Active immunization with amyloid-β (Aβ) peptide 1–42 reverses amyloid plaque deposition in the CNS of patients with Alzheimer’s disease and in amyloid precursor protein transgenic mice. However, this treatment may also cause severe, life-threatening meningoencephalitis. Physiological responses to immunization with Aβ1–42 are poorly understood. In this study, we characterized cognitive and immunological consequences of Aβ1–42/CFA immunization in C57BL/6 mice. In contrast to mice immunized with myelin oligodendrocyte glycoprotein (MOG)35–55/CFA or CFA alone, Aβ1–42/CFA immunization resulted in impaired exploratory activity, habituation learning, and spatial-learning abilities in the open field. As morphological substrate of this neurocognitive phenotype, we identified a disseminated, nonfocal immune cell infiltrate in the CNS of Aβ1–42/CFA-immunized animals. In contrast to MOG35–55/CFA and PBS/CFA controls, the majority of infiltrating cells in Aβ1–42/CFA-immunized mice were CD11b+CD14+ and CD45high, indicating their blood-borne monocyte/macrophage origin. Immunization with Aβ1–42/CFA was significantly more potent than immunization with MOG35–55/CFA or CFA alone in activating macrophages in the secondary lymphoid compartment and peripheral tissues. Studies with TLR2/4-deficient mice revealed that the TLR2/4 pathway mediated the Aβ1–42-dependent proinflammatory cytokine release from cells of the innate immune system. In line with this, TLR2/4 knockout mice were protected from cognitive impairment upon immunization with Aβ1–42/CFA. Thus, this study identifies adjuvant effects of Aβ1–42, which result in a clinically relevant neurocognitive phenotype highlighting potential risks of Aβ immunotherapy.

Alzheimer’s disease (AD) is the most common neurodegenerative disorder characterized by compact extracellular plaques, which are largely composed of amyloid-β (Aβ) peptide. Several therapeutic strategies have been developed to eliminate or reduce Aβ deposits within the CNS. Active immunization in which Aβ peptide is combined with an adjuvant to stimulate an Ab response against Aβ was shown to lower brain amyloid burden and partially restore behavioral deficits in animal models (1, 2). These promising observations led to a clinical trial with active immunization using synthetic Aβ1–42. However, the trial was discontinued due to the occurrence of meningoencephalitis in 6% of cases without a clear correlation to the strength of the anti-Aβ Ab response in these patients (35). Furthermore, immunotherapeutic approaches in animal models of AD provided controversial results with respect to cognitive improvements. In amyloid precursor protein (APP) transgenic mice, behavioral tasks were not improved postimmunization (p.i.) with Aβ (6), whereas in another study, spatial memory was recovered in a small subset of immunized mice with very low hippocampal Aβ levels (7).

More recent data indicate that immunization with Aβ peptide provokes activation of microglia, which initiates inflammation within the CNS (8). Whereas microglia seems to play a key role in Aβ clearance, it remains to be determined whether microglial cells also play an active proinflammatory role when exposed to Aβ. Indeed, activated microglial cells located in close proximity to Aβ plaques are found in patients with AD (9) and transgenic mouse models of AD (10). Moreover, it has been shown that Aβ peptide itself provokes activation of microglia, stimulating critical signaling responses that lead to increased IL-6 production, inducing the death of cultured neurons (11). In addition, Aβ peptide was shown to enhance the action of TLR2 and 4 agonists in primary mouse microglial cells (12), suggesting a role of the TLRs in mediating Aβ1–42-driven inflammatory responses.

Whereas the majority of studies on immune interventions targeting Aβ were conducted in APP transgenic mice with conditional overexpression of APP in the CNS, little is known about the systemic and organ specific inflammatory responses upon immunization with Aβ in nontransgenic animals. In this study, we aimed at characterizing: 1) the cognitive profile and the histopathological manifestation of wild-type mice challenged with Aβ1–42 immunization; 2) distinguishing the inflammatory response in Aβ1–42-immunized animals from classical experimental autoimmune encephalomyelitis (EAE); and 3) unraveling the immunological mechanisms behind the inflammatory processes in Aβ1–42-immunized mice in the systemic compartment and within the CNS.

Female C57BL/6 mice were obtained from Charles River Laboratories (Sulzfeld, Germany) and were used in experimental paradigms at the age of 6–8 wk. TLR2/4-deficient mice on the C57BL/6 background were provided by C. Kirschning (Institute of Medical Microbiology, Technische Universität Munich, Munich, Germany). All procedures were conducted in compliance with the local guidelines for animal experimentation.

Animals were immunized s.c. with 100 μg/animal human Aβ1–42 peptide (American Peptide Company, Sunnyvale, CA; EZBiolab, Carmel, CA) emulsified in CFA containing 5 mg/ml Mycobacterium tuberculosis extract (strain H37Ra, DIFCO Laboratories, Detroit, MI). EAE induction was performed by s.c. injection of 100 μg/animal of myelin oligodendrocyte glycoprotein (MOG) peptide 35–55 (Jerini, Berlin, Germany) emulsified in CFA. Control animals received CFA with PBS. On days 0 and 2, all animals were injected with 500 ng/animal pertussis toxin (PTX; Sigma-Aldrich, Munich, Germany) i.p.

Open field.

For evaluation of habituation and visuospatial learning, the open field test was conducted as previously described (13, 14). Briefly, the open field was a square arena (30 × 30 × 40 cm) with clear Plexiglas walls and a grid square floor composed of nine equal quadrants. At the beginning of the test, mice were placed in the center of the open field and left to freely explore. The total number of quadrant borders the mice crossed and the number of rearings were counted by a blinded observer during a 10-min observation period. Baseline values were assessed prior to immunization.

According to O’Keefe and Nadel’s cognitive map theory (15), exploration of a novel environment is used by the animal to construct a cognitive map, and activity wanes once such a map is established. Therefore, habituation to an open field is a measure of memory, and the faster a cognitive map is established, the sooner exploration activity will decrease. To asses a habituation learning measure (habituation learning index), the difference of crossed segments in the first and last 150 s of each 10-min observation period was determined (16). The open field test was repeated every 3 d.

Clinical signs of EAE were ranked with an established score from 0–5: 0 (normal); 1 (tail limpness), 2 (paraparesis with clumsy gait); 3 (hind limb paralysis); 4 (hind limb and forelimb paralysis); and 5 (death). All ratings were done by observers blinded to the treatment.

Visuospatial learning task.

Visuospatial learning performance was tested in the open field paradigm with slight modifications from published protocols (17). For ethical reasons, the water maze paradigm was not applied, as some of the animals in the MOG35–55/CFA-immunized control group developed severe pareses.

For 3 consecutive learning d, mice were placed into the open field in which two identical objects (bottles) in terms of height, color, shape, and surface texture were located. Spatial configuration did not change for three training sessions. On day 4, the bottle in the corner was moved to the opposite corner, leaving the configuration and distance of the objects undisturbed. The total exploration time for each object was determined during a 10-min observation period. Object exploration was defined as physical contact with the bottle by mouth, vibrissae, and forepaws. Compassing or sitting inactively next to the objects was not regarded as object exploration. For statistical evaluation, the initial exploration time for each stimulus in the first session was calculated, and the relative change in exploration time of the replaced stimulus in the fourth session was determined.

For systemic depletion of macrophages, mice were given i.p. injections of clodronate liposomes according to established protocols (18). Briefly, mice received an initial dose of 100 mg/kg clodronate liposomes (kindly provided by R. Schwendener, Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland) followed by subsequent injections of 50 mg/kg every fourth day. Control mice were injected with empty liposomes. Immunization with PBS/CFA or Aβ1–42/CFA plus PTX was performed 3 d after the initial clodronate injection.

