Skip to main content
Erschienen in: Fluids and Barriers of the CNS 1/2005

Open Access 01.12.2005 | Research

The circumventricular organs participate in the immunopathogenesis of experimental autoimmune encephalomyelitis

verfasst von: Martina Schulz, Britta Engelhardt

Erschienen in: Fluids and Barriers of the CNS | Ausgabe 1/2005

Abstract

Background

During inflammatory conditions of the central nervous system (CNS), such as in multiple sclerosis or in its animal model, experimental autoimmune encephalomyelitis (EAE), immune cells migrate from the blood stream into the CNS parenchyma and into the cerebrospinal fluid (CSF) spaces. The endothelial blood-brain barrier (BBB) has been considered the most obvious entry site for circulating immune cells. Recently, the choroid plexus has been considered as an alternative entry site for circulating lymphocytes into the CSF. The choroid plexus, belongs to the circumventricular organs (CVOs) localized in the walls of the ventricles. Other CVOs, which similar to the choroid plexus lack an endothelial BBB, have not been considered as possible entry sites for immune cells into the CNS parenchyma or the CSF. Here we asked, whether CVOs are involved in the recruitment of inflammatory cells into the brain during EAE.

Methods

We performed an extensive immunohistological study on the area postrema (AP), the subfornical organ (SFO), the organum vasculosum of the lamina terminalis (OVLT) and the median eminence (ME) in frozen brain sections from healthy SJL mice and mice suffering from EAE. Expression of cell adhesion molecules, the presence of leukocyte subpopulations and the detection of major histocompatibility complex antigen expression was compared.

Results

Similar changes were observed for all four CVOs included in this study. During EAE significantly increased numbers of CD45+ leukocytes were detected within the four CVOs investigated, the majority of which stained positive for the macrophage markers F4/80 and Mac-1. The adhesion molecules ICAM-1 and VCAM-1 were upregulated on the fenestrated capillaries within the CVOs. A considerable upregulation of MHC class I throughout the CVOs and positive immunostaining for MHC class II on perivascular cells additionally documented the immune activation of the CVOs during EAE. A significant enrichment of inflammatory infiltrates was observed in close vicinity to the CVOs.

Conclusion

Our data indicate that the CVOs are a site for the entry of immune cells into the CNS and CSF and consequently are involved in the inflammatory process in the CNS during EAE.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1743-8454-2-8) contains supplementary material, which is available to authorized users.

Competing interests

The author(s) declare that they have no competing interests.

Authors' contributions

The entire experimental work described in this paper was performed by MS. Evaluation of the data as well as the documentation was performed by both authors. BE designed and supervised the study, and wrote the manuscript. Both authors have read and approved the final manuscript.
Abkürzungen
AP
area postrema
BBB
blood brain barrier
CD
cluster of differentiation
CNS
central nervous system
CVOs
circumventricular organs
EAE
experimental autoimmune encephalomyelitis
ICAM-1
intercellular adhesion molecule-1
LFA-1
leukocyte function associated antigen-1
MAdCAM-1
mucosal addressin cell adhesion molecule-1
ME
median eminence
MHC
major histocompatibility complex
OVLT
organum vasculosum of the lamina terminalis
PECAM-1
platelet endothelial cell adhesion molecule -1
SFO
subfornical organ
VCAM-1
vascular cell adhesion molecule -1

