RESEARCH ARTICLE

Lymphoid Chemokines CCL19 and CCL21 are Expressed in the Central Nervous System During Experimental Autoimmune Encephalomyelitis: Implications for the Maintenance of Chronic Neuroinflammation Sandra Columba-Cabezas, Barbara Serafini, Elena Ambrosini, Francesca Aloisi Laboratory of Organ and System Pathophysiology, Istituto Superiore di Sanità, Roma, Italy S. C.-C. and B. S. equally contributed to this work.

The simultaneous presence of dendritic, T- and Bcells in the central nervous system (CNS) of mice with experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis, suggests that interactions among these cell types might be instrumental in the local induction and maintenance of autoimmune reactions. In this study, we explored the possibility that such aberrant leukocyte recruitment in the CNS could be sustained by “lymphoid” chemokines which orchestrate dendritic cell and lymphocyte homing to lymphoid organs. Transcripts for CCL19 and CCL21 and their common receptor CCR7 were induced in the CNS of mice undergoing relapsing-remitting and chronic-relapsing EAE. While CCL21 immunoreactivity was confined to the endothelium of some inflamed blood vessels, CCL19 was expressed by many infiltrating leukocytes and some astrocytes and microglia in the CNS parenchyma. CCR7+ cells accumulated in inflammatory lesions during EAE progression, when abundant infiltration of the CNS by mature dendritic cells, B-cells and cells expressing naive Tcell markers also occurred. These findings suggest that CCL19 and CCL21 produced in the EAE-affected CNS may be critical for the homing of antigen presenting cells and lymphocytes, resulting in continuous local antigenic stimulation and maintenance of chronic neuroinflammation. Brain Pathol 2003;13:38-51.

Introduction

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) characterized by focal myelin destruction, varying degree of axonal

pathology and progressive neurological dysfunction (35, 44). Perivascular infiltrates of T-cells, B-cells and monocytes/macrophages, are a prominent histological feature of the discrete lesions that develop in the brain and spinal cord in typical cases of MS (35). Studies in patients and in the animal model, experimental autoimmune encephalomyelitis (EAE), indicate that a Th1mediated immune response to various myelin antigens and myelin-specific autoantibodies might be implicated in the pathogenesis of MS (52, 60). However, the initial events triggering autoimmune responses and the mechanisms sustaining CNS inflammation in MS remain to be elucidated. In recent years, dendritic cells (DCs) have received increasing attention as key players in the pathogenesis of autoimmunity (18, 37). DCs have the unique ability to activate resting T-cells and play an important role in the initiation and regulation of immune responses (5, 28). DCs capturing self-antigen in the target organ and migrating to draining lymph nodes or spleen may initiate stimulation of autoreactive T-cells and sustain new waves of priming of naive T-cells. In several organ-specific autoimmune diseases and related animal models, DCs have been shown to infiltrate the target organ and to interact with T and B lymphocytes to form lymphoidlike structures (25, 33, 36, 55). This process, which is also referred to as lymphoid neogenesis, is thought to be crucial for continuous immune activation in the target tissue itself (33). Recent studies have documented that cells expressing DC-specific markers accumulate in the CNS tissue and cerebrospinal fluid during EAE (39, 51, 53) and MS (47), respectively, suggesting that DC trafficking to the CNS (a site where DCs are normally confined to the meninges and choroid plexuses) could contribute to tolerance breakdown and sustain the inflammatory self-recognition process. The intracerebral expression of maturation signals for DCs and chemokines/chemokine receptors regulating trafficking

Corresponding author: Dr Francesca Aloisi, Laboratory of Organ and System Pathophysiology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Roma, Italy (e-mail: [email protected])

and positioning of DCs and lymphocytes may thus represent critical elements in the progression of CNS autoimmunity. A recently identified set of chemokines, referred to as “lymphoid,” have emerged as key signals for the migration of lymphocyte subsets into T- and B-cell zones of secondary lymphoid organs and for the development of lymphoid tissue compartments (15, 41, 50, 64). Among these, CCL19, also known as macrophage inflammatory protein-3 or EBV-induced molecule 1 ligand, and CCL21, also known as 6Ckine, secondary lymphoid tissue chemokine, Exodus-2 and thymus-derived chemotactic agent-4, are expressed in T-cell zones by stromal cells and interdigitating DCs and by HEV endothelia and stromal cells, respectively (24, 38, 43). CCL19 and CCL21 are structurally related chemokines and share a common receptor, CCR7 (62, 63), that is expressed on mature DCs, B-cells, naive and central memory T-cells (7, 30, 50, 59, 61). CCR7-deficient mice and paucity of lymph node T-cell (plt) mice lacking CCL19 and CCL21 expression show defective migration of lymphocytes and DCs into T-cell zones, decreased B-cell homing, and profound morphological alterations of secondary lymphoid organs (20, 23, 38, 58). Recent studies in chronic infectious and autoimmune diseases have demonstrated expression of lymphoid chemokines, including CCL19 and CCL21, in the target organ, indicating that these molecules may orchestrate leukocyte microenvironmental homing resulting in the formation of ectopic lymphoid tissue (25, 26, 40, 55). In this study, we explored the hypothesis that CNS autoimmunity could be sustained by aberrant lymphoid chemokine expression favoring cell trafficking and interactions between DCs and lymphocytes in the neural tissue. Using RT-PCR and immunohistochemical techniques, we show that expression of CCL19, CCL21 and their cognate receptor CCR7 is induced in the CNS of mice developing EAE, particularly during disease progression. These findings provide novel insights into the signals regulating leukocyte homing to the inflamed CNS and suggest novel approaches to treat CNS autoimmune diseases by targeting the lymphoid chemokine system. Materials and Methods