Serum Abs against human and murine Aβ1–42 peptides were determined by ELISA according to established protocols (19). On day 28 p.i. with PBS/CFA or human Aβ1–42/CFA, anti-Aβ1–42 Abs in the sera of the animals were captured by solid-phase human or murine Aβ1–42 followed by detection of mouse IgG with HRP-labeled goat anti-mouse IgG (AbD Serotec, Raleigh, NC).

Cells immunoreactive for CD11b and CD11b/CD11c were isolated from naive mouse spleen tissue by magnetic cell sorting with MACS (Miltenyi Biotec, Bergisch Gladbach, Germany) according to the manufacturer’s instructions. Purity of cells (>90%) was confirmed by FACS analysis.

Primary macrophages were isolated from the peritoneal cavity of mice 12 d p.i. according to previously published protocols (20). For assessing cytokine production, these cells were cultured (2 × 106 cells/ml) in media (DMEM medium containing 10% FCS, nonessential amino acids, HEPES, l-glutamine, and antibiotics) for 48 h at 37°C in a humidified incubator at 5% CO2. For gene expression studies, mRNA was isolated directly after harvesting the cells from the peritoneal cavity.

Mice were perfused with cold PBS through the left cardiac ventricle on day 10 p.i. The brain was dissected, and the spinal cord was flushed out by hydrostatic pressure. CNS tissue was cut into pieces and digested with 2.5 mg/ml Collagenase D (Roche Diagnostics, Indianapolis, IN) and 1 mg/ml DNAse I (Sigma-Aldrich) in DMEM medium at 37°C for 40 min. Single-cell suspensions were prepared using a 70-μm cell strainer followed by percoll gradient centrifugation (70/37%). Mononuclear cells were removed from the interphase, washed, and resuspended in culture medium.

Mononuclear cells were stained for CD11b, CD14, and CD45 (BD Biosciences, San Jose, CA) according to the manufacturer’s instructions. Analyses were performed on a Dako CyAn flow cytometer system (DakoCytomation, Glostrup, Denmark). Flow cytometric data were analyzed with FlowJo (Tree Star, Ashland, OR).

Lyophilized human Aβ1–42 peptide (obtained from American Peptide Company or EZBiolab) was reconstituted with PBS at a concentration of 2 mg/ml. Dissolved peptide was stored at 4°C for up to 48 h. Where indicated, murine Aβ1–42 (American Peptide Company) was used. In stimulation experiments, CD11b+ and CD11b+CD11c+ cells (2 × 106 cells/ml) were stimulated with different concentrations of Aβ1–42 peptide (0.1–50 μg/ml) or 100 ng/ml LPS (Sigma-Aldrich) for 48 h at 37°C in culture medium in a humidified incubator at 5% CO2.

Cytokine levels were determined in sera and culture supernatants. Cell culture supernatants were collected after indicated incubation periods and stored at −80°C until analysis. Levels of IL-1β, IL-6, IFN-γ, and TNF were measured by commercial ELISA kits (R&D Systems, Minneapolis, MN) according to the manufacturer’s instructions.

Isolation of RNA (RNeasy, Quiagen, Hilden, Germany) from whole brain tissue and immune cells, its quantification, and the RT reactions (High-capacity RT Kit, Applied Biosystems, Foster City, CA) were performed according to established protocols. Expression of mRNA of target genes and the endogenous control gene GAPDH was assessed by real-time PCR (with TaqMan Gene Expression Assay products on StepOne Plus PCR System, Applied Biosystems) according to the manufacturer’s recommendations. Expression levels for each gene of interest were calculated by normalizing the quantified mRNA amount to GAPDH. Relative gene expression was determined and used to test significance between different groups. The following gene expression assays (Applied Biosystems) were used: IL-1β (Mm00434228_m1), IL-6 (Mm00446190_m1), CD14 (Mm00438094_g1), glial fibrillary acidic protein (GFAP; Mm01253033_m1), S100A8 (Mm00496696_g1), and TNF (Mm00443258_m1).

Mice were anesthesized with isoflurane and perfused with ice-cold PBS and 4% paraformaldehyde. Brains were dissected and embedded in paraffin. Immunohistochemistry was performed with a rat Ab against mouse Mac-3 (1:200; clone M3/84, BD Biosciences) as described previously (21, 22). Briefly, tissues were pretreated by microwaving in 10 mM citrate buffer (pH 6) for two cycles of 5 min each. Immunolabeling was detected by the avidin-peroxidase method and visualized with diaminobenzidine by incubation for 5 min. Control sections were incubated in the absence of primary Ab or with nonimmune sera. Slides were counterstained with hematoxylin and coverslipped.

For statistical comparisons, a one-way multiple-range ANOVA test for multiple comparisons was employed. Unpaired t tests were used for comparison of two groups. Values of p < 0.05 were considered significant. Graphs were generated using GraphPad Prism software (GraphPad, San Diego, CA).

Because APP and its cleavage products, the Aβ peptides, are present in the normal CNS, we wished to investigate in more detail how immunotherapeutic approaches designed to remove Aβ deposits interfere with regular functions of the CNS. Active immunization with Aβ1–42/CFA significantly altered the psychomotor and cognitive phenotype of mice in comparison with various control groups. Observations in the open field revealed pronounced deficits in three cognitive parameters: first, open field testing of Aβ1–42/CFA-immunized mice showed a significant reduction of locomotion (Fig. 1A) as compared with MOG/CFA- or PBS/CFA-immunized animals. Changes in locomotion were detected as early as on day 10 p.i., and reduced locomotion in Aβ1–42-immunized mice persisted over the entire observation period until day 28. Reduced rearing behavior was detected already on day 4 and persisted until day 18 (Fig. 1B). Second, we observed a significant decrease in habituation learning ability. Whereas control animals showed habituation to a persisting environment by reduction of exploration over time, Aβ1–42/CFA-immunized mice had a significantly lower habituation learning index (Fig. 1C) from day 3 p.i. Even compared with MOG35–55/CFA-immunized mice (EAE scores are shown in Fig. 1D), we found significant differences in Aβ1–42- immunized mice on days 10, 17, and 28 p.i. Experiments in aged mice (12 mo old) revealed similar deficits in explorative behavior after Aβ1–42/CFA immunization (Supplemental Fig. 1). Third, we found that mice immunized with Aβ1–42/CFA developed profound deficits in visuospatial learning both in the acute (observation between days 9 and 14 p.i.) and chronic (observation between days 23 and 28 p.i.) phases of disease (Fig. 2A, 2B). As compared with controls, Aβ1–42/CFA-immunized animals spent significantly less time to explore a novel stimulus in a known environment (reduced memory gain) both in the acute and chronic phases of disease. Together, these behavioral data indicate a profound and persistent decline in motivational and cognitive performance in Aβ1–42/CFA-immunized animals.

FIGURE 1.

Active immunization with Aβ1–42 impairs psychomotor functioning and habituation learning in the open field. Groups of female C57BL/6 mice (n = 10/group) were immunized with PBS/CFA, MOG35–55/CFA, or Aβ1–42/CFA plus PTX and evaluated for locomotion (A) and explorative behavior (B) as measured by the number of crossed quadrants and rearing events at different time points p.i. Habituation learning was assessed in a setting that tested the habituation to visuospatial cues (C; for habituation learning index, see 1Materials and Methods). Mean performances and SEM are illustrated for each group preimmunization and p.i. At least three independent experiments were performed. D, EAE scores illustrating that paralytic disease in the MOG35–55/CFA group started around day 11, but did not mar the specific readout parameters of the open field tests. Statistical comparisons are based on the relative change to baseline performance. “ma” and “ca” denote significant differences between the MOG35–55/CFA versus Aβ1–42/CFA and PBS/CFA versus Aβ1–42/CFA groups, respectively. *p < 0.05; **p < 0.01.

FIGURE 1.