Background

In multiple sclerosis and in its animal model, experimental autoimmune encephalomyelitis (EAE), inflammatory cells obtain access to the central nervous system (CNS) parenchyma and the cerebrospinal fluid (CSF) and initiate the events leading to signs of paralysis. The endothelial blood-brain barrier (BBB) has been considered the obvious place for entry for circulating lymphocytes into the CNS. Therefore most investigations have focused on defining the molecular mechanisms involved in leukocyte recruitment from the circulating blood across the endothelial BBB. The adhesion molecules, intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1), both members of the immunoglobulin superfamily, are upregulated on the endothelial cells of cerebral vessels during EAE and actively involved in the recruitment of inflammatory cells across the BBB (summarized in [1]).
Trafficking pathways for the entry of immune cells into the CSF remain unknown to date. The CSF of healthy individuals contains between 150,000 cells and 500,000 cells. During multiple sclerosis this number increases dramatically. Neither in the healthy individual nor during multiple sclerosis does the cellular composition of the CSF reflect that of the peripheral blood, suggesting a stringent control for leukocyte entry into the CSF at all times [2]. Recently it was considered that leukocytes enter the CSF using a direct pathway through the choroid plexus. The microvessels within the choroid plexus are different to those in brain parenchyma, the most significant of which is that the endothelial cells allow free movement of molecules via fenestrations and intercellular gaps (reviewed in [3]). Instead, the barrier is located at the level of the choroid plexus epithelial cells, which form tight junctions inhibiting paracellular diffusion of water soluble molecules [4].
Migration of leukocytes through the choroid plexus into the CSF has been suggested by the finding that fluorescently labeled splenocytes are present in the choroid plexus stroma two hours after intravenous injection in mice [5]. The adhesion molecules ICAM-1 and VCAM-1, which are required for leukocyte entry into the CNS, are expressed on the choroid plexus epithelium [6], become upregulated during EAE, and can mediate lymphocyte binding in vitro [7]. These observations suggest that the choroid plexus is involved in the communication of the immune system with the CNS probably by allowing the entry of immune cells directly into the CSF spaces.
Besides the choroid plexus there are additional structures in the CNS of mammals lacking an endothelial BBB. These areas fulfill neurohemal and neurosecretory functions, in that the neurons monitor hormonal stimuli and other substances within the blood or secrete neuroendocrines into the blood, and are commonly referred to as the circumventricular organs (CVOs; reviewed in [8, 9]). CVOs are localized at strategic points close to the midline of the brain within the ependymal walls lining the 3rd and the 4th ventricle. Because they lack an endothelial BBB they lie within the blood milieu and thus form a blood-CSF barrier. At the cellular level the barrier between the CVOs and the neuropil is established by specialized epithelial cells called tanycytes. The median eminence (ME) belongs to the purely neuroendocrine CVOs also including the pineal gland and the neurohypophysis. The ME is localized between the stem of the infundibular or pituitary stalk of the hypophysis and the hypothalamus at the base of the 3rd ventricle. The ME contains the terminations of axons from hypothalamic neurons and specialized glial cells. Neuroendocrine functions of the ME include the release of gonadotropin-releasing hormone by which the ME influences reproductive functions [10].
The sensory CVOs such as the subfornical organ (SFO), the organum vasculosum of the lamina terminalis (OVLT) and the area prostrema (AP) are characterized by a high density of small neurons, astrocytes and microglial cells [11]. The SFO and the OVLT are structures in the anterior wall of the 3rd cerebral ventricle. The SFO bulges from the roof of the 3rd ventricle into its lumen at the level of the interventricular foramina, whereas the OVLT is localized in the lamina terminalis between the chiasma opticum and the anterior commissure. The AP can be found at the caudal end of the fossa rhomboidea in the floor of the 4th ventricle. The sensory CVOs monitor the changes in osmotic, ionic and hormonal composition of the blood and are therefore involved in regulating thirst and fluid metabolism [12].
The strategic localization of the CVOs in the wall of the ventricles prompted us to ask whether the CVOs might, similar to the choroid plexus, be involved in the recruitment of immune cells into the brain during EAE. We have performed an extensive immunohistological study localizing adhesion molecules, MHC class I and II antigens and leukocyte subpopulations within the subfornical organ (SFO), the median eminence (ME), the organum vasculosum of the lamina terminalis (OVLT) and the area postrema (AP) of the SJL/N mouse. During EAE similar changes were observed within all four CVOs included in this study. Specifically, the presence of significantly increased numbers of CD45+ leukocytes in the CVOs suggested the recruitment of inflammatory cells into the the parenchyma of the CVOs probably mediated via endothelial ICAM-1 and VCAM-1 which were both upregulated on the microvessels within the CVOs. These changes were accompanied by a significant upregulation of MHC class I throughout the CVOs and induction of MHC class II on perivascular cells. Our observations indicate an active communication between the CVOs and the CNS parenchyma and thus the involvement of the CVOs in the inflammatory process of the CNS during EAE.

Methods

Induction of EAE

Female SJL/N mice were obtained from Bomholdgård Breeding, Ry, Denmark and used for experiments at the age of 10 weeks. Active EAE (aEAE) was induced by immunization with syngeneic spinal cord homogenate as described in detail before [13, 14]. Briefly, SJL/N mice were immunized with 100 μg of spinal cord homogenate (SCH) from syngeneic mice in complete Freund's Adjuvant (CFA) (Gibco Laboratories, Grand Island, NY), containing 60 μg/ml Mycobacterium tuberculosis H37Ra and 10 μg/ml Mycobacterium butyricum (Difco Laboratories, Detroit, MI) subcutaneously. Heat-killed Bordetella pertussis organisms (3 × 109; kindly provided by Dr. Kolbe, Behring Werke, Marburg, Germany or Dr. Kohler, Berna Biotech, Bern, Switzerland) were injected in 0.5 ml of PBS on days 1 and 3 post immunization. Animals were checked daily and clinical severity was documented as follows: 0.5 = limp tail; 1 = weak hindlimbs, unsteady gate; 2 = paraplegic, 3 = paraplegic plus incontinent and weakness. Clinical disease occurred approximately 14 days post immunization with spinal cord homogenate. A total of 46 mice with a clinical severity of EAE of 1 to 2 during their first clinical episode are presented in this study. As control, 10 untreated littermates, which were kept in the same cages – were sacrificed and investigated in the same way. All animal experiments were performed in accordance with the German and the Swiss legislation on the protection of animals and approved by the respective government authorities (permission numbers B2/127 and 55/04).

Monoclonal Antibodies

Mec13.3 (anti-mouse PECAM-1/CD31), MK2.7 (anti-mouse VCAM-1), 3C4 (anti-mouse ICAM-2), RB40.34/4 (anti-mouse P-selectin), Lyt 2 (anti-mouse CD8), 145-2C11 (anti-mouse CD3), F4/80 (anti-mouse macrophages), FD441.8 (anti-mouse LFA-1), Mel-14 (anti mouse L-selectin) were purchased form BD Pharmingen, Germany, where detailed information on the antibodies can be obtained http://​www.​bdbiosciences.​com/​pharmingen. C363 (anti mouse CD3ε) was purchased from Southern Biotechnology Associates, Birmingham, AL, USA, http://​www.​southernbiotech.​com. ER-TR2 (rat- anti mouse MHC class II) and ER-MP42 (rat anti-mouse MHC class I) were purchased from Dianova, Hamburg, Germany, http://​www.​dianova.​com/​index_​flash.​php. The hybridomas PS/2 (anti-mouse α4-integrin), M1/9 (anti-mouse CD45), M1/70 (anti-mouse Mac-1), B220 (anti-mouse CD45R), GK1.5 (anti-mouse CD4), MECA-367 and MECA-89 (anti- mouse MAdCAM-1) and Hermes-1 (= 9B5, anti-human CD44, used as an isotype-matched control) were obtained from ATCC (Rockville, MA, USA; http://​www.​atcc.​org). 25ZC7 (anti-mouse ICAM-1) and 9DB3 (anti-mouse VCAM-1) were kindly provided by D. Vestweber (Münster, Germany; [13]. UZ 4 and UZ 7 (both anti-mouse E-selectin) and MECA 32 were kindly provided by R. Hallmann (Münster, Germany; [14, 15]).