Animals. Female adult SJL mice and Biozzi AB/H mice, 6 to 8 weeks of age, were obtained from Charles River (Calco, Italy) and Harlan U.K. Ltd (Bicester, United Kingdom), respectively. Mice were housed in a controlled environment in accordance with the guidelines

of the European Community Council of the Welfare of Experimental Animals (86/609/EEC). All experimental procedures were approved by the Italian Ministry of Health. EAE induction. For the induction of relapsingremitting EAE, 42 female SJL mice were injected subcutaneously in the flank with 0.2 mg proteolipid protein (PLP) 139-151 peptide (Primm, Milano, Italy), in complete Freund’s adjuvant (CFA; Difco Laboratories, Detroit, Mich) on day 0 and 7, and with pertussis toxin (0.2 g/mouse; Sigma Chemical Co., St. Louis, Mo) intraperitoneally on day 0, 1, 7 and 8, as previously described (17). For the induction of chronic-relapsing EAE, 26 female Biozzi AB/H mice were injected subcutaneously in the flank with 1 mg of lyophilized, autologous whole spinal cord homogenate reconstituted in PBS and mixed with CFA (incomplete Freund’s adjuvant [IFA] supplemented with 320 g/ml of Mycobacterium Tuberculosis H37 RA and 40 g/ml of Mycobacterium Butyricum [Difco Laboratories]) on day 0 and 7, and with pertussis toxin intraperitoneally on day 0, 1, 7 and 8, following published procedures (21). Control mice (6 for each strain) were injected with PBS in CFA and pertussis toxin, according to the same schedule. Mice were weighed and examined daily for clinical signs of EAE, which was scored on the following scale: grade 0, no abnormality; grade 1, reduced tail tonus or slightly clumsy gait; grade 2, tail atony, moderately clumsy gait, impaired righting ability, or any combination of these signs; grade 3, additional hind limb weakness; grade 4, hind limb paralysis and fore limb weakness; grade 5, tetraplegia or moribund state. Mice were sacrificed during the preclinical stage (day 10-13 post-immunization [p.i.]) and at different times after appearance of clinical signs. SJL mice were sacrificed during the first peak of disease (day 15-16 p.i.; EAE grade 2-4), during the early remission phase (day 20-22 p.i., 24 hours after disappearance of clinical signs), and 24 to 48 hours after the onset of relapses that followed complete remissions (up to 2 months p.i.; EAE grade 1-4). Biozzi mice were sacrificed immediately after disease onset (day 15 p.i.; EAE grade 3-4), and after partial remissions (EAE grade 1) and relapses occurring during the chronic-relapsing phase (up to 3 months p.i.; EAE grade 3-4). For each EAE stage examined, at least 6 mice were sacrificed for molecular and immunohistochemical studies. Reverse transcription-polymerase chain reaction (RT-PCR). Following anesthesia, mice were perfused intracardiacally with cold PBS. Different CNS areas

Lymphoid Chemokines CCL19 and CCL21 in the CNS During EAE—Columba-Cabezas et al

39

Figure 1. Disease patterns of EAE-affected mice. A. Time course of the clinical score of three SJL mice developing relapsing-remitting EAE after immunization with PLP 139-151 peptide in CFA; mice were sacrificed at day 26 (), 29 () and 63 () p.i. B. Time course of the clinical score of three Biozzi AB/H mice developing chronic-relapsing EAE after immunization with spinal cord homogenate in CFA; mice were sacrificed at day 42 (), 50 () and 86 () p.i.

(cerebrum, cerebellum/brain stem and spinal cord) and positive control tissues (spleen and thymus) were collected, immediately frozen in liquid nitrogen and kept at -80°C until RNA extraction. Total RNAs were extracted from normal mouse control tissues and from the CNS of control and EAE-affected SJL and Biozzi AB/H mice using the SV Total RNA Isolation System (Promega, Madison, Wis), according to the manufacturer's instructions. One g of purified RNA was reverse-transcribed using the Reverse Transcription System (Promega), with 15 U of Avian Myeloblastosis Virus Reverse Transcriptase. For detection of murine CCL19 transcripts, one tenth of this reaction was added to 40 l of PCR mix and amplified for 29 cycles (45 s/94°C, 45 s/60°C, 60 s/72°C) with 2.5 units of Taq polymerase (Promega), using the following primers: 5GCC TCA GAT TAT CTG CCA T-3 (sense) and 5-AGA CAC AGG GCT CCT TCT GGT-3 (antisense) (size of the amplified product of CCL19 cDNA was 332 bp). Due to the presence of one functional CCL19 gene and three CCL19 pseudogenes in the mouse genome, all of which are transcribed, the PCR primer pairs were chosen to

40

detect expression of the functional CCL19 gene bearing the ATG start codon (38). For detection of murine CCL21 transcripts, cDNA was subjected to PCR amplification for 28 to 30 cycles (30 s/94°C, 60 s/60°C, 60 s/72°C), using the following primers: 5-GAA AAT TCC CTA CAG TAT TGT CCG AG-3 (sense) and 5GAC TTA GAG GTT CCC CGG TTC-3 (antisense) (size of the amplified product of CCL21 cDNA was 222 bp). CCL21-specific primers were designed based on the sequence deposited in the EMBL/GenBank (accession number AF171085) (59). For detection of CCR7 transcripts, cDNA was subjected to PCR amplification for 28 cycles (60 s/94°C, 120 s/60°C, 90 s/72°C) using the following primers: 5-CCA GGAAAAACG TGC TGG TG3 (sense) and 5–GGC CAG GTT GAG CAG GTA GG–3 (antisense) (size of the amplified product of CCR7 cDNA was 287 bp) (10). As control, transcripts of the “housekeeping” gene glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were amplified for 25 cycles (30 s/94°C, 30 s/60°C, 45 s/72°C), using the following primers: 5-CCA TGG AGA AGG CCG GGG-3 (sense) and 5-CAA AGT TGT CAT GGA TGA CC-3 (antisense) (size of the amplified product of GAPDH cDNA was 194 bp). Negative controls lacking template RNA or RT were included in each experiment. The PCR products were fractionated on a 3% NuSieve® 3:1 agarose gel (FMC BioProducts, Rockland, Mass) and visualized by ethidium bromide staining. The identity of the PCR amplified fragments was verified by size comparison with DNA standards (Promega) and by direct DNA sequencing of purified fragments (M-Medical, Roma, Italy). At least 3 mice were examined for each time point tested. In some of the EAE-affected mice used for RT-PCR studies, histopathological analysis was also performed on unfixed, frozen CNS sections to confirm the presence of immune infiltrates. The RT-PCR data shown were obtained in individual, representative mice. Immunohistochemistry. For specimen collection, mice were anesthesized with xylazine chloride hydrate and ketamine and perfused intracardiacally with PBS followed by cold 4% paraformaldehyde in 0.1 M phosphate buffer (pH 7.4). Brains, spinal cords and inguinal lymph nodes were removed, kept overnight in 4% paraformaldehyde at 4°C, passed in 15% and 30% sucrose, frozen in dry ice-chilled isopentane, cut in serial sections with cryostat and stored at -20°C. For immunohistochemistry, 10 m-thick sections were air dried, passed in 70%, 95% and 100% ethanol and then dried again. After rehydratation with PBS and 20minute incubation with 0.3% H2O2 in PBS (to eliminate