Active immunization with Aβ1–42 impairs psychomotor functioning and habituation learning in the open field. Groups of female C57BL/6 mice (n = 10/group) were immunized with PBS/CFA, MOG35–55/CFA, or Aβ1–42/CFA plus PTX and evaluated for locomotion (A) and explorative behavior (B) as measured by the number of crossed quadrants and rearing events at different time points p.i. Habituation learning was assessed in a setting that tested the habituation to visuospatial cues (C; for habituation learning index, see 1Materials and Methods). Mean performances and SEM are illustrated for each group preimmunization and p.i. At least three independent experiments were performed. D, EAE scores illustrating that paralytic disease in the MOG35–55/CFA group started around day 11, but did not mar the specific readout parameters of the open field tests. Statistical comparisons are based on the relative change to baseline performance. “ma” and “ca” denote significant differences between the MOG35–55/CFA versus Aβ1–42/CFA and PBS/CFA versus Aβ1–42/CFA groups, respectively. *p < 0.05; **p < 0.01.

Close modal
FIGURE 2.

Active immunization with Aβ1–42 impairs performance in a visuospatial object recognition task in the open field. Groups of female C57BL/6 mice were immunized with PBS/CFA, MOG35–55/CFA, or Aβ1–42/CFA plus PTX and evaluated in a visuospatial object recognition paradigm in the acute phase (A, acquisition period between days 9 and 14 p.i.) and the chronic phase of disease (B, acquisition period between days 23 and 28 p.i.). The fraction of additional exploration time of the novel stimulus in a habituated environment is illustrated for each individual mouse. Dashed lines indicate mean performances of individual groups. Results were replicated at least three times. Note that in contrast to Aβ1–42/CFA immunization, immunization with MOG35–55/CFA induced paralytic disease, but did not affect visuospatial memory performance. **p < 0.01; ***p < 0.001.

FIGURE 2.

Active immunization with Aβ1–42 impairs performance in a visuospatial object recognition task in the open field. Groups of female C57BL/6 mice were immunized with PBS/CFA, MOG35–55/CFA, or Aβ1–42/CFA plus PTX and evaluated in a visuospatial object recognition paradigm in the acute phase (A, acquisition period between days 9 and 14 p.i.) and the chronic phase of disease (B, acquisition period between days 23 and 28 p.i.). The fraction of additional exploration time of the novel stimulus in a habituated environment is illustrated for each individual mouse. Dashed lines indicate mean performances of individual groups. Results were replicated at least three times. Note that in contrast to Aβ1–42/CFA immunization, immunization with MOG35–55/CFA induced paralytic disease, but did not affect visuospatial memory performance. **p < 0.01; ***p < 0.001.

Close modal

In contrast to immunization with MOG35–55/CFA or CFA only, immunization with Aβ1–42 emulsified in CFA induced profound and persistent behavioral changes in wild-type animals. To investigate the potential immunological substrate of this behavioral phenotype, we performed immunohistochemical studies of CNS tissue specimens 18 d p.i. Immunohistochemistry revealed perivascular and subpial infiltrates of mononuclear cells in the brain and brainstem of Aβ1–42/CFA-immunized mice (Fig. 3B), but not in PBS/CFA controls (Fig. 3A). These infiltrates in the hippocampal region mainly consisted of macrophages as shown by MAC-3 staining with few CD3+ T cells. Infiltrates in Aβ1–42/CFA-immunized mice (Fig. 3D) were disseminated and nonfocal, whereas MOG35–55/CFA controls showed focal meningeal and perivascular inflammatory infiltrates (Fig. 3C). We wondered whether the cellular infiltrate in MOG35–55/CFA- versus Aβ1–42/CFA-immunized mice was also quantitatively different. To quantify various immune cell populations in the CNS of immunized mice, we isolated mononuclear cells from the CNS and performed flow cytometric analysis. Because behavioral differences between the groups of MOG35–55/CFA-versus Aβ1–42/CFA-immunized animals were evident as early as 10 d p.i., whereas animals in the MOG35–55/CFA group did not yet show signs of paralytic disease at this time point, we chose to perform quantitative analyses of CNS infiltrates on day 10 p.i. In PBS/CFA-immunized control animals, most of the CNS-derived CD11b+ cells were CD45low, indicating their microglial origin (Fig. 4A, 4D, 4G). Whereas in MOG35–55/CFA-immunized mice, T cells (CD11b-CD45high) were already starting to accumulate in the CNS on day 10 p.i., the majority of CD11b+ cells were still CD45low, again suggesting their microglial origin (Fig. 4B, 4E, 4G). In contrast, the majority of CD11b+ cells isolated from the CNS of Aβ1–42/CFA-immunized mice were CD45high, indicating that these CD11b+ cells were macrophages that had invaded the CNS as early as on day 10 p.i. (Fig. 4C, 4F, 4G). Moreover, the fraction of CD14+ cells within the population of CD11b+CD45high macrophages in the CNS was significantly higher in Aβ1–42/CFA-immunized mice than in either control group (Fig. 4H, 4I). Together, these data indicate that Aβ1–42/CFA immunization leads to early and massive recruitment of blood-borne macrophages into the CNS.

FIGURE 3.

Disseminated, nonfocal immune cell infiltration in the CNS of Aβ1–42/CFA-immunized mice. Mice were immunized with PBS/CFA, MOG35–55/CFA, or Aβ1–42/CFA plus PTX and prepared for histopathological analysis 18 d p.i. Representative MAC-3 immunostainings (original magnification ×20) of coronar sections from the hippocampus region prepared from PBS/CFA (A) and Aβ1–42/CFA-immunized (B) mice are shown. Vessels with perivascular immune cell infiltrates (original magnification ×40) located in the cerebrum of MOG35–55/CFA (C), and Aβ1–42/CFA-immunized (D) mice are illustrated. Note that in Aβ1–42/CFA-immunized mice, nonfocal infiltrates of macrophages are widely distributed within the hippocampus region and other regions of the cerebrum. PBS/CFA-immunized mice are largely devoid of infiltrating cells, whereas MOG35–55/CFA-immunized animals showed EAE-typical focal meningeal and perivascular infiltrates.

FIGURE 3.

Disseminated, nonfocal immune cell infiltration in the CNS of Aβ1–42/CFA-immunized mice. Mice were immunized with PBS/CFA, MOG35–55/CFA, or Aβ1–42/CFA plus PTX and prepared for histopathological analysis 18 d p.i. Representative MAC-3 immunostainings (original magnification ×20) of coronar sections from the hippocampus region prepared from PBS/CFA (A) and Aβ1–42/CFA-immunized (B) mice are shown. Vessels with perivascular immune cell infiltrates (original magnification ×40) located in the cerebrum of MOG35–55/CFA (C), and Aβ1–42/CFA-immunized (D) mice are illustrated. Note that in Aβ1–42/CFA-immunized mice, nonfocal infiltrates of macrophages are widely distributed within the hippocampus region and other regions of the cerebrum. PBS/CFA-immunized mice are largely devoid of infiltrating cells, whereas MOG35–55/CFA-immunized animals showed EAE-typical focal meningeal and perivascular infiltrates.

Close modal
FIGURE 4.

Infiltrating cells in Aβ1–42/CFA-immunized animals are primarily of blood-borne macrophage origin. Mononuclear cells were isolated from the CNS of PBS/CFA, MOG35–55/CFA, and Aβ1–42/CFA-immunized animals on day 10 p.i. and analyzed by flow cytometry. Representative cytograms (AC) and histograms (DF) are shown to illustrate the fractions of CD11b+CD45low cells (microglia) and CD11b+CD45high cells (macrophages) in PBS/CFA, MOG35–55/CFA, and Aβ1–42/CFA-immunized animals, respectively. G, Ratio of macrophages versus microglial cells in the CNS of the three experimental groups. One symbol represents one animal. Note that in contrast to PBS/CFA and MOG35–55/CFA-immunized controls, the majority of cells isolated from the CNS of Aβ1–42/CFA-immunized mice on day 10 p.i. were CD11b+CD14+CD45high cells (H, I), indicating their macrophage origin. *p < 0.05 as determined by Mann-Whitney U test.