Immunohistochemistry

Animals were anesthetized using isoflurene anesthesia (Abbott, Wiesbaden, Germany), and were perfused with either phosphate buffered saline (PBS) or 1% formaldehyde (PFA) in PBS through the left ventricle of the heart. Tissue was removed, embedded in Tissue-tec, (OCT, Miles Inc., Vogel, Giessen, Germany), and snap-frozen in a 2-methylbutane (Merck, Darmstadt, Germany) bath at -80°C. Cryostat sections (6 μm) were air dried overnight, acetone fixed, and stained using a three-step immunoperoxidase technique. Sections were incubated sequentially with primary mAbs, biotinylated secondary goat-anti-rat IgG, (Vector, Boehringer Ingelheim Bioproducts, Heidelberg, Germany), and horseradish peroxidase-conjugated Streptavidin (Vector) for 30 min each step in a humidified chamber, with PBS washes in between the single steps. Sections were developed with 0.07% amino-ethylcarbazol (AEC, Sigma, Germany) and 0.009% hydrogen peroxide in 0.01 M acetate buffer (pH 5.2) for 10 min. Sections were counterstained with Hematoxylin (Gill's formula, Merck), coverslipped with Aquatex (Merck, Darmstadt, Germany) and immediately analyzed.
To detect presence of luminal antigens in cerebral blood vessels mice were injected intravenously with 250 μg primary antibody, anesthetized 15 or 30 minutes later, perfused first with PBS to remove unbound antibody then with 1% PFA to fix bound antibody. Tissue was processed as described above and immunohistology was performed accordingly leaving out the first antibody. This study summarizes results from a total of 49 individual immunostainings where CVOs from 46 EAE mice and 10 healthy mice were compared.

Results

In the mouse the panendothelial antigen MECA-32 becomes specifically downregulated in brain endothelia during the maturation of the blood-brain barrier [15]. As a consequence, the MECA-32 antigen was absent on the mature cerebral endothelium, whereas it remained present on vessels outside of the CNS and the microvessels within the choroid plexus and the circumventricular organs (CVOs, Figures 1, 2, 3, 4, 5, 6, 7, 8).
In the present study we investigated four CVOs, namely the subfornical organ (SFO), the area postrema (AP), the median eminence (ME) and the organum vasculosum of the lamina terminalis (OVLT), which all lack an endothelial blood-brain barrier (BBB) and demonstrate fenestrated MECA-32+ microvessels.

The circumventricular organs in healthy SJL/N mice

In general, characteristics of the 4 CVOs analyzed were similar. As expected in the healthy SJL/N mice we detected MECA-32+ microvessels within the SFO (Fig. 1), in the AP (Fig. 2) and the ME and OVLT (Table 1). Additionally, CVO microvessels stained positive for PECAM-1 and some of them stained for ICAM-2 (Table 1). Adhesion molecules such as ICAM-1 and VCAM-1 (Figs. 1, 2 and Table 1) or MAdCAM-1, E- and P-selectin (Table 1) could not be detected on the microvessels within the CVOs of healthy mice. A low number of CD45-positive perivascular cells was regularly observed in all CVOs (Figs. 1, 2, 3 and Table 1). In contrast, in only 1 or 2 out of 10 immunostainings performed for each CVO, were individual perivascular cells staining positive for macrophage markers such as F4/80 or Mac-1 or MHC class I or II and LFA-1 detected in CVOs of healthy mice (Table 1). This suggests a variable and rarely detectable presence of individual perivascular macrophages within the CVOs during health. In contrast, parenchymal staining for these molecules was never detected in the CVOs of healthy mice. Similarly, staining for CD3, CD4, CD8 or B220 was always negative in the CVOs of healthy mice (Table 1).
Table 1
Summary of the immunohistochemical characterization of CVOs in health and EAE
Molecule
Healthy1
EAE2
Microvessels
MECA-32
+ on all vessels
+ on all vessels
PECAM-1
+ on all vessels
+ on all vessels
ICAM-2
+ on some vessels
+ on some vessels
ICAM-1
No staining
+ on most vessels luminally and abluminally/perivascular
VCAM-1
No staining
+ on few vessels luminally and abluminally or perivascular no luminal VCAM-1 detected in ME
MAdCAM-1
No staining
no staining
E-selectin
no staining
no staining
P-selectin
no staining
no staining
Immune cells
CD45
+ on few perivascular cells
+ on many cells within the CVOs
CD3
no staining
+ on few round cells ca. 4 cells/section in SFO ca. 10 cells/section in AP
CD4
no staining
+ on few round cells
CD8
no staining
+ on few round cells
B220
no staining
+ on few round cells
F4/80
only rarely observed on single perivascular cells*
+ on many exclusively perivascular cells
Mac-1 (αM-integrin)
only rarely observed on single perivascular cells*
+ on round and branched cells located perivascularly and throughout the CVOs
LFA-1 (αL-integrin)
only rarely observed on single perivascular cells*
+ on round and branched cells located perivascularly and throughout the CVOs
α4-integrin
no staining
very faint staining on few round cells – not regularly detected
MHC class I
only rarely observed on few perivascular cells§
considerable staining throughout the CVOs; negative on endothelium
MHC class II
only rarely observed on single perivascular cells*
+ on many perivascular cells
1All four CVOs were analysed in a total of 10 healthy mice. Results therefore summarize 10 immunostainings. 2A total of 46 mice has been analyzed. Staining for each marker in each CVO was observed at least 10 times. If not stated differently, stainings were repeatedly observed in all CVOs analysed. *Single immunopositive cells were observed in only 1 or 2 of 10 immunostainings. §Few immunopositive cells were observed in 2 out of 10 immunostainings.