Lymphoid Chemokines CCL19 and CCL21 in the CNS During EAE—Columba-Cabezas et al

Figure 2. CCL19, CCL21 and CCR7 transcripts are upregulated in the CNS of SJL mice with PLP 139-151-induced EAE (A) and of Biozzi AB/H mice with whole spinal cord-induced EAE (B). RNA was extracted from different CNS areas and from control tissues (thymus and spleen), reverse transcribed and subjected to PCR amplification using CCL19, CCL21 and CCR7 specific primers, as described in Materials and Methods. RT-PCR using GAPDH specific primers is also shown, as an internal control. RNA was extracted from control and EAE-affected mice at the indicated disease stages. The PCR products were visualized by ethidium bromide staining. Results from individual mice are shown. Two relapsing SJL mice are shown, which were sacrificed at day 29 (grade 1) and 63 p.i. (grade 4), respectively. The two chronic-relapsing Biozzi AB/H mice were sacrificed at days 35 and 28 p.i. (grade 4), respectively. Lanes: 1 = cerebrum; 2 = cerebellum/brain stem; 3 = spinal cord; 4 = thymus; 5 = spleen.

endogenous peroxidase activity), sections were preincubated with 10% normal rabbit or goat serum and then incubated at 4°C overnight with the following purified primary antibodies: goat polyclonal anti-mouse CCL19 (R&D Systems, Minneapolis, Minn); rabbit polyclonal anti-mouse CCL21 (PeproTech, London, United Kingdom); goat polyclonal anti-mouse CCR7 (M18; Santa Cruz Biotechnology, Santa Cruz, Calif); rat monoclonal antibody MIDC-8, binding to a still unidentified antigen expressed by interdigitating DCs in T-cell areas of murine lymphoid organs (Serotec, Oxford, United

Kingdom) (8); rat monoclonal antibody anti-mouse CD11b/Mac-1 (5C6, Serotec); and rat monoclonal antibodies anti-mouse CD4 (RM4-5), CD45R/B220 (RA36B2), CD45RB (16A) and CD62L (MEL-14) (Pharmingen, San Diego, Calif). All antibodies were optimally diluted in PBS/1%BSA (2-5 g/ml). After extensive washings with PBS, sections were incubated with the corresponding biotinylated secondary antibodies (rabbit anti-goat IgG, goat anti-rabbit IgG and rabbit anti-rat IgG, all from Vector Laboratories, Burlingame, Calif) and avidin:biotinylated peroxidase complex

Lymphoid Chemokines CCL19 and CCL21 in the CNS During EAE—Columba-Cabezas et al

41

42

Lymphoid Chemokines CCL19 and CCL21 in the CNS During EAE—Columba-Cabezas et al

Figure 3. (Opposing page) Immunohistochemical detection of CCL19 in the CNS of normal and EAE-affected mice. Cryostat sections were stained for CCL19 (A, B, F-K), CD11b (C), CD4 (D) or MIDC-8 (E) and visualized with DAB, as described in Materials and methods. A-F: stainings in SJL mice. CCL19+ cells in the cerebellar meninges (A) of normal SJL mice. Serial spinal cord sections from SJL mice at EAE onset were stained for CCL19 (B), CD11b (C), CD4 (D) and MIDC-8 (E). CCL19 immunoreactivity is detected in the cytoplasm of numerous cells within a large submeningeal and perivascular infiltrate comprising mainly CD11b+ macrophages and fewer CD4+ T-cells and MIDC-8+ DCs. CCL19+ cells in perivascular infiltrates and adjacent spinal cord parenchyma during a relapse (F). G, H: CCL19 stainings in the spinal cord of Biozzi AB/H mice at EAE onset (G) and during chronic-relapsing disease (H). I-K: higher magnifications of CCL19+ cells showing typical astrocyte (arrow) and microglia (arrow head) morphologies in the spinal cord of relapsing SJL mice. Original magnifications: 500 (A-D, F-H), 1000 (E), and 1575 (I-K).

(ABC), using the ABC Vectastain Elite kit (Vector Laboratories), according to the manufacturer’s instructions. Staining reactions were performed with 3,3 diaminobenzidine (DAB; Sigma Chemical Co., St. Louis, Mo) as substrate. MIDC-8 stainings were performed using the amplification procedure described by Adams, with minor modifications (1). Following incubation with ABC, sections were incubated with 0.01% H2O2 and 28 mM biotinylated tyramine, prepared as described, for 5 minutes at room temperature. After extensive washings, sections were incubated with ABC for 20 minutes, PBS for 30 minutes, and DAB for another 2 to 5 minutes. For negative controls, the primary antibody was omitted from the diluent or replaced with preimmune serum. Sections were counterstained with hematoxylin and viewed with a Zeiss Axiophot photomicroscope. Results

Detection of CCL19, CCL21 and CCR7 mRNAs in the CNS of EAE-affected mice. To explore the expression of CCL19, CCL21, and their cognate receptor CCR7 in different phases of CNS autoimmunity, EAE was induced in 2 mouse strains, SJL and Biozzi AB/H mice, which develop a relapsing-remitting and chronicrelapsing form of paralytic disease, respectively. After immunization with the PLP peptide 139-151 in CFA, 60 to 80% of the SJL mice developed the first EAE clinical signs around days 14 to 18, followed by complete remission, and about 20% of these underwent one or more relapses (Figure 1A). In all Biozzi AB/H mice immunized with whole spinal cord homogenate in CFA, EAE clinical signs first appeared at day 13-15 after immunization and the disease progressed as a continuum of relapses and incomplete or, less frequently, complete remissions (Figure 1B). Mice immunized with PBS/CFA showed no signs of disease (not shown). Expression of transcripts specific for CCL19, CCL21 and CCR7 was investigated in control and EAE-affected mice by RT-PCR analysis. The CNS areas examined included cerebrum, cerebellum/brain