FIGURE 4.

Infiltrating cells in Aβ1–42/CFA-immunized animals are primarily of blood-borne macrophage origin. Mononuclear cells were isolated from the CNS of PBS/CFA, MOG35–55/CFA, and Aβ1–42/CFA-immunized animals on day 10 p.i. and analyzed by flow cytometry. Representative cytograms (AC) and histograms (DF) are shown to illustrate the fractions of CD11b+CD45low cells (microglia) and CD11b+CD45high cells (macrophages) in PBS/CFA, MOG35–55/CFA, and Aβ1–42/CFA-immunized animals, respectively. G, Ratio of macrophages versus microglial cells in the CNS of the three experimental groups. One symbol represents one animal. Note that in contrast to PBS/CFA and MOG35–55/CFA-immunized controls, the majority of cells isolated from the CNS of Aβ1–42/CFA-immunized mice on day 10 p.i. were CD11b+CD14+CD45high cells (H, I), indicating their macrophage origin. *p < 0.05 as determined by Mann-Whitney U test.

Close modal

Consistent with the immunohistochemical and flow cytometric analyses, the expression of macrophage-associated genes, such as S100A8 and CD14 (Fig. 5A, 5B), was upregulated in whole brain tissue of Aβ1–42/CFA-immunized animals compared with PBS/CFA and MOG35–55/CFA controls. Quantitative RT-PCR from whole brain tissue isolated from Aβ1–42/CFA-immunized mice demonstrated a 10-fold higher expression of S100A8 as compared PBS/CFA-immunized animals and a 3-fold higher expression as compared with MOG35–55/CFA-immunized mice (Fig. 5A). CD14 expression was ∼2-fold increased as compared with either control group (Fig. 5B). When comparing the CNS parenchyma between the groups at late stages of the disease (4 wk p.i.), we found prominent signs of astrogliosis in the Aβ1–42/CFA-immunized mice as determined by a disproportionate upregulation of GFAP mRNA expression in Aβ1–42/CFA-immunized animals (Fig. 5C).

FIGURE 5.

1–42/CFA immunization induces mRNA expression of genes related to macrophage and glia cell activation. The expression of macrophage-associated genes was quantified by RT-PCR in whole CNS tissue. Expression of S100A8 (A) and CD14 (B) was significantly higher in Aβ1–42/CFA-immunized mice compared with controls. Increased astrogliosis was confirmed in Aβ1–42/CFA-immunized mice by quantitative GFAP expression analysis in whole brain tissue 4 wk p.i. (C). Numbers represent the relative gene expression for individual animals as determined by RT-PCR. Dashes indicate mean relative gene expression of individual groups. *p < 0.05; **p < 0.01; ***p < 0.001.

FIGURE 5.

1–42/CFA immunization induces mRNA expression of genes related to macrophage and glia cell activation. The expression of macrophage-associated genes was quantified by RT-PCR in whole CNS tissue. Expression of S100A8 (A) and CD14 (B) was significantly higher in Aβ1–42/CFA-immunized mice compared with controls. Increased astrogliosis was confirmed in Aβ1–42/CFA-immunized mice by quantitative GFAP expression analysis in whole brain tissue 4 wk p.i. (C). Numbers represent the relative gene expression for individual animals as determined by RT-PCR. Dashes indicate mean relative gene expression of individual groups. *p < 0.05; **p < 0.01; ***p < 0.001.

Close modal

Cognitive changes in Aβ1–42/CFA-immunized mice developed in the absence of focal neurologic symptoms. We wondered whether systemic and CNS specific release of inflammatory cytokines induced by immunization with Aβ1–42/CFA was responsible for the neurocognitive phenotype. To test this hypothesis, we determined the level of TNF in the sera of mice immunized with Aβ1–42/CFA. On day 15 p.i., serum TNF was significantly increased in Aβ1–42/CFA-immunized mice as compared with control groups (Fig. 6A). To identify possible cellular sources of TNF, we measured both the expression and the production of TNF and IL-6 in peritoneal macrophages of the various experimental groups. Consistent with the elevated serum concentration of TNF, peritoneal macrophages isolated from Aβ1–42/CFA-immunized mice showed a higher expression TNF, IL-6, CD14, and S100A8 on a per-cell basis and higher secretion of TNF and IL-6 into the culture supernatant than macrophages that were isolated from MOG/CFA- or CFA only-immunized mice (Fig. 6B, 6C). Taken together, these data suggest that immunization with Aβ1–42/CFA induces an exaggerated systemic inflammatory response by activating cells of the innate immune system.

FIGURE 6.

1–42/CFA immunization induces systemic inflammation. A, Serum TNF levels were measured in groups of mice immunized with PBS/CFA, MOG35–55/PBS, or Aβ1–42/CFA 15 d p.i. Cytokine levels of single animals are depicted; dashes indicate mean concentrations. B and C, Peritoneal macrophages were isolated from PBS/CFA, MOG35–55/CFA, or Aβ1–42/CFA-immunized mice and tested for gene expression by quantitative RT-PCR directly ex vivo. The n-fold difference in gene expression of macrophages from Aβ1–42/CFA- and MOG35–55/CFA-immunized mice relative to the PBS/CFA group is shown (B). To confirm the mRNA data on the protein level, peritoneal macrophages were isolated from the individual groups of mice and cultured without further stimulation for 48 h. Secretion of IL-6 and TNF in the culture supernatant was measured by ELISA (C). Mean cytokine concentrations plus SD are shown. Similar results were obtained in three independent experiments. **p < 0.01.

FIGURE 6.

1–42/CFA immunization induces systemic inflammation. A, Serum TNF levels were measured in groups of mice immunized with PBS/CFA, MOG35–55/PBS, or Aβ1–42/CFA 15 d p.i. Cytokine levels of single animals are depicted; dashes indicate mean concentrations. B and C, Peritoneal macrophages were isolated from PBS/CFA, MOG35–55/CFA, or Aβ1–42/CFA-immunized mice and tested for gene expression by quantitative RT-PCR directly ex vivo. The n-fold difference in gene expression of macrophages from Aβ1–42/CFA- and MOG35–55/CFA-immunized mice relative to the PBS/CFA group is shown (B). To confirm the mRNA data on the protein level, peritoneal macrophages were isolated from the individual groups of mice and cultured without further stimulation for 48 h. Secretion of IL-6 and TNF in the culture supernatant was measured by ELISA (C). Mean cytokine concentrations plus SD are shown. Similar results were obtained in three independent experiments. **p < 0.01.

Close modal

To assess the functional relevance of systemic activation of macrophages and their recruitment to the CNS for the neurocognitive phenotype of Aβ1–42/CFA-immunized mice, we performed macrophage-depletion experiments. Clodronate liposomes were used to deplete macrophages in the secondary lymphoid tissues prior to immunization with Aβ1–42/CFA or PBS/CFA. The depletion of macrophages by clodronate liposomes prevented Aβ1–42-induced effects on psychomotor behavior (Fig. 7). In contrast to control mice receiving empty liposomes, macrophage-depleted animals did not show impaired locomotion (Fig. 7A), rearing (Fig. 7B), and habituation performance (Fig. 7C) as a result of challenge with Aβ1–42/CFA, suggesting that peripheral macrophages are crucial effector cells in inducing the clinical phenotype of Aβ1–42/CFA-challenged animals. Accordingly, resident microglial cells (CD11b+CD45low) and not blood-borne macrophages (CD11b+CD45high) constituted the majority of CD11b+ cells in the CNS of macrophage-depleted Aβ1–42/CFA-immunized mice (Supplemental Fig. 3). Thus, clodronate administration was able to abrogate both the behavioral and the immunopathological phenotype in Aβ1–42/CFA-immunized mice.

FIGURE 7.