The circumventricular organs during EAE in SJL/N mice

During EAE the changes observed within the four CVOs analysed were overall very similar. The microvessels within the CVOs retained their characteristic staining for the MECA-32 antigen, PECAM-1 and ICAM-2 (Figs. 3, 4, 5, 6, 7, 8, Table 1). In contrast to healthy mice, a high number of CD45+ cells could be detected within the CVOs (Fig. 3, 4, 5, 6), with the most prominent increase of CD45+ cells detected within the OVLT (Fig. 3). CD45+ cells within all four CVOs were localized in close proximity to microvessels and were also found within the CVO parenchyma. Besides the intense CD45 staining on cells of round appearance, a fainter immunostaining for CD45 was detected on cells with a more ramified appearance, indicating that, in addition to the recruitment of CD45+ cells into these areas, CD45 might also be upregulated on the resident microglial cells within the CVOs (Fig. 4, 5, 6).
In order to further characterize the CD45+ cells detected within the CVOs during EAE we performed immunostaining for T cell, B cell, and macrophage markers. A large number of F4/80+ and Mac-1+ macrophages or microglial cells were detected within the CVOs (Figs. 4, 5). Whereas F4/80+ cells were exclusively detected adjacent to CVO microvessels, Mac-1 positive cells could also be found within the parenchyma of the different CVOs (Fig. 4 and 5). Mac-1 positive cells presented either as round cells or cells with many processes, suggesting that Mac-1 was detected on macrophages recruited into the CVOs and also upregulated on resident microglial cells. Besides macrophages, few B220+ B lymphocytes as well as few CD3+ T lymphocytes could regularly be detected within the CVOs during EAE, especially within the SFO (Fig. 4). Most of the CD3 positive T cells recruited into the SFO were CD4+ T helper cells as indicated by the detection of mostly CD4+ but rarely CD8+ cells within this CVO during EAE (Fig. 4 and Table 1).
The adhesion molecules from the integrin family, namely leukocyte function-associated antigen (LFA)-1 (αLβ2-integrin) and α4-integrins can be involved in inflammatory cell recruitment across the BBB during EAE. Therefore we investigated, whether CD45+ cells localized within the CVOs also express these adhesion molecules. Whereas a high number of LFA-1+ cells was detected in the CVOs (Fig. 4), α4-integrin positive cells were detected only rarely (Table 1).
Next we asked whether the potential endothelial ligands for Mac-1, LFA-1 and α4-integrins, namely intercellular adhesion molecule (ICAM)-1 and vascular cell adhesion molecule (VCAM)-1, are expressed on the microvascular endothelial cells within the CVOs. We found prominent induction of ICAM-1 and to a lesser degree of VCAM-1 (Figs. 4, 5, 6), but not of mucosal addressin cell adhesion molecule (MAdCAM)-1, E-selectin or P-selectin (Table 1) on CVO microvessels during EAE. In order to evaluate, whether ICAM-1 and VCAM-1 are available on the luminal surface of the CVOs capillary endothelial cells for the recruitment of circulating Mac-1+, LFA-1+ and α4-integrin+ leukocytes into the CVOs, we injected anti-ICAM-1 or anti-VCAM-1 antibodies into live animals suffering from EAE and investigated their binding to CVO microvessels by immunohistology. Whereas ICAM-1 was detected on the luminal surface of the capillaries of all CVOs investigated (Figs. 7, 8 and data not shown), VCAM-1 although present on the luminal surface of microvessels in the SFO, OVLT and AP (Fig. 7 and data not shown) could not be detected on the luminal surface of the capillaries within the ME (Fig. 8). Taken together, these observations suggest that recruitment of inflammatory cells into the CVOs mainly depend on LFA-1/ICAM-1 or Mac-1/ICAM-1 rather than VCAM-1/α4-integrin-mediated interactions.
In addition to the increased number of immunocompetent cells present in the CVOs during EAE, we observed a significant upregulation of major histocompatibility complex (MHC) antigens class I on cells throughout the entire parenchyma of the CVOs, where expression was absent before in healthy mice (Figs. 4, 6 and Table 1). In contrast, expression of MHC class II was solely induced on perivascular cells, a significant number of which could now regularly be found within all four CVOs during EAE (Fig. 4, 6 and Table 1).
Finally, besides the leukocyte infiltration of the CVOs proper, we observed the preferential localization of massive inflammatory cuffs within the brain parenchyma or the ventricular walls localized in close vicinity or directly adjacent to CVOs, suggesting an involvement of the CVOs in immune cell recruitment into these distinct areas (Fig. 9).