stem and spinal cord, whereas thymus and spleen were used as positive control tissues for CCR7 and for CCL19 and CCL21, respectively. Very low levels of CCL19 mRNA, but no CCL21 and CCR7 mRNAs, were detected in the CNS, mainly cerebrum and cerebellum/brain stem, of control SJL (Figure 2A) and Biozzi AB/H (Figure 2B) mice (untreated and injected with PBS/CFA). In the CNS of preclinical SJL mice (day 10 to 13 post-immunization), levels of CCL19 and CCL21 mRNAs were either unchanged or only slightly increased, whereas CCR7 mRNA was weakly expressed. During the first peak of disease, expression of CCL19 and CCR7 mRNAs was increased in all CNS areas from SJL mice, whereas levels of CCL21 transcripts remained low. All transcripts analyzed returned to baseline levels during the early remission phase (24 h after disappearance of clinical signs). Following EAE relapses, CCL19 and CCR7 mRNAs were detected in the CNS of all SJL mice examined, with CCR7 being expressed predominantly in the spinal cord and at higher levels compared to disease onset. CCL21 mRNA was weakly induced only in some relapsing SJL mice. In the CNS of Biozzi AB/H mice, CCL19 mRNA was slightly induced before (day 10 p.i.) and immediately after the appearance of clinical signs but its levels markedly increased during the chronic-relapsing phase, the highest expression being detected in the cerebellum/brain stem and spinal cord (Figure 2B). Similarly, CCR7 mRNA levels were low at disease onset and increased as disease progressed, particularly in the spinal cord. CCL19 and CCR7 mRNAs were detected in the CNS also during partial remissions (Figure 2B). While CCL21 mRNA was not detected in the CNS of Biozzi AB/H mice before and immediately after EAE onset, low levels of CCL21 mRNA were induced in the cerebellum/brain stem and spinal cord of Biozzi AB/H mice undergoing chronic-relapsing disease (Figure 2B). Immunohistochemical detection of CCL19 and CCL21 in the CNS of EAE-affected mice. The expression of CCL19 and CCL21 was then evaluated by

Lymphoid Chemokines CCL19 and CCL21 in the CNS During EAE—Columba-Cabezas et al

43

Figure 4. Immunohistochemical detection of CCL21 in peripheral lymph node and CNS of normal and EAE-affected mice. A-C: stainings in SJL mice. Lymph node section showing positive reaction for CCL21 on HEV (A). Absence of CCL21 immunoreactivity in the normal spinal cord (B). CCL21 immunoreactivity on an intrameningeal blood vessel in the spinal cord of a SJL mouse developing chronic EAE (C); the meninges and intrameningeal leukocytes are negative. D-F: stainings in the spinal cord of Biozzi AB/H mice during chronic-relapsing EAE. CCL21 is expressed in cells lining the lumen of inflamed blood vessels. In a severely affected Biozzi AB/H mouse (day 49 p.i., EAE grade 4) (F), CCL21 immunoreactivity is also detected in some process-bearing cells scattered in the adjacent white matter. Original magnifications: 500 (B, D) and 1000 (A, C, E, F).

44

Lymphoid Chemokines CCL19 and CCL21 in the CNS During EAE—Columba-Cabezas et al

immunohistochemistry on cryostat sections of normal and EAE-affected CNS using polyclonal anti-mouse CCL19 and CCL21 antibodies. Sections of inguinal lymph nodes were also stained to check the specificity of the two anti-chemokine antibodies. Consistent with the distribution of CCL19 and CCL21 in lymphoid tissue (15, 24, 38, 43), anti-CCL19 antibody labeled dendritiform cells, likely DCs and stromal cells, in T-cell zones (not shown), whereas anti-CCL21 antibodies labeled HEV endothelia and stromal cells in T-cell zones of normal mouse lymph nodes (Figure 4A). In control SJL and Biozzi AB/H mice, anti-CCL19 antibody labeled few, distinct cells in the cerebral, cerebellar and spinal cord meninges (Figure 3A). No staining of normal CNS tissue was observed with rabbit anti-CCL21 antibody (Figure 4B). After the appearance of the first clinical signs (EAE grade 3-4), CCL19 immunoreactivity was found associated with numerous immune cells accumulating in the meninges, and in the perivascular and submeningeal lesions of the brain stem and spinal cord of both mouse strains (Figure 3B, G). Rare CCL19+ cells were also present in the parenchyma adjacent to the inflammatory infiltrates. Compared to Biozzi AB/H mice, SJL mice exhibited more prominent submeningeal cell infiltration, but fewer perivascular lesions. At these sites, the cell infiltrates comprise a major population of CD11b+ macrophages, and less numerous CD4+ T-cells and MIDC-8+ DCs (Fig. 3 C-E; only stainings performed in SJL mice are shown). Conversely, very few CCL19+ cells were detected in the perivascular inflammatory cuffs of the cerebellum, which contain mainly T-cells and very few macrophages and DCs (not shown). These observations, together with previous findings that CCL19 is produced by macrophages and DCs, but not Tcells (15), suggest that macrophages, and to a lesser extent DCs, are the major source of CCL19 in EAE lesions. Numerous CCL19+ cells were also detected in EAE lesions of relapsing SJL mice (EAE grade 2) and chronically affected Biozzi AB/H mice (EAE grade 3-4) (Figure 3F, H). Staining was particularly abundant in the CNS of Biozzi AB/H mice which exhibited the most invasive and widespread lesions (Figure 3H). As disease progressed, CCL19 immunoreactivity was also detected in some astrocyte-like and microglia-like cells in the CNS parenchyma of both mouse strains (Figure 3I-K). At EAE onset, no CCL21+ cells were detected in CNS lesions of either SJL or Biozzi AB/H mice (not shown). Despite weak induction of CCL21 mRNA (see Figure 2A), CCL21 immunoreactivity remained unde-

tectable also in the CNS of relapsing SJL mice. This suggests that in a EAE model characterized by acute disease attacks followed by complete remissions, the self-limiting inflammatory process is not sufficient to trigger CCL21 protein synthesis, at least at levels detectable by immunohistochemical techniques. Interestingly, only in one SJL mouse developing an atypical form of chronic stable EAE (grade 3 up to day 30 p.i.), CCL21 immunoreactivity localized to the endothelial cells of an intrameningeal blood vessel in the spinal cord (Figure 4C). In Biozzi AB/H mice undergoing disease progression, anti-CCL21 antibody reacted with endothelial cells of intrameningeal vessels (not shown) and of some inflamed vessels in the spinal cord and brain stem white matter (Figure 4D, E). Although the intensity of CCL21 staining was variable, we calculated that about 40% of the inflamed vessels examined expressed CCL21. Only in one severely diseased Biozzi AB/H mouse, some CCL21+ cells displaying an elongated or irregular cell body and thin cytoplasmic processes were found in a discrete area of the spinal cord white matter containing scattered inflammatory infiltrates (Figure 4F). Immunohistochemical detection of CCR7-expressing cells and potential cellular targets for lymphoid chemokines in the EAE-affected CNS. Immunohistochemical analysis of CCR7 expression was performed using a polyclonal anti-mouse CCR7 antibody. While strong CCR7 immunoreactivity was detected on lymphocytes in T- and B-cell zones of control lymph nodes, no CCR7 staining was detected in normal CNS (not shown). A few CCR7+ cells were present in CNS lesions of SJL and Biozzi AB/H mice at EAE onset (Figure 5A, B). In agreement with the RT-PCR data, more numerous CCR7+ cells were found in the CNS of relapsing SJL and chronically-affected Biozzi AB/H mice, mainly within perivascular and intrameningeal cell infiltrates (Figure 5C, D). Because CCR7 has been reported to be expressed on mature DCs, B-cells, naive and central memory T-cells (7, 50, 59, 61), we next examined the expression of markers specific for these cell types in spinal cord lesions. As previously shown (51), cells labeled by MIDC-8 antibody, a marker for mature interdigitating DCs, enter the CNS at disease onset (see also Figure 3E) and markedly increase in number during EAE progression in both mouse strains (Figure 5E; only staining in Biozzi AB/H mice is shown). CD45R/B220+ B-cells were detected only in the spinal cord of relapsing SJL and chronic-relapsing Biozzi AB/H mice, mainly in intrameningeal and perivascular locations (Figure 5F). We also stained for L-selectin