1–42/CFA immunization does not affect the behavioral phenotype in macrophage-depleted mice. Groups of female wild-type mice (n = 5/group) were control treated or depleted of macrophages by clodronate as described in 1Materials and Methods followed by immunization with PBS/CFA or Aβ1–42/CFA plus PTX. Mice were tested for locomotion (number of crossed quadrants in the open field) (A), rearing behavior (B), and habituation (C) preimmunization [2 d postdepletion, BL] and on day 10 p.i. The mean performances are summarized for both depletion and control groups after PBS/CFA or Aβ1–42/CFA challenge. Statistical comparisons are based on the surplus effect of Aβ1–42/CFA immunization as compared with PBS/CFA immunization within the respective treatment group. Note that the surplus effect of Aβ1–42/CFA immunization was abrogated in the clodronate group. **p < 0.01. BL, baseline.

FIGURE 7.

1–42/CFA immunization does not affect the behavioral phenotype in macrophage-depleted mice. Groups of female wild-type mice (n = 5/group) were control treated or depleted of macrophages by clodronate as described in 1Materials and Methods followed by immunization with PBS/CFA or Aβ1–42/CFA plus PTX. Mice were tested for locomotion (number of crossed quadrants in the open field) (A), rearing behavior (B), and habituation (C) preimmunization [2 d postdepletion, BL] and on day 10 p.i. The mean performances are summarized for both depletion and control groups after PBS/CFA or Aβ1–42/CFA challenge. Statistical comparisons are based on the surplus effect of Aβ1–42/CFA immunization as compared with PBS/CFA immunization within the respective treatment group. Note that the surplus effect of Aβ1–42/CFA immunization was abrogated in the clodronate group. **p < 0.01. BL, baseline.

Close modal

Because we observed a profound activation of the innate immune system p.i. with Aβ1–42, we investigated the stimulatory properties of Aβ peptide in vitro and tested the relevance of specific TLR systems that have been implicated with immunostimulatory effects of Aβ peptide in previous studies. It has been reported that the activation of microglial cells by Aβ peptide requires both TLR2 and TLR4 pathways to activate intracellular signaling (23). In this study, stimulatory effects of Aβ1–42 on CD11b+ macrophages and CD11b+CD11c+ dendritic cells isolated from naive wild-type and TLR2/4-deficient mice were evaluated in vitro. Aβ1–42 induced large amounts of IL-6 and TNF in macrophages (Fig. 8A, 8B) and IFN-γ in dendritic cells from wild-type mice in a dose-dependent manner (Fig. 8C). Similar effects were observed poststimulation with murine Aβ1–42 peptide (Supplemental Fig. 2). In contrast, this effect was not detected in macrophages and dendritic cells derived from TLR2/4-deficient mice, suggesting that either TLR2 or TLR4 or the combined activation of these TLRs mediate the stimulatory effect of Aβ1–42.

FIGURE 8.

1–42 induces IL-6 and TNF production in naive macrophages and IFN-γ in dendritic cells through activation of TLR2 and TLR4. MACS purified CD11b+ cells (A, B, macrophages) and CD11b+CD11c+ cells (C, dendritic cells) from untreated wild-type or TLR2/4-deficient mice were stimulated with increasing concentrations of Aβ1–42 for 48 h. Levels of IL-6, TNF, and IFN-γ were determined in the supernatants by ELISA. Data are representative of three independent experiments.

FIGURE 8.

1–42 induces IL-6 and TNF production in naive macrophages and IFN-γ in dendritic cells through activation of TLR2 and TLR4. MACS purified CD11b+ cells (A, B, macrophages) and CD11b+CD11c+ cells (C, dendritic cells) from untreated wild-type or TLR2/4-deficient mice were stimulated with increasing concentrations of Aβ1–42 for 48 h. Levels of IL-6, TNF, and IFN-γ were determined in the supernatants by ELISA. Data are representative of three independent experiments.

Close modal

To corroborate whether activation of the TLR2/4 pathway by Aβ1–42 was relevant in vivo, we immunized TLR2/4 knockout (KO) animals with Aβ1–42/CFA. We consistently observed a differential disease-promoting effect of Aβ1–42/CFA versus PBS/CFA treatment in wild-type animals. Indeed, we determined a significant decrease in locomotion and rearing in wild-type C57BL/6 mice immunized with Aβ1–42 as compared with immunization with CFA only (Fig. 9). In contrast, we did not find any additional neurocognitive phenotype (surplus effect) upon immunization with Aβ1–42/CFA as compared with the CFA only condition in TLR2/4-deficient mice. When evaluating the surplus effect induced by Aβ1–42/CFA immunization in wild-type animals versus TLR2/4 KO mice, the differences were significant as of day 4 p.i. regarding locomotion and as of day 8 with respect to the rearing behavior. Taken together, these data corroborate the critical involvement of the TLR2/4 pathways in the macrophage-induced behavioral changes following active immunization with Aβ1–42 in vivo.

FIGURE 9.

1–42/CFA immunization does not affect the behavioral phenotype in TLR2/4-deficient mice. TLR2/4-deficient and wild-type mice (n = 8/group) were immunized with PBS/CFA or Aβ1–42/CFA and evaluated for locomotion (A) and explorative behavior as measured by the number of crossed quadrants and rearing events (B) at different time points p.i. The mean performances pre- and p.i. are summarized for both wild-type and TLR2/4 KO mice upon PBS/CFA or Aβ1–42/CFA challenge. Statistical comparisons are based on the surplus effect of Aβ1–42/CFA as compared with PBS/CFA challenge within the respective mouse strain. Data were reproduced in three different experiments. Note that in contrast to wild-type mice, TLR2/4-deficient mice were resistant to Aβ1–42/CFA induced neurocognitive impairment. Significant differences are indicated. *p < 0.05; ***p < 0.001.

FIGURE 9.

1–42/CFA immunization does not affect the behavioral phenotype in TLR2/4-deficient mice. TLR2/4-deficient and wild-type mice (n = 8/group) were immunized with PBS/CFA or Aβ1–42/CFA and evaluated for locomotion (A) and explorative behavior as measured by the number of crossed quadrants and rearing events (B) at different time points p.i. The mean performances pre- and p.i. are summarized for both wild-type and TLR2/4 KO mice upon PBS/CFA or Aβ1–42/CFA challenge. Statistical comparisons are based on the surplus effect of Aβ1–42/CFA as compared with PBS/CFA challenge within the respective mouse strain. Data were reproduced in three different experiments. Note that in contrast to wild-type mice, TLR2/4-deficient mice were resistant to Aβ1–42/CFA induced neurocognitive impairment. Significant differences are indicated. *p < 0.05; ***p < 0.001.

Close modal

In this study, we showed that active immunization with Aβ1–42/CFA induced sustained cognitive and behavioral impairment in wild-type C57BL/6 mice. We identified a disseminated, nonfocal infiltrate of CD11b+CD14+CD45high cells in the CNS of Aβ1–42/CFA-immunized mice. We propose that this infiltrate of activated macrophages represented the immunopathogenetic correlate of the neurocognitive phenotype in Aβ1–42/CFA-immunized mice because challenge with Aβ1–42/CFA failed to induce neurocognitive impairment in animals that had been depleted of macrophages. Furthermore, immunization with Aβ1–42/CFA induced a systemic inflammatory response including the systemic release of cytokines. Peritoneal macrophages from Aβ1–42/CFA-immunized animals were characterized by an increased activation state as compared with MOG35–55/CFA-immunized mice, suggesting the capacity of Aβ peptide to activate the innate immune system in a manner reminiscent of pathogen-associated molecular patterns (PAMPs). Using TLR2/4-deficient mice, we showed that the TLR2/4 pathway mediated the Aβ1–42-induced proinflammatory cytokine release from cells of the innate immune system. Accordingly, TLR2/4 KO mice were protected from cognitive impairment upon immunization with Aβ1–42/CFA. We concluded that vaccination with Aβ1–42/CFA lead to the activation of innate immune cells in the systemic and CNS compartments in a TLR-dependent manner. Thus, this study identified adjuvant effects of Aβ1–42, which resulted in a clinically relevant and sustained neurocognitive phenotype.