Discussion

In the present study we provide evidence that immune cell invasion into the CNS during EAE is accompanied by a prominent immune activation of the CVOs. Changes within the CVOs are characterized by the presence of an increased number of CD45+ cells, the considerable induction of MHC class I and MHC class II molecules within the CVOs as well as microglial activation documented by increased expression of Mac-1. Upregulated expression of adhesion molecules on the CVO microvessels suggest their involvement in the recruitment of macrophages, T- and B cells into the CVOs during EAE. Taken together, these observations indicate an active participation of the CVOs in the immunopathogenesis of EAE.
The involvement of CVOs in the communication of the immune system with the nervous system has been considered before. Due to their fenestrated capillaires the CVOs are often referred to as "windows of the brain" and have been thought to serve as entry points for pro-inflammatory cytokines into the CNS [16]. In fact, receptors for inflammatory cytokines and bacterial fragments are constitutively expressed in cells within the sensory CVOs and the release of pro-inflammatory cytokines such as interleukin-1 or TNF-α outside the CNS has been demonstrated to deliver signals into the CNS via the CVOs leading to neuroendocrine responses such as elevations in adrenocorticotropic hormone (ACTH) in the plasma, development of fever and the activation of the hypothalamic-pituitary-adrenal (HPA) axis [17]. Additionally, CVOs play an active role in the development of brain dysfunction during sepsis. Induction of experimental endotoxin shock by injection of lipopolysaccaride induces the sequential expression of molecules involved in innate immune responses such as CD14 or Toll-like receptors first in the CVOs and subsequently in the brain parencyhma (reviewed by [18]). Furthermore, involvement of CVOs in the establishment of an acute phase response during peripheral immune stimuli was demonstrated [19]. Finally, a role for CVOs in sending signals into the brain during systemic inflammation is supported by observations that lesions of CVOs block several components of brain-controlled illness responses (i.e. fever or neuroendocrine modifications) [20].
An active participation of CVOs in the auto-immunopathogenesis in EAE has not been reported to date. In the present study, we found that CVOs in the healthy SJL/N mouse presented a low number of perivascular CD45+ cells, probably macrophages, similar to other regions of the CNS. We only occasionally detected expression of MHC class I or MHC class II on individual perivascular cells suggesting that MHC-positive cells rarely reside within CVOs of healthy mice. Adhesion molecules associated with inflammation could not be detected. In contrast, during clinical EAE at the time when immune cells have penetrated the BBB and perivascular infiltration of T cells and macrophages is observed within the CNS parenchyma, the CVOs demonstrated a dramatic increase in the presence of CD45+ cells. As CD45 is upregulated on activated microglial cells, which reside in high numbers in CVOs [21], the increased number of CD45+ cells detected within the CVOs during EAE is most probably due to both the recruitment of circulating CD45+ immune cells from the periphery into these areas and additionally to the upregulation of CD45 on microglial cells residing within the CVOs.
Most of the CD45+ cells also expressed the macrophage marker F4/80 or Mac-1, which can be expressed on macrophages or activated microglial cells. Based on the immunological and morphological criteria, these cells most probably can be classified as perivascular macrophages or ramified microglial cells, as classical parenchymal microglial cells, as well as scattered rounded macrophage-like cells. Activation of microglial cells by upregulated expression of CD45 and induction of Mac-1 therefore parallels the events observed within the CNS parenchyma protected by the BBB during EAE.
Microglial activation within the CVOs during EAE is further supported by our observation that MHC class I antigens were induced throughout the BBB-deficient parenchyma in all CVOs. In addition, MHC class II molecules could now regularly be detected on perivascular microglial cells or macrophages within the CVOs, which may function as a cellular barrier against blood-borne pathogens. Thus, increased expression of MHC class I and II molecules on microglial cells and perivascular macrophages, which are known characteristic immunopathological changes observed in the CNS parenchyma during EAE, are similarly observed in the BBB-deficient areas of the CVOs [22].
Besides the immune activation of the CVOs, recruitment of a low number of CD3+ T cells and B220+ B cells was detected in the parencyhma of CVOs during EAE, demonstrating that immune cells are invading these areas during EAE. It has been shown that endothelial VCAM-1 and ICAM-1 are involved in lymphocyte recruitment across brain endothelium in vitro and in vivo [13, 2325]. The upregulated expression of ICAM-1 and VCAM-1 on the microvessels of the CVOs during EAE suggests that the same molecules mediating immune cell entry across the BBB might be involved in guiding inflammatory cells into the CVOs. This notion was confirmed by our findings that both molecules are available on the luminal surface of CVO microvessels, with the exception of the median eminence, where luminal VCAM-1 could not be detected. Inflammatory cells within the CVOs were found to stain positive for Mac-1+ and LFA-1+, therefore resemling those, present in the inflammatory cuffs surrounding brain venules forming a barrier [26]. In contrast, whereas during EAE inflammatory cells within the brain express high levels of α4-integrins [26] only few α4-integrin+ leukocytes were found within the CVOs. These observations indicate that inflammatory cell recruitment into the CVOs is not necessarily mediated by the same mechanisms as those regulating leukocyte recruitment across the inflamed BBB, which during EAE was shown to be mainly dependent on α4-integrin and VCAM-1[1, 25].
Whether immune cells enter the brain or CSF via the CVOs cannot be determined by this study. It is, however, tempting to speculate that CVOs have an active role in determining inflammatory cell recruitment into the brain as we observed a significantly increased number of inflammatory cuffs and infiltrating cells in close vicinity to the CVOs as compared to regions of the brain with a greater distance to the CVOs. These observations suggest that during EAE the inflamed CVOs deliver cellular or molecular signals into the CNS, which may influence immune cell entry into the CNS and CSF.

Conclusion

Taken together our observations demonstrate that the CVOs are involved in the immunopathogenesis of EAE. Considering their strategic localization within the walls of the ventricles, they may be involved in immune cell entry from the blood into the CNS parenchmyma and also from the blood into the CSF. Therefore, the development of neuroprotective strategies will require consideration of the molecular changes in the CVOs during CNS inflammation.

Acknowledgements

We owe great thanks to Hiltrud Hölzinger for her expert assistance in photodocumentation. Most of this research was funded by the Max-Planck Society, Germany and the Deutsche Forschungsgemeinschaft (DFG), Germany. Martina Schulz was funded by the DFG.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The author(s) declare that they have no competing interests.