Lymphoid Chemokines CCL19 and CCL21 in the CNS During EAE—Columba-Cabezas et al

45

46

Lymphoid Chemokines CCL19 and CCL21 in the CNS During EAE—Columba-Cabezas et al

Figure 5. (Opposing page) Presence of CCR7+ cells and potential cellular targets for lymphoid chemokine action in EAE lesions. At disease onset, CCR7 immunoreactivity is detected on the surface of some round cells in the spinal cord lesions of SJL (A) and Biozzi AB/H (B) mice. In relapsing SJL (C) and chronic-relapsing Biozzi AB/H (D) mice, numerous CCR7+ cells are present in the meningeal and perivascular infiltrates. E-H: Sections from the spinal cord of a chronic-relapsing Biozzi AB/H mouse were stained for MIDC-8 (E), a marker for mature DC, B220/CD45R (F), a marker for B-cells, CD62L (G) and CD45RB (H), two molecules highly expressed in naive T-cells. Sections were cut from a 200-m thick tissue block; F and G show adjacent sections. Original magnifications: 500 (E) and 1000 (A-D, F-H).

(CD62L) and CD45RB, since these molecules are known to be highly expressed on murine naive and central memory T-cells that preferentially home to lymphoid tissues (54, 59). Numerous CD62L+ and CD45RB+ cells were detected in the meninges and some inflamed blood vessels of relapsing SJL and chronic-relapsing Biozzi AB/H mice (Figure 5G, H). Although anti-CD62L and anti-CD45RB antibodies may also stain B-cells, we would exclude labeling of other cell types (eg, neutrophils, monocytes/macrophages) based on the observation that cells expressing CD62L or CD45RB were absent or only occasionally seen in CNS lesions at EAE onset (not shown). CNS tissues from control mice failed to stain with all the above antibodies. Taken together, these findings indicate that, during EAE progression, the inflamed CNS provides a suitable environment for recruiment and positioning of immune cell types that are potential targets for CCL19 and CCL21. Discussion

Major unsolved issues regarding the immunopathology of MS concern the nature of the immunologic drive responsible for continuous T-cell accumulation/activation in the CNS and the site for priming and expansion of CNS-targeted T-cells. Because DCs are thought to play an essential role in the pathogenesis of autoimmunity (5, 18, 37), signals regulating DC-T-cell homing and interactions in the diseased CNS may be the key to understanding how the inflammatory self-recognition process is sustained. The presence of DCs in the CSF of MS patients (47) and CNS lesions of EAE-affected rodents (39, 51, 53) suggests that these potent antigen presenting cells may internalize CNS antigen(s) and either transport it to secondary lymphoid organs or present it to T-cells locally to maintain autoimmune responses. The first scenario is supported by studies showing that progression to chronicity in EAE and MS is associated with the appearance of circulating T-cells with new CNS-antigenic specificities, a phenomenon referred to as epitope spreading (34, 57), and by the presence of macrophages/DCs containing myelin components in the cervical lymph nodes of EAE-affected monkeys (16). The second scenario is favored by the demonstra-

tion of T-cell clonal expansions in the CSF and active lesions of MS patients, most of which are not identifiable in blood (3). Furthermore, Targoni et al (56) have recently shown that in SJL mice with PLP 139-151 peptide-induced EAE a second wave of effector T-cells recognizing PLP 178-191 peptide is detected first in the CNS and only at later time points in the spleen, suggesting that priming of new autoreactive T-cells could occur in the target organ itself. The characteristic intrathecal synthesis of oligoclonal immunoglobulins (2) and the accumulation of clonally related B-cells in the CSF and brain lesions of MS patients (6, 13) are also suggestive of antigen-driven, intrathecal B-cell clonal expansion which, according to the requirements for B-cell stimulation, should be sustained by local B-, T-cell and DC interactions (32, 41). This study supports the concept that compartmental leukocyte homing to the CNS may contribute to autoimmunity by showing that EAE development in SJL and Biozzi AB/H mice is associated with the intracerebral expression of 2 chemokines, CCL19 and CCL21, that regulate trafficking of lymphocytes and DCs in lymphoid organs (15, 41, 50). While CCL19 mRNA and immunoreactivity are detected in the CNS already at EAE onset, CCL21 expression is much lower and confined to some CNS blood vessels during chronic disease. The demonstration that immune cells expressing CCR7, the common receptor for CCL19 and CCL21, accumulate in the CNS during EAE progression further implies a role for these chemokines in maintaining neuroinflammation. CCL19 and CCL21 have been shown to stimulate 2 integrin-mediated adhesion of lymphocytes to intercellular adhesion molecules (ICAM)-1 and cause rapid arrest of circulating lymphocytes to the vascular endothelium (9, 14). CCL21 also induces 47 integrinmediated adhesion of lymphocytes to mucosal addressin cell adhesion molecule-1 (MadCAM-1) expressed in mucosal HEV (46).The unique combination of traffic molecules found on HEV, including ICAM-1, MadCAM-1, peripheral node addressin (PNAd) or CD62L ligand, and CCL21, is thought to be essential for efficient lymphocyte homing into secondary lymphoid organs (22, 50). In EAE, blood-brain barrier endothelia