Induction of EAE with myelin Ags emulsified in CFA is a widely used model to study autoimmune diseases of the CNS, such as multiple sclerosis. We used classical MOG35–55/CFA-induced EAE as a control condition for our vaccination protocol with Aβ1–42/CFA. MOG35–55-induced EAE has been extensively investigated (2427). In MOG35–55/CFA-induced EAE, focal perivascular and parenchymal infiltrates of T cells and macrophages primarily in the spinal cord lead to demyelination and axonal damage resulting in paralytic disease. However, less is known about the neurocognitive status in MOG35–55/CFA-immunized animals. A depression-like syndrome is reported, but cognitive alterations are rarely seen (28). In contrast, only a few days p.i. with Aβ1–42/CFA, mice exhibited a significant decrease in their psychomotor performance as well as deficient habitual learning abilities and impaired performance in complex visuospatial tasks in the absence of apparent focal neurologic deficits. Although neurocognitive phenotypes have been extensively characterized in previous reports and in our present study, memory performance in mice can only be captured in the context of locomotive behavior tasks that reflect both motivational and cognitive components. Dissection of these components is not possible in the absence of verbal communication. Thus, we cannot exclude that the extent of exploration in our setup was also influenced by the lack of motivation due to systemic release of proinflammatory cytokines.

The clinical syndrome exhibited by Aβ1–42/CFA-immunized mice was reminiscent of the apathic condition that was the result of a cytokine release syndrome. In fact, deficits in visuospatial tasks are reported in mice injected with LPS. After LPS treatment, mice show impaired performance in tests of cognition that require effective integration of new information to complete a spatial task (29). A further study in mice provides evidence for hippocampus-dependent learning and memory impairment after LPS injection (30). Systemic administration of LPS is reported to induce TLR4-dependent secretion of proinflammatory cytokines such as IL-1β, IL-6, and TNF in the CNS (3134). Furthermore, activation of TLR4 by LPS increases IFN-γ levels in mice and stimulates IDO in peripheral tissues and the brain (35). IDO activation results in decreased tryptophan levels and increased production of kynurenine promoting depression-like behavior in mice (36). LPS-induced sickness behavior is characterized both by systemic inflammation (35) and activation of local microglial cells (37) in the absence of cellular infiltrates in the CNS.

In contrast to detrimental effects of proinflammatory cytokines on cognitive functioning, other cytokines may have beneficial effects on cognitive processes in the normal brain (38, 39). For example, IL-4 in meningeal T cells is involved in maintaining cognitive abilities in spatial learning and memory tasks (40). In the absence of T cell-derived IL-4, mononuclear cells in the meningeal compartment become activated and contribute to impaired learning abilities. In line with this concept, we showed that extensive production of proinflammatory cytokines like TNF and IL-6 by Aβ1–42-activated macrophages resulted in reduced cognitive performance.

A study by Furlan et al. (19) reports an Aβ1–42-specific CD4+ T cell response and mild neurologic signs p.i. of C57BL/6 mice with Aβ1–42/CFA, but did not test for behavioral deficits. Mice develop an inflammatory disease of the CNS characterized by the presence of perivenular inflammatory foci containing macrophages, T and B cells, and Igs both in the brain and spinal cord. In our immunohistochemical analyses of Aβ1–42/CFA-immunized mice, we consistently observed disseminated macrophage infiltrates without focal accumulation of immune cells. S100A8 and CD14 were more prominently expressed in the CNS of Aβ1–42/CFA-immunized mice than in control animals. Calprotectin (S100A8) induces cytokine-like effects in the local environment and is expressed in activated macrophages, endothelial cells, and epithelial cells (41). CD14 is reported to function as a coreceptor for Aβ1–42 (42, 43). Both Ab blocking of CD14 and knockdown of the CD14 gene reduce the Aβ peptide-induced release of inflammatory cytokines and NO in microglial cells and peritoneal macrophages (44). We hypothesize that the upregulation of CD14 p.i. with Aβ1–42 might be part of a positive feedback loop in that Aβ1–42 induced components of its own receptor system both in the peripheral immune compartment and the CNS. Thus, induction and subsequent activation of the CD14 receptor would result in sustained secretion of proinflammatory cytokines in the presence of Aβ peptide.

1–42/CFA immunization strongly stimulated the production of proinflammatory cytokines in the serum and in peritoneal macrophages. These data suggested that Aβ1–42 acted in a PAMP-like manner on cells of the innate immune system. PAMPs (e.g., LPS) are recognized by pattern recognition receptors such as TLRs, triggering the expression of proinflammatory molecules (45). It has been demonstrated that Aβ1–42 has the capability to engage TLR2 to transduce intracellular signaling into microglial cells (46). Mice transgenic for a chimeric mouse/human APP and the human presenilin-1 gene that are also deficient for TLR2 exhibit increased Aβ deposition in the CNS and accelerated cognitive decline (47) due to deficient microglia activation, indicating the possibility of a direct interaction of Aβ1–42 with TLRs in the CNS. By activating TLR2, Aβ1–42 induces the secretion of proinflammatory molecules like TNF, IL-6, and IL-1β in mouse primary microglia (23). Similarly, both TLR2 and -4 mediate Aβ1–42-induced proinflammatory responses in human monocytic cell lines (48). In contrast, TLR2 and -4 are not required for the induction of EAE by active immunization with myelin Ags emulsified in CFA. In TLR2-deficient mice, the severity of MOG35–55/CFA-induced EAE is similar to wild-type animals (49). TLR4 and TLR9 KO animals are even hypersusceptible to EAE (50). Thus, each of TLR2 and TLR4 are dispensable for inducing a paralytic syndrome upon immunization with MOG35–55/CFA, suggesting that adjuvant effects of CFA are mediated by other pattern recognition receptors or a combination of these TLRs. However, the neurocognitive phenotype induced by immunization with Aβ1–42/CFA was absolutely dependent on TLR2 and TLR4. Thus, we propose that unique effects of Aβ1–42 were mediated by TLRs and were the molecular basis of the clinical neurocognitive phenotype induced by immunization with Aβ1–42. Because there is also a weak Ag-specific T cell response to Aβ1–42 promoting inflammation in tissues with relevant expression of Aβ (27), activated macrophages may subsequently be recruited to the CNS. In this study, macrophages were further activated and induced to release proinflammatory cytokines, resulting in clinically manifest psychomotor impairment.

In conclusion, Aβ1–42 peptide had the capacity to stimulate cells of the innate immune system in a TLR2/4-dependent manner. We propose that Aβ1–42 had adjuvant-like properties and by this mechanism induced a profound inflammatory response syndrome. Aβ1–42, perhaps by activating TLR2, triggered the expression of CD14, which could act as a coreceptor for Aβ peptide. Therefore, this might represent a feed-forward loop enhancing Aβ1–42-driven activation of macrophages, leading to sustained secretion of proinflammatory cytokines in situ. Thus, the current study highlights potential risks of Aβ immunotherapy and potential mechanisms involved in the induction of cognitive deficits.

Disclosures The authors have no financial conflicts of interest.

This work was supported by Deutsche Forschungsgemeinschaft Grant He 2386/4-1 and -2. T.K. is the recipient of a Heisenberg award and other grants from the Deutsche Forschungsgemeinschaft (KO-2964/2-1 and KO2964/3-1) as well as from the Gemeinnützige Hertie-Stiftung. P.V. was supported by the Studienstiftung des Deutschen Volkes.

The online version of this article contains supplemental material.

Abbreviations used in this paper:

amyloid β

AD

Alzheimer’s disease

APP

amyloid precursor protein

BL

baseline

EAE

experimental autoimmune encephalomyelitis

GFAP

glial fibrillary acidic protein

KO

knockout

MOG

myelin oligodendrocyte glycoprotein

PAMP

pathogen-associated molecular pattern

p.i.

postimmunization

PTX

pertussis toxin.