Authors' contributions

The entire experimental work described in this paper was performed by MS. Evaluation of the data as well as the documentation was performed by both authors. BE designed and supervised the study, and wrote the manuscript. Both authors have read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Engelhardt B, Ransohoff R: The ins and outs of T cell trafficking to the central nervous system: Anatomic sites and molecular mechanisms. Trends Immunol. 2005, Engelhardt B, Ransohoff R: The ins and outs of T cell trafficking to the central nervous system: Anatomic sites and molecular mechanisms. Trends Immunol. 2005,
2.
Zurück zum Zitat Kivisakk P, Mahad DJ, Callahan MK, Trebst C, Tucky B, Wei T, Wu L, Baekkevold ES, Lassmann H, Staugaitis SM, Campbell JJ, Ransohoff RM: Human cerebrospinal fluid central memory CD4+ T cells: evidence for trafficking through choroid plexus and meninges via P-selectin. Proc Natl Acad Sci U S A. 2005, 100: 8389-94. Epub 2003 Jun 26.. 10.1073/pnas.1433000100.CrossRef Kivisakk P, Mahad DJ, Callahan MK, Trebst C, Tucky B, Wei T, Wu L, Baekkevold ES, Lassmann H, Staugaitis SM, Campbell JJ, Ransohoff RM: Human cerebrospinal fluid central memory CD4+ T cells: evidence for trafficking through choroid plexus and meninges via P-selectin. Proc Natl Acad Sci U S A. 2005, 100: 8389-94. Epub 2003 Jun 26.. 10.1073/pnas.1433000100.CrossRef
3.
Zurück zum Zitat Betz LA, Goldstein GW, Katzman R: Blood-Brain-Cerebrospinal Fluid Barriers. Basic Neurochemistry: Molecular, cellular, and medical aspects. Edited by: Siegel GJ. 1989, New York, Raven Press, 4th Ed.: 591-606. Betz LA, Goldstein GW, Katzman R: Blood-Brain-Cerebrospinal Fluid Barriers. Basic Neurochemistry: Molecular, cellular, and medical aspects. Edited by: Siegel GJ. 1989, New York, Raven Press, 4th Ed.: 591-606.
4.
Zurück zum Zitat Wolburg H, Wolburg-Buchholz K, Liebner S, Engelhardt B: OSP/claudin-11, claudin-1 and claudin-2 are present in tight junctions of choroid plexus epithelium of the mouse. Neurosci Lett. 2001, 13: 77-80. 10.1016/S0304-3940(01)01927-9.CrossRef Wolburg H, Wolburg-Buchholz K, Liebner S, Engelhardt B: OSP/claudin-11, claudin-1 and claudin-2 are present in tight junctions of choroid plexus epithelium of the mouse. Neurosci Lett. 2001, 13: 77-80. 10.1016/S0304-3940(01)01927-9.CrossRef
5.
Zurück zum Zitat Carrithers MD, Visintin I, Viret C, Janeway CSJ: Role of genetic background in P selectin-dependent immune surveillance of the central nervous system. J Neuroimmunol. 2002, 129: 51-57. 10.1016/S0165-5728(02)00172-8.CrossRefPubMed Carrithers MD, Visintin I, Viret C, Janeway CSJ: Role of genetic background in P selectin-dependent immune surveillance of the central nervous system. J Neuroimmunol. 2002, 129: 51-57. 10.1016/S0165-5728(02)00172-8.CrossRefPubMed
6.
Zurück zum Zitat Engelhardt B, Wolburg-Buchholz K, Wolburg H: Involvement of the choroid plexus in central nervous system inflammation. Microsc Res Tech. 2001, 52: 112-129. 10.1002/1097-0029(20010101)52:1<112::AID-JEMT13>3.0.CO;2-5.CrossRefPubMed Engelhardt B, Wolburg-Buchholz K, Wolburg H: Involvement of the choroid plexus in central nervous system inflammation. Microsc Res Tech. 2001, 52: 112-129. 10.1002/1097-0029(20010101)52:1<112::AID-JEMT13>3.0.CO;2-5.CrossRefPubMed
7.
Zurück zum Zitat Steffen BJ, Breier G, Butcher EC, Schulz M, Engelhardt B: ICAM-1, VCAM-1, and MAdCAM-1 are expressed on choroid plexus epithelium but not endothelium and mediate binding of lymphocytes in vitro. American Journal of Pathology. 1996, 148: 1819-1838.PubMedCentralPubMed Steffen BJ, Breier G, Butcher EC, Schulz M, Engelhardt B: ICAM-1, VCAM-1, and MAdCAM-1 are expressed on choroid plexus epithelium but not endothelium and mediate binding of lymphocytes in vitro. American Journal of Pathology. 1996, 148: 1819-1838.PubMedCentralPubMed
8.
Zurück zum Zitat Johnson AK, Gross PM: Sensory circumventricular organs and brain homeostatic pathways. FASEB J. 1993, 7: 678-686.PubMed Johnson AK, Gross PM: Sensory circumventricular organs and brain homeostatic pathways. FASEB J. 1993, 7: 678-686.PubMed
9.
Zurück zum Zitat Leonhardt H: Ependym und zirkumventrikuläre Organe. Handbuch der mikroskopischen Anatomie des Menschen. Edited by: Oksche, A. and Vollrath L. 1980, Berlin, Springr Verlag, 177-666. Leonhardt H: Ependym und zirkumventrikuläre Organe. Handbuch der mikroskopischen Anatomie des Menschen. Edited by: Oksche, A. and Vollrath L. 1980, Berlin, Springr Verlag, 177-666.
10.
Zurück zum Zitat Daftar SS, Gore AC: IGF-1 in the brain as a regulator of reproductive neuroendocrine function. Exp Biol Med (Maywood). 2005, 230: 292-306. Daftar SS, Gore AC: IGF-1 in the brain as a regulator of reproductive neuroendocrine function. Exp Biol Med (Maywood). 2005, 230: 292-306.
11.
Zurück zum Zitat McKinley MJ, McAllen RM, Davern P, Giles ME, Penschow J, Sunn N, Uschakov A, Oldfield BJ: The sensory circumventricular organs of the mammalian brain. Adv Anat Embryol Cell Biol. 2003, 172:III-XII: 1-122.CrossRef McKinley MJ, McAllen RM, Davern P, Giles ME, Penschow J, Sunn N, Uschakov A, Oldfield BJ: The sensory circumventricular organs of the mammalian brain. Adv Anat Embryol Cell Biol. 2003, 172:III-XII: 1-122.CrossRef
12.
Zurück zum Zitat Gebke E, Muller AR, Jurzak M, Gerstberger R: Angiotensin II-induced calcium signalling in neurons and astrocytes of rat circumventricular organs. Neuroscience. 1998, 85: 509-520. 10.1016/S0306-4522(97)00601-5.CrossRefPubMed Gebke E, Muller AR, Jurzak M, Gerstberger R: Angiotensin II-induced calcium signalling in neurons and astrocytes of rat circumventricular organs. Neuroscience. 1998, 85: 509-520. 10.1016/S0306-4522(97)00601-5.CrossRefPubMed
13.
Zurück zum Zitat Engelhardt B, Laschinger M, Schulz M, Samulowitz U, Vestweber D, Hoch G: The development of experimental autoimmune encephalomyelitis in the mouse requires alpha4-integrin but not alpha4beta7-integrin. Journal of Clinical Investigation. 1998, 102: 2096-2105.PubMedCentralCrossRefPubMed Engelhardt B, Laschinger M, Schulz M, Samulowitz U, Vestweber D, Hoch G: The development of experimental autoimmune encephalomyelitis in the mouse requires alpha4-integrin but not alpha4beta7-integrin. Journal of Clinical Investigation. 1998, 102: 2096-2105.PubMedCentralCrossRefPubMed
14.
Zurück zum Zitat Engelhardt B, Vestweber D, Hallmann R, Schulz M: E- and P-selectin are not involved in the recruitment of inflammatory cells across the blood-brain barrier in experimental autoimmune encephalomyelitis. Blood. 1997, 90: 4459-4472.PubMed Engelhardt B, Vestweber D, Hallmann R, Schulz M: E- and P-selectin are not involved in the recruitment of inflammatory cells across the blood-brain barrier in experimental autoimmune encephalomyelitis. Blood. 1997, 90: 4459-4472.PubMed