Lymphoid Chemokines CCL19 and CCL21 in the CNS During EAE—Columba-Cabezas et al

47

acquire some properties of HEV, like expression of ICAM-1 and MadCAM-1 (11, 45). Although the lack of PNAd in inflamed CNS blood vessels (11) implies that these do not sustain lymphocyte emigration as efficiently as HEV, it is conceivable that the combined expression of CCL21, ICAM-1 and MadCAM-1 is sufficient to enable the intracerebral migration of circulating lymphocytes that normally home to lymphoid tissues (15, 41, 50). This idea is consistent with the accumulation of cells expressing B-cell (CD45R/B220), naive and central memory T-cell (CD45RB, CD62L) markers in CNS lesions of mice undergoing EAE progression. Interestingly, in a severely diseased Biozzi AB/H mouse, CCL21 was found expressed by some intraparenchymal process-bearing cells whose identity remains to be established. A possible explanation for this finding is that CNS inflammation facilitates the recruitment and/or induces the local differentiation of cells with properties of stromal cells, another major source of CCL21 in lymphoid T-cell zones (15, 38). Lymphotoxin- and tumor necrosis factor, which have a pivotal role in regulating the synthesis of CCL19 and CCL21 in lymphoid tissues (26, 42), are both produced in the EAE-affected CNS (27, 29) and could induce production of lymphoid chemokines during inflammation. In secondary lymphoid organs, CCL19 is produced by T zone stromal cells and interdigitating DCs and is thought to facilitate interaction of naive T-cells with DCs (15, 38). Interestingly, perivascular CCL19 can be transcytosed to the luminal surfaces of HEV and may also participate in CCR7-mediated triggering of leukocyte arrest (4). The observation that during EAE CCL19 is expressed by CNS-infiltrating immune cells, likely macrophages and DCs, as well as by resident microglia and astrocytes, suggests that this chemokine may help to retain CCR7-expressing cells in the lesion site and favor encounter of mature DCs with pathogenic T-cells. Notably, DCs present in EAE lesions are stained by mAb MIDC-8, which recognizes interdigitating DCs in T-cell zones of lymphoid organs, where T-cell responses are initiated (8, 51). Thus, a number of similarities in the cellular and chemokine environment of lymphoid Tcell zones and chronic EAE lesions suggest that DCs maturing in the inflamed CNS, where myelin destruction leads to continuous exposure of self-antigens, could support the local generation of autoreactive T-cells. The findings that in the normal CNS CCL19 mRNA is expressed at low levels and that some cells in the meninges stain positively for CCL19 raise the question of whether CCL19 regulates the constitutive trafficking of specific leukocyte subsets to the CNS. Because

48

CCL19 has been reported to attract hematopoietic progenitor cells, particularly macrophage progenitors (31), we propose that it could contribute to the normal turnover of macrophages residing in the meningeal compartment. Recent studies have shown that ectopic expression of CCL21 in the pancreas is sufficient to trigger lymphoid neogenesis (19), whereas transgenic mice expressing CCL21 in oligodendrocytes develop CNS inflammation and neurologic abnormalities, but show neither lymphocyte recruitment nor lymphoid neogenesis in the CNS (12). Although these findings highlight the importance of tissue-specific environments in regulating chemokine effects, they do not exclude that lymphoid chemokines might play a role in CNS immunopathologies. So far, only limited morphological evidence has been provided that lymphoid-like structures develop in the CNS during autoimmune disease (48, 49). The simultaneous presence of CCR7+ cells, mature DCs, Bcells and cells expressing naive/central memory T-cell markers in the inflamed meninges and CNS perivascular spaces during EAE progression suggests that these may represent preferential sites for the establishment of functional interactions mimicking those occurring in lymphoid organs. In conclusion, this study describes the expression of lymphoid chemokines in EAE lesions and suggests their possible involvement in promoting cellular interactions that could be important for priming and expansion of autoreactive lymphocytes within the inflamed CNS. It remains to be seen if blockade of the intracerebral production or activity of lymphoid chemokines might represent an effective therapeutic approach for inhibiting EAE progression, and ultimately for the treatment of MS patients. Acknowledgments

We thank Luciano Adorini for critical reading of the manuscript. This work was supported by Project “Dendritic cells in central nervous system autoimmunity” of the Istituto Superiore di Sanità, and Projects “Multiple Sclerosis” and “Inflammatory, oxidative and autoimmune mechanisms in disabling CNS diseases” of the Italian Ministry of Health. Note Added in Proof

After the submission of this manuscript, Alt et al reported that CCL19 and CCL21 are expressed in venules surrounded by inflammatory cells in the CNS of EAE-affected SJL mice (Eur J Immunol 32:21332144.)

Lymphoid Chemokines CCL19 and CCL21 in the CNS During EAE—Columba-Cabezas et al

References 1.

Adams JC (1992) Biotin amplification of biotin and horseradish peroxidase signals in histochemical stains. J Histochem Cytochem 40:1457-1463.

2.

Archelos JJ, Hartung H-P (2000) Pathogenic role of autoantibodies in neurological diseases. Trends Neurosci 23:317-327.

3.

Babbe H, Roers A, Waisman A, Lassmann H, Goebels N, Hohlfeld R, Friese M, Schröder R, Deckert M, Schmidt S, Ravid R, Rajewsky K (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 192:393404.

4.

Baekkevold ES, Yamanaka T, Palframan RT, Carlsen HS, Reinholt FP, von Andrian UH, Brandtzaeg P, Haraldsen G (2001) The CCR7 ligand ELC (CCL19) is transcytosed in high endothelial venules and mediates T-cell recruitment. J Exp Med 193:1105-1111.

5.

Banchereau J, Steinman RM (1998) Dendritic cells and the control of immunity. Nature 392:245-252.

6.

Baranzini SE, Jeong MC, Butunoi C, Murray RS, Bernard CC, Oksenberg JR (1999) B-cell repertoire diversity and clonal expansion in multiple sclerosis brain lesions. J Immunol 163:5133-5144.

7.

Bowman EP, Campbell JJ, Soler D, Dong Z, Manlongat N, Picarella D, Hardy RR, Butcher EC (2000) Developmental switches in chemokine response profiles during B-cell differentiation and maturation. J Exp Med 191:13031317.

8.

Breel M, Mebius RE, Kraal G (1987) Dendritic cells of the mouse recognized by two monoclonal antibodies. Eur J Immunol 17:1555-1559.

9.

Campbell JJ, Hedrick J, Zlotnik A, Siani MA, Thompson DA, Butcher EC (1998) Chemokines and the arrest of lymphocytes under flow conditions. Science 279:381-384.