1
Schenk
D.
,
Barbour
R.
,
Dunn
W.
,
Gordon
G.
,
Grajeda
H.
,
Guido
T.
,
Hu
K.
,
Huang
J.
,
Johnson-Wood
K.
,
Khan
K.
, et al
.
1999
.
Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse.
Nature
400
:
173
177
.
2
Bard
F.
,
Cannon
C.
,
Barbour
R.
,
Burke
R. L.
,
Games
D.
,
Grajeda
H.
,
Guido
T.
,
Hu
K.
,
Huang
J.
,
Johnson-Wood
K.
, et al
.
2000
.
Peripherally administered antibodies against amyloid beta-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease.
Nat. Med.
6
:
916
919
.
3
Orgogozo
J. M.
,
Gilman
S.
,
Dartigues
J. F.
,
Laurent
B.
,
Puel
M.
,
Kirby
L. C.
,
Jouanny
P.
,
Dubois
B.
,
Eisner
L.
,
Flitman
S.
, et al
.
2003
.
Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization.
Neurology
61
:
46
54
.
4
Robinson
S. R.
,
Bishop
G. M.
,
Münch
G.
.
2003
.
Alzheimer vaccine: amyloid-beta on trial.
Bioessays
25
:
283
288
.
5
Münch
G.
,
Robinson
S. R.
.
2002
.
Potential neurotoxic inflammatory responses to Abeta vaccination in humans.
J. Neural Transm.
109
:
1081
1087
.
6
Austin
L.
,
Arendash
G. W.
,
Gordon
M. N.
,
Diamond
D. M.
,
DiCarlo
G.
,
Dickey
C.
,
Ugen
K.
,
Morgan
D.
.
2003
.
Short-term beta-amyloid vaccinations do not improve cognitive performance in cognitively impaired APP + PS1 mice.
Behav. Neurosci.
117
:
478
484
.
7
Chen
G.
,
Chen
K. S.
,
Kobayashi
D.
,
Barbour
R.
,
Motter
R.
,
Games
D.
,
Martin
S. J.
,
Morris
R. G.
.
2007
.
Active beta-amyloid immunization restores spatial learning in PDAPP mice displaying very low levels of beta-amyloid.
J. Neurosci.
27
:
2654
2662
.
8
Shie
F. S.
,
Woltjer
R. L.
.
2007
.
Manipulation of microglial activation as a therapeutic strategy in Alzheimer’s disease.
Curr. Med. Chem.
14
:
2865
2871
.
9
Rozemuller
A. J.
,
van Gool
W. A.
,
Eikelenboom
P.
.
2005
.
The neuroinflammatory response in plaques and amyloid angiopathy in Alzheimer’s disease: therapeutic implications.
Curr. Drug Targets CNS Neurol. Disord.
4
:
223
233
.
10
Bornemann
K. D.
,
Staufenbiel
M.
.
2000
.
Transgenic mouse models of Alzheimer’s disease.
Ann. N. Y. Acad. Sci.
908
:
260
266
.
11
Sondag
C. M.
,
Dhawan
G.
,
Combs
C. K.
.
2009
.
Beta amyloid oligomers and fibrils stimulate differential activation of primary microglia.
J. Neuroinflammation
6
:
1
.
12
Lotz
M.
,
Ebert
S.
,
Esselmann
H.
,
Iliev
A. I.
,
Prinz
M.
,
Wiazewicz
N.
,
Wiltfang
J.
,
Gerber
J.
,
Nau
R.
.
2005
.
Amyloid beta peptide 1-40 enhances the action of Toll-like receptor-2 and -4 agonists but antagonizes Toll-like receptor-9-induced inflammation in primary mouse microglial cell cultures.
J. Neurochem.
94
:
289
298
.
13
Mathis
C.
,
Paul
S. M.
,
Crawley
J. N.
.
1994
.
Characterization of benzodiazepine-sensitive behaviors in the A/J and C57BL/6J inbred strains of mice.
Behav. Genet.
24
:
171
180
.
14
Holmes
A.
,
Yang
R. J.
,
Crawley
J. N.
.
2002
.
Evaluation of an anxiety-related phenotype in galanin overexpressing transgenic mice.
J. Mol. Neurosci.
18
:
151
165
.
15
O’Keefe
J.
1990
.
A computational theory of the hippocampal cognitive map.
Prog. Brain Res.
83
:
301
312
.
16
Streijger
F.
,
Jost
C. R.
,
Oerlemans
F.
,
Ellenbroek
B. A.
,
Cools
A. R.
,
Wieringa
B.
,
Van der Zee
C. E.
.
2004
.
Mice lacking the UbCKmit isoform of creatine kinase reveal slower spatial learning acquisition, diminished exploration and habituation, and reduced acoustic startle reflex responses.
Mol. Cell. Biochem.
256–257
:
305
318
.
17
Dere
E.
,
Huston
J. P.
,
De Souza Silva
M. A.
.
2005
.
Episodic-like memory in mice: simultaneous assessment of object, place and temporal order memory.
Brain Res. Brain Res. Protoc.
16
:
10
19
.
18
Schwendener
R. A.
2007
.
Liposomes in biology and medicine.
Adv. Exp. Med. Biol.
620
:
117
128
.
19
Furlan
R.
,
Brambilla
E.
,
Sanvito
F.
,
Roccatagliata
L.
,
Olivieri
S.
,
Bergami
A.
,
Pluchino
S.
,
Uccelli
A.
,
Comi
G.
,
Martino
G.
.
2003
.
Vaccination with amyloid-beta peptide induces autoimmune encephalomyelitis in C57/BL6 mice.
Brain
126
:
285
291
.
20
Ousman
S. S.
,
Tomooka
B. H.
,
van Noort
J. M.
,
Wawrousek
E. F.
,
O’Connor
K. C.
,
Hafler
D. A.
,
Sobel
R. A.
,
Robinson
W. H.
,
Steinman
L.
.
2007
.
Protective and therapeutic role for alphaB-crystallin in autoimmune demyelination.
Nature
448
:
474
479
.
21
Nessler
S.
,
Boretius
S.
,
Stadelmann
C.
,
Bittner
A.
,
Merkler
D.
,
Hartung
H. P.
,
Michaelis
T.
,
Brück
W.
,
Frahm
J.
,
Sommer
N.
,
Hemmer
B.
.
2007
.
Early MRI changes in a mouse model of multiple sclerosis are predictive of severe inflammatory tissue damage.
Brain
130
:
2186
2198
.
22
Vollmar
P.
,
Nessler
S.
,
Kalluri
S. R.
,
Hartung
H. P.
,
Hemmer
B.
.
2009
.
The antidepressant venlafaxine ameliorates murine experimental autoimmune encephalomyelitis by suppression of pro-inflammatory cytokines.
Int. J. Neuropsychopharmacol.
12
:
525
536
.
23
Reed-Geaghan
E. G.
,
Savage
J. C.
,
Hise
A. G.
,
Landreth
G. E.
.
2009
.
CD14 and toll-like receptors 2 and 4 are required for fibrillar Abeta-stimulated microglial activation.
J. Neurosci.
29
:
11982
11992
.
24
Kerlero de Rosbo
N.
,
Milo
R.
,
Lees
M. B.
,
Burger
D.
,
Bernard
C. C.
,
Ben-Nun
A.
.
1993
.
Reactivity to myelin antigens in multiple sclerosis. Peripheral blood lymphocytes respond predominantly to myelin oligodendrocyte glycoprotein.
J. Clin. Invest.
92
:
2602
2608
.
25
Lehmann
P. V.
,
Forsthuber
T.
,
Miller
A.
,
Sercarz
E. E.
.
1992
.
Spreading of T-cell autoimmunity to cryptic determinants of an autoantigen.
Nature
358
:
155
157
.
26
Korn
T.
,
Reddy
J.
,
Gao
W.
,
Bettelli
E.
,
Awasthi
A.
,
Petersen
T. R.
,
Bäckström
B. T.
,
Sobel
R. A.
,