15.
Zurück zum Zitat Hallmann R, Mayer DN, Berg EL, Broermann R, Butcher EC: Novel mouse endothelial cell surface marker is suppressed during differentation of the blood-brain barrier. Developmental Dynamics. 1995, 202: 325-332.CrossRefPubMed Hallmann R, Mayer DN, Berg EL, Broermann R, Butcher EC: Novel mouse endothelial cell surface marker is suppressed during differentation of the blood-brain barrier. Developmental Dynamics. 1995, 202: 325-332.CrossRefPubMed
16.
Zurück zum Zitat Elmquist JK, Scammell TE, Saper CB: Mechanisms of CNS response to systemic immune challenge: the febrile response. Trends Neurosci. 1997, 20: 565-570. 10.1016/S0166-2236(97)01138-7.CrossRefPubMed Elmquist JK, Scammell TE, Saper CB: Mechanisms of CNS response to systemic immune challenge: the febrile response. Trends Neurosci. 1997, 20: 565-570. 10.1016/S0166-2236(97)01138-7.CrossRefPubMed
17.
Zurück zum Zitat Lee HY, Whiteside MB, Herkenham M: Area postrema removal abolishes stimulatory effects of intravenous interleukin-1beta on hypothalamic-pituitary-adrenal axis activity and c-fos mRNA in the hypothalamic paraventricular nucleus. Brain Res Bull. 1998, 46: 495-503. 10.1016/S0361-9230(98)00045-8.CrossRefPubMed Lee HY, Whiteside MB, Herkenham M: Area postrema removal abolishes stimulatory effects of intravenous interleukin-1beta on hypothalamic-pituitary-adrenal axis activity and c-fos mRNA in the hypothalamic paraventricular nucleus. Brain Res Bull. 1998, 46: 495-503. 10.1016/S0361-9230(98)00045-8.CrossRefPubMed
18.
Zurück zum Zitat Sharshar T, Hopkinson NS, Orlikowski D, Annane D: Science review: The brain in sepsis--culprit and victim. Crit Care. 2005, 9: 37-44. Epub 2004 Sep 8.. 10.1186/cc2951.PubMedCentralCrossRefPubMed Sharshar T, Hopkinson NS, Orlikowski D, Annane D: Science review: The brain in sepsis--culprit and victim. Crit Care. 2005, 9: 37-44. Epub 2004 Sep 8.. 10.1186/cc2951.PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Buller KM: Role of circumventricular organs in pro-inflammatory cytokine-induced activation of the hypothalamic-pituitary-adrenal axis. Clin Exp Pharmacol Physiol. 2001, 28: 581-589. 10.1046/j.1440-1681.2001.03490.x.CrossRefPubMed Buller KM: Role of circumventricular organs in pro-inflammatory cytokine-induced activation of the hypothalamic-pituitary-adrenal axis. Clin Exp Pharmacol Physiol. 2001, 28: 581-589. 10.1046/j.1440-1681.2001.03490.x.CrossRefPubMed
20.
Zurück zum Zitat Roth J, Harre EM, Rummel C, Gerstberger R, Hubschle T: Signaling the brain in systemic inflammation: role of sensory circumventricular organs. Front Biosci. 2004, 9: 290-300.CrossRefPubMed Roth J, Harre EM, Rummel C, Gerstberger R, Hubschle T: Signaling the brain in systemic inflammation: role of sensory circumventricular organs. Front Biosci. 2004, 9: 290-300.CrossRefPubMed
21.
Zurück zum Zitat Pedersen EB, McNulty JA, Castro AJ, Fox LM, Zimmer J, Finsen B: Enriched immune-environment of blood-brain barrier deficient areas of normal adult rats. J Neuroimmunol. 1997, 76: 117-131. 10.1016/S0165-5728(97)00038-6.CrossRefPubMed Pedersen EB, McNulty JA, Castro AJ, Fox LM, Zimmer J, Finsen B: Enriched immune-environment of blood-brain barrier deficient areas of normal adult rats. J Neuroimmunol. 1997, 76: 117-131. 10.1016/S0165-5728(97)00038-6.CrossRefPubMed
22.
Zurück zum Zitat Raivich G, Banati R: Brain microglia and blood-derived macrophages: molecular profiles and functional roles in multiple sclerosis and animal models of autoimmune demyelinating disease. Brain Res Brain Res Rev. 2004, 46: 261-281. 10.1016/j.brainresrev.2004.06.006.CrossRefPubMed Raivich G, Banati R: Brain microglia and blood-derived macrophages: molecular profiles and functional roles in multiple sclerosis and animal models of autoimmune demyelinating disease. Brain Res Brain Res Rev. 2004, 46: 261-281. 10.1016/j.brainresrev.2004.06.006.CrossRefPubMed
23.
Zurück zum Zitat Lyck R, Reiss Y, Gerwin N, Greenwood J, Adamson P, Engelhardt B: T cell interaction with ICAM-1/ICAM-2-double-deficient brain endothelium in vitro: the cytoplasmic tail of endothelial ICAM-1 is necessary for transendothelial migration of T cells. Blood. 2003, 102: 3675-83. Epub 2003 Jul 31.. 10.1182/blood-2003-02-0358.CrossRefPubMed Lyck R, Reiss Y, Gerwin N, Greenwood J, Adamson P, Engelhardt B: T cell interaction with ICAM-1/ICAM-2-double-deficient brain endothelium in vitro: the cytoplasmic tail of endothelial ICAM-1 is necessary for transendothelial migration of T cells. Blood. 2003, 102: 3675-83. Epub 2003 Jul 31.. 10.1182/blood-2003-02-0358.CrossRefPubMed
24.
Zurück zum Zitat Laschinger M, Engelhardt B: Interaction of alpha4-integrin with VCAM-1 is involved in adhesion of encephalitogenic T cell blasts to brain endothelium but not in their transendothelial migration in vitro. J Neuroimmunol. 2000, 102: 32-43. 10.1016/S0165-5728(99)00156-3.CrossRefPubMed Laschinger M, Engelhardt B: Interaction of alpha4-integrin with VCAM-1 is involved in adhesion of encephalitogenic T cell blasts to brain endothelium but not in their transendothelial migration in vitro. J Neuroimmunol. 2000, 102: 32-43. 10.1016/S0165-5728(99)00156-3.CrossRefPubMed
25.
Zurück zum Zitat Yednock TA, Cannon C, Fritz LC, Sanchez-Madrid F, Steinman L, Karin N: Prevention of experimental autoimmune encephalomyelitis by antibodies against alpha 4 beta 1 integrin. Nature. 1992, 356: 63-66. 10.1038/356063a0.CrossRefPubMed Yednock TA, Cannon C, Fritz LC, Sanchez-Madrid F, Steinman L, Karin N: Prevention of experimental autoimmune encephalomyelitis by antibodies against alpha 4 beta 1 integrin. Nature. 1992, 356: 63-66. 10.1038/356063a0.CrossRefPubMed
26.
Zurück zum Zitat Steffen BJ, Butcher EC, Engelhardt B: Evidence for involvement of ICAM-1 and VCAM-1 in lymphocyte interaction with endothelium in experimental autoimmune encephalomyelitis in the central nervous system in the SJL/J mouse. Am J Pathol. 1994, 145(N1): 189-201. Steffen BJ, Butcher EC, Engelhardt B: Evidence for involvement of ICAM-1 and VCAM-1 in lymphocyte interaction with endothelium in experimental autoimmune encephalomyelitis in the central nervous system in the SJL/J mouse. Am J Pathol. 1994, 145(N1): 189-201.
Metadaten
Titel
The circumventricular organs participate in the immunopathogenesis of experimental autoimmune encephalomyelitis
verfasst von
Martina Schulz
Britta Engelhardt
Publikationsdatum
01.12.2005
Verlag
BioMed Central
Erschienen in
Fluids and Barriers of the CNS / Ausgabe 1/2005
Elektronische ISSN: 2045-8118
DOI
https://doi.org/10.1186/1743-8454-2-8