10. Campbell JJ, Pan J, Butcher EC (1999) Developmental switches in chemokine responses during T-cell maturation. J Immunol 163:2353-2357. 11. Cannella B, Cross AH, Raine CS (1991) Relapsing autoimmune demyelination: a role for vascular addressins. J Neuroimmunol 35:295-300. 12. Chen S-C, Leach MW, Chen Y, Cai X-Y, Sullivan L, Wiekowski M, Dovey-Hartman BJ, Zlotnik A, Lira SA (2002) Central nervous system inflammation and neurological disease in transgenic mice expressing the CC chemokine CCL21 in oligodendrocytes. J Immunol 168:1009-1017. 13. Colombo M, Dono M, Gazzola P, Roncella S, Valetto A, Chiorazzi M, Mancardi GL, Ferrarini M (2000) Accumulation of clonally related B lymphocytes in the cerebrospinal fluid of multiple sclerosis patients. J Immunol 164:2782-2798. 14. Constantin G, Majeed M, Giagulli C, Piccio L, Kim JY, Butcher EC, Laudanna C (2000) Chemokines trigger immediate 2 integrin affinity and mobility changes: differential regulation and roles in lymphocyte arrest under flow. Immunity 13:759-769.

15. Cyster JG (1999) Chemokines and cell migration in secondary lymphoid organs. Science 286:2098-2102. 16. De Vos AF, van Meurs M, Brok HPM, Boon L, Rensing S, ‘t Hart BA, Laman JD (2001) Transfer of CNS autoantigens and presentation in secondary lymphoid organs. J Neuroimmunol 118:75. 17. Di Rosa F, Serafini B, Scognamiglio P, Di Virgilio A, Finocchi L, Aloisi F, Barnaba V (2000) Short-lived immunization site inflammation in self-limited active experimental allergic encephalomyelitis. Int Immunol 12:711719. 18. Drakesmith H, Chain B, Beverley P (2000) How can dendritic cells cause autoimmune disease? Immunol Today 21:214-217. 19. Fan L, Reilly CR, Luo Y, Dorf ME, Lo D (2000) Ectopic expression of the chemokine TCA4/SLC is sufficient to trigger lymphoid neogenesis. J Immunol 164:3955-3959. 20. Förster R, Schubel A, Breitfeld D, Kremmer E, RennerMüller I, Wolf E, Lipp M (1999) CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell 99:23-33. 21. Furlan R, Poliani PL, Galbiati F, Bergami A, Grimaldi LM, Comi G, Adorini L, Martino G (1998) Central nervous system delivery of interleukin 4 by a nonreplicative herpes simplex type 1 viral vector ameliorates autoimmune demyelination. Hum Gene Ther 9:2605-2617. 22. Girard J-P, Springer TA (1995) High endothelial venules (HEVs): specialized endothelium for lymphocyte migration. Immunol Today 16:449-457. 23. Gunn MD, Kyuwa S, Tam C, Kakiuchi T, Matsuzawa A, Williams LT, Nakano H (1999) Mice lacking expression of secondary lymphoid organ chemokine have defects in lymphocyte homing and dendritic cell localization. J Exp Med 189:451-460. 24. Gunn MD, Tangemann K, Tam C, Cyster GC, Rosen SD, Williams LT (1998) A chemokine expressed in lymphoid high endothelial venules promotes the adhesion and chemotaxis of naive T lymphocytes. Proc Natl Acad Sci USA 95:258-263. 25. Hjelmström P (2001) Lymphoid neogenesis: de novo formation of lymphoid tissue in chronic inflammation through expression of homing chemokines. J Leukoc Biol 69:331-339. 26. Hjelmström P, Fjell J, Nakagawa T, Sacca R, Cuff CA, Ruddle NH (2000) Lymphoid tissue homing chemokines are expressed in chronic inflammation. Am J Pathol 156:1133-1138. 27. Issazadeh S, Ljungdahl A, Hojeberg B, Mustafa M, Olsson T (1995) Cytokine production in the central nervous system of Lewis rats with experimental autoimmune encephalomyelitis: dynamics of mRNA expression for interleukin-10, interleukin-12, cytolysin, tumor necrosis factor alpha and tumor necrosis factor beta. J Neuroimmunol 61:205-212. 28. Jonuleit H, Schmitt E, Steinbrink K, Enk AH (2001) Dendritic cells as a tool to induce anergic and regulatory T-cells. Immunol Today 22:394-400.

Lymphoid Chemokines CCL19 and CCL21 in the CNS During EAE—Columba-Cabezas et al

49

29. Juedes AE, Hjelmström P, Bergman CM, Neild AL, Ruddle NH (2000) Kinetics and cellular origin of cytokines in the central nervous system: insight into mechanisms of myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis. J Immunol 164:419426. 30. Kellermann S-A, Hudak S, Oldham ER, Liu Y-J, McEvoy LM (1999) The CC chemokine receptor-7 ligands 6Ckine and macrophage inflammatory protein-3 are potent chemoattractants for in vitro- and in vivo-derived dendritic cells. J Immunol 162:3859-3864. 31. Kim CH, Pelus LM, White JR, Broxmeyer HE (1998) Macrophage-inflammatory protein-3/EBI1-ligand chemokine/CKb-11, a CC chemokine, is a chemoattractant with a specificity for macrophage progenitors among myeloid progenitor cells. J Immunol 161:2580-2585. 32. Kim CH, Rott LS, Clark-Lewis I, Campbell DJ, Wu L, Butcher EC (2001) Subspecialization of CXCR5+ T-cells: B helper activity is focalized in a germinal center-localized subset of CXCR5+ T-cells. J Exp Med 193:1373-138.

43. Ngo VN, Tang LH, Cyster JG (1998) Epstein-Barr virusinduced molecule 1 ligand chemokine is expressed by dendritic cells in lymphoid tissues and strongly attracts naive T-cells and activated B-cells. J Exp Med 188:181191 44. Noseworthy JH (1999) Progress in determining the causes and treatment of multiple sclerosis. Nature 399:A40-A47 45. O’Neill JK, Butter C, Baker D, Gschmeissner SE, Kraal G, Butcher EC (1991) Expression of vascular addressins and ICAM-1 by endothelial cells in the spinal cord during chronic relapsing experimental allergic encephalomyelitis in the Biozzi AB/H mouse. Immunology 72:520-525 46. Pachynski RK, Wu SW, Gunn MD, Erle DJ (1998) Secondary lymphoid-tissue chemokine (SLC) stimulates integrin 47-mediated adhesion of lymphocytes to mucosal addressin cell adhesion molecule-1 (MadCAM-1) under flow. J Immunol 161:952-956 47. Pashenkov M, Huang Y-M, Kostulas V, Haglund M, Södeström M, Link H (2001) Two subsets of dendritic cells are present in human cerebrospinal fluid. Brain 124:480-492

33. Kratz A, Campos-Neto A, Hanson MS, Ruddle NH (1996) Chronic inflammation caused by lymphotoxin is lymphoid neogenesis. J Exp Med 183:1461-1472.