Wucherpfennig
K. W.
,
Strom
T. B.
, et al
.
2007
.
Myelin-specific regulatory T cells accumulate in the CNS but fail to control autoimmune inflammation.
Nat. Med.
13
:
423
431
.
27
Brown
D. A.
,
Sawchenko
P. E.
.
2007
.
Time course and distribution of inflammatory and neurodegenerative events suggest structural bases for the pathogenesis of experimental autoimmune encephalomyelitis.
J. Comp. Neurol.
502
:
236
260
.
28
Pollak
Y.
,
Orion
E.
,
Goshen
I.
,
Ovadia
H.
,
Yirmiya
R.
.
2002
.
Experimental autoimmune encephalomyelitis-associated behavioral syndrome as a model of ‘depression due to multiple sclerosis’.
Brain Behav. Immun.
16
:
533
543
.
29
Chen
J.
,
Buchanan
J. B.
,
Sparkman
N. L.
,
Godbout
J. P.
,
Freund
G. G.
,
Johnson
R. W.
.
2008
.
Neuroinflammation and disruption in working memory in aged mice after acute stimulation of the peripheral innate immune system.
Brain Behav. Immun.
22
:
301
311
.
30
Richwine
A. F.
,
Sparkman
N. L.
,
Dilger
R. N.
,
Buchanan
J. B.
,
Johnson
R. W.
.
2009
.
Cognitive deficits in interleukin-10-deficient mice after peripheral injection of lipopolysaccharide.
Brain Behav. Immun.
23
:
794
802
.
31
Layé
S.
,
Parnet
P.
,
Goujon
E.
,
Dantzer
R.
.
1994
.
Peripheral administration of lipopolysaccharide induces the expression of cytokine transcripts in the brain and pituitary of mice.
Brain Res. Mol. Brain Res.
27
:
157
162
.
32
Gatti
S.
,
Bartfai
T.
.
1993
.
Induction of tumor necrosis factor-alpha mRNA in the brain after peripheral endotoxin treatment: comparison with interleukin-1 family and interleukin-6.
Brain Res.
624
:
291
294
.
33
Zhang
H.
,
Ching
S.
,
Chen
Q.
,
Li
Q.
,
An
Y.
,
Quan
N.
.
2008
.
Localized inflammation in peripheral tissue signals the CNS for sickness response in the absence of interleukin-1 and cyclooxygenase-2 in the blood and brain.
Neuroscience
157
:
895
907
.
34
Sellner
J.
,
Grandgirard
D.
,
Gianinazzi
C.
,
Landmann
R. M.
,
Leib
S. L.
.
2009
.
Effects of Toll-like receptor 2 agonist Pam(3)CysSK(4) on inflammation and brain damage in experimental pneumococcal meningitis.
J. Neuroimmunol.
206
:
28
31
.
35
Dantzer
R.
,
O’Connor
J. C.
,
Freund
G. G.
,
Johnson
R. W.
,
Kelley
K. W.
.
2008
.
From inflammation to sickness and depression: when the immune system subjugates the brain.
Nat. Rev. Neurosci.
9
:
46
56
.
36
Lestage
J.
,
Verrier
D.
,
Palin
K.
,
Dantzer
R.
.
2002
.
The enzyme indoleamine 2,3-dioxygenase is induced in the mouse brain in response to peripheral administration of lipopolysaccharide and superantigen.
Brain Behav. Immun.
16
:
596
601
.
37
van Dam
A. M.
,
Poole
S.
,
Schultzberg
M.
,
Zavala
F.
,
Tilders
F. J.
.
1998
.
Effects of peripheral administration of LPS on the expression of immunoreactive interleukin-1 alpha, beta, and receptor antagonist in rat brain.
Ann. N. Y. Acad. Sci.
840
:
128
138
.
38
Kipnis
J.
,
Cohen
H.
,
Cardon
M.
,
Ziv
Y.
,
Schwartz
M.
.
2004
.
T cell deficiency leads to cognitive dysfunction: implications for therapeutic vaccination for schizophrenia and other psychiatric conditions.
Proc. Natl. Acad. Sci. USA
101
:
8180
8185
.
39
Brynskikh
A.
,
Warren
T.
,
Zhu
J.
,
Kipnis
J.
.
2008
.
Adaptive immunity affects learning behavior in mice.
Brain Behav. Immun.
22
:
861
869
.
40
Derecki
N. C.
,
Cardani
A. N.
,
Yang
C. H.
,
Quinnies
K. M.
,
Crihfield
A.
,
Lynch
K. R.
,
Kipnis
J.
.
2010
.
Regulation of learning and memory by meningeal immunity: a key role for IL-4.
J. Exp. Med.
207
:
1067
1080
.
41
Rahimi
F.
,
Hsu
K.
,
Endoh
Y.
,
Geczy
C. L.
.
2005
.
FGF-2, IL-1beta and TGF-beta regulate fibroblast expression of S100A8.
FEBS J.
272
:
2811
2827
.
42
Liu
Y.
,
Walter
S.
,
Stagi
M.
,
Cherny
D.
,
Letiembre
M.
,
Schulz-Schaeffer
W.
,
Heine
H.
,
Penke
B.
,
Neumann
H.
,
Fassbender
K.
.
2005
.
LPS receptor (CD14): a receptor for phagocytosis of Alzheimer’s amyloid peptide.
Brain
128
:
1778
1789
.
43
Triantafilou
M.
,
Triantafilou
K.
.
2002
.
Lipopolysaccharide recognition: CD14, TLRs and the LPS-activation cluster.
Trends Immunol.
23
:
301
304
.
44
Fassbender
K.
,
Walter
S.
,
Kühl
S.
,
Landmann
R.
,
Ishii
K.
,
Bertsch
T.
,
Stalder
A. K.
,
Muehlhauser
F.
,
Liu
Y.
,
Ulmer
A. J.
, et al
.
2004
.
The LPS receptor (CD14) links innate immunity with Alzheimer’s disease.
FASEB J.
18
:
203
205
.
45
Mogensen
T. H.
2009
.
Pathogen recognition and inflammatory signaling in innate immune defenses.
Clin. Microbiol. Rev.
22
:
240
273
.
46
Jana
M.
,
Palencia
C. A.
,
Pahan
K.
.
2008
.
Fibrillar amyloid-beta peptides activate microglia via TLR2: implications for Alzheimer’s disease.
J. Immunol.
181
:
7254
7262
.
47
Richard
K. L.
,
Filali
M.
,
Préfontaine
P.
,
Rivest
S.
.
2008
.
Toll-like receptor 2 acts as a natural innate immune receptor to clear amyloid beta 1-42 and delay the cognitive decline in a mouse model of Alzheimer’s disease.
J. Neurosci.
28
:
5784
5793
.
48
Udan
M. L.
,
Ajit
D.
,
Crouse
N. R.
,
Nichols
M. R.
.
2008
.
Toll-like receptors 2 and 4 mediate Abeta(1-42) activation of the innate immune response in a human monocytic cell line.
J. Neurochem.
104
:
524
533
.
49
Prinz
M.
,
Garbe
F.
,
Schmidt
H.
,
Mildner
A.
,
Gutcher
I.
,
Wolter
K.
,
Piesche
M.
,
Schroers
R.
,
Weiss
E.
,
Kirschning
C. J.
, et al
.
2006
.
Innate immunity mediated by TLR9 modulates pathogenicity in an animal model of multiple sclerosis.
J. Clin. Invest.
116
:
456
464
.
50
Marta
M.
,
Andersson
A.
,
Isaksson
M.
,
Kämpe
O.
,
Lobell
A.
.
2008
.
Unexpected regulatory roles of TLR4 and TLR9 in experimental autoimmune encephalomyelitis.
Eur. J. Immunol.
38
:
565
575
.