Weitere Artikel der Ausgabe 1/2005

Fluids and Barriers of the CNS 1/2005 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Schützt Olivenöl vor dem Tod durch Demenz?

10.05.2024 Morbus Alzheimer Nachrichten

Konsumieren Menschen täglich 7 Gramm Olivenöl, ist ihr Risiko, an einer Demenz zu sterben, um mehr als ein Viertel reduziert – und dies weitgehend unabhängig von ihrer sonstigen Ernährung. Dafür sprechen Auswertungen zweier großer US-Studien.

Bluttest erkennt Parkinson schon zehn Jahre vor der Diagnose

10.05.2024 Parkinson-Krankheit Nachrichten

Ein Bluttest kann abnorm aggregiertes Alpha-Synuclein bei einigen Menschen schon zehn Jahre vor Beginn der motorischen Parkinsonsymptome nachweisen. Mit einem solchen Test lassen sich möglicherweise Prodromalstadien erfassen und die Betroffenen früher behandeln.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Wartezeit nicht kürzer, aber Arbeit flexibler

Psychotherapie Medizin aktuell

Fünf Jahren nach der Neugestaltung der Psychotherapie-Richtlinie wurden jetzt die Effekte der vorgenommenen Änderungen ausgewertet. Das Hauptziel der Novellierung war eine kürzere Wartezeit auf Therapieplätze. Dieses Ziel wurde nicht erreicht, es gab jedoch positive Auswirkungen auf andere Bereiche.

Update Neurologie

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