48. Prineas JW (1979) Multiple sclerosis: presence of lymphatic capillaries and lymphoid tissue in the brain and spinal cord. Science 203:1123-1125

34. Lehmann PV, Forsthuber T, Miller A, Sercarz EE (1992) Spreading of T-cell autoimmunity to cryptic determinants of an autoantigen. Nature 358:155-157.

49. Raine CS, Mokhtarian F, McFarlin DE (1984) Adoptively transferred chronic relapsing experimental autoimmune encephalomyelitis in the mouse. Neuropathologic analysis. Lab Invest 51:534-546

35. Lucchinetti CF, Bruck W, Lassmann H (1998) Multiple sclerosis: lessons learned from neuropathology. Semin Neurol 18:337-349. 36. Ludewig B, Odermatt B, Landmann S, Hengartner H, Zinkernagel RM (1998) Dendritic cells induce autoimmune diabetes and maintain disease via de novo formation of local lymphoid tissue. J Exp Med 188:1493-1501. 37. Ludewig B, Odermatt B, Ochsenbein AF, Zinkernagel RM, Hengartner H (1999) Role of dendritic cells in the induction and maintenance of autoimmune diseases. Immunol Rev 169:45-54. 38. Luther SA, Tang LH, Hyman PL, Garr AG, Cyster JG (2000) Coexpression of the chemokines ELC and SLC by T zone stromal cells and deletion of the ELC gene in the plt/plt mouse. Proc Natl Acad Sci USA 97:12694-12699. 39. Matyszak MK, Perry VH (1996) The potential role of dendritic cells in immune-mediated inflammatory diseases in the central nervous system. Neuroscience 74:599-608. 40. Mazzucchelli L, Blaser A, Kappeler A, Scharli P, Laissue JA, Baggiolini M, Uguccioni M (1999) BCA-1 is highly expressed in Helicobacter pylori-induced mucosa-associated lymphoid tissue and gastric lymphoma. J Clin Invest 104:R49-54. 41. Moser B, Loetscher P (2001) Lymphocyte traffic control by chemokines. Nature Immunol 2:123-128. 42. Ngo VN, Korner H, Gunn MD, Schmidt KN, Riminton DS, Cooper MD, Browning JL, Sedgwick JD, Cyster JG (1999) Lymphotoxin alpha/beta and tumor necrosis factor are required for stromal cell expression of homing chemokines in B and T-cell areas of the spleen. J Exp Med 189:403-412

50

50. Sallusto F, McKay CR, Lanzavecchia A (2000) The role of chemokine receptors in primary, effector, and memory immune responses. Annu Rev Immunol 18:593-620 51. Serafini B, Columba-Cabezas S, Di Rosa F, Aloisi F (2000) Intracerebral recruitment and maturation of dendritic cells in the onset and progression of experimental autoimmune encephalomyelitis. Am J Pathol 157:19912002 52. Steinman L (1999) Assessment of animal models for MS and demyelinating disease in the design of rational therapy. Neuron 24:511-514 53. Suter T, Malipiero U, Otten L, Ludewig B, Muelethaler-Mottet A, Mach B, Reith W, Fontana A (2000) Dendritic cells and differential usage of the MHC class II transactivator promoters in the central nervous system in experimental autoimmune encephalitis. Eur J Immunol 30:794-802 54. Swain SL, Croft, M, Dubey C, Haynes L, Rogers P, Zhang X, Bradley LM (1996) From naive to memory T-cells. Immunol Rev 150:143-167 55. Takemura S, Braun A, Crowson C, Kurtin PJ, Cofield RH, O’Fallon WM, Goronzy JJ, Weyand CM (2001) Lymphoid neogenesis in rheumatoid synovitis. J Immunol 167:1072-1080 56. Targoni OS, Baus J, Hofstetter HH, Hesse MD, Karulin AY, Boehm BO, Forsthuber TG, Lehmann PV (2001) Frequencies of neuroantigen-specific T-cells in the central nervous system versus the immune periphery during the course of experimental allergic encephalomyelitis. J Immunol 166:4757-4764

Lymphoid Chemokines CCL19 and CCL21 in the CNS During EAE—Columba-Cabezas et al

57. Tuohy VK, Yu M, Weinstock-Guttman B, Kinkel RP (1997) Diversity and plasticity of self recognition during the development of multiple sclerosis. J Clin Invest 99:16821690 58. Vassileva G, Soto H Zlotnik A, Nakano H, Kakiuchi T, Hedrick JA, Lira SA (1999) The reduced expression of 6Ckine in the plt mouse results from the deletion of one of two 6Ckine genes. J Exp Med 190:1183-1188 59. Weninger W, Crowley MA, Manjunath N, von Andrian UH (2001) Migratory properties of naive, effector and memory CD8+ T-cells. J Exp Med 194:953-966 60. Wingerchuk DM, Lucchinetti CF, Noseworthy JH (2001) Multiple sclerosis: current pathophysiological concepts. Lab Invest 81:263-281 61. Yanagihara S, Komura E, Nagafune J, Watarai H, Yamaguchi Y (1998) EBI1/CCR7 is a new member of dendritic cell chemokine receptor that is up-regulated upon maturation. J Immunol 161:3096-3102 62. Yoshida R, Imai T, Hieshima K, Kusuda J, Baba M, Kitaura M, Nishimura M, Kakizaki M, Nomiyama H, Yoshie O (1997) Molecular cloning of a novel human CC chemokine EBI1-ligand chemokine that is a specific functional ligand for EBI1, CCR7. J Biol Chem 272:1380313809 63. Yoshida R, Nagira M, Kitaura M, Imagawa N, Imai T, Yoshie O (1998) Secondary lymphoid-tissue chemokine is a functional ligand for the CC chemokine receptor CCR7. J Biol Chem 273:7118-7122 64. Zlotnik A, Yoshie O (2000) Chemokines: a new classification system and their role in immunity. Immunity 12:121-127

Lymphoid Chemokines CCL19 and CCL21 in the CNS During EAE—Columba-Cabezas et al

51

The cell-based L-glutathione protection assays to study endocytosis and recycling of plasma membrane proteins.

Membrane trafficking involves transport of proteins from the plasma membrane to the cell interior (i.e. endocytosis) followed by trafficking to lysoso...
818KB Sizes 0 Downloads 0 Views