Skip to main content
Journal of Virology logoLink to Journal of Virology
. 2013 Dec;87(24):13433–13445. doi: 10.1128/JVI.02072-13

Absence of CD14 Delays Progression of Prion Diseases Accompanied by Increased Microglial Activation

Keiko Sakai 1, Rie Hasebe 1, Yusuke Takahashi 1, Chang-Hyun Song 1,*, Akio Suzuki 1, Takeshi Yamasaki 1, Motohiro Horiuchi 1,
PMCID: PMC3838238  PMID: 24089559

Abstract

Prion diseases are fatal neurodegenerative disorders characterized by accumulation of PrPSc, vacuolation of neurons and neuropil, astrocytosis, and microglial activation. Upregulation of gene expressions of innate immunity-related factors, including complement factors and CD14, is observed in the brains of mice infected with prions even in the early stage of infections. When CD14 knockout (CD14−/−) mice were infected intracerebrally with the Chandler and Obihiro prion strains, the mice survived longer than wild-type (WT) mice, suggesting that CD14 influences the progression of the prion disease. Immunofluorescence staining that can distinguish normal prion protein from the disease-specific form of prion protein (PrPSc) revealed that deposition of PrPSc was delayed in CD14−/− mice compared with WT mice by the middle stage of the infection. Immunohistochemical staining with Iba1, a marker for activated microglia, showed an increased microglial activation in prion-infected CD14−/− mice compared to WT mice. Interestingly, accompanied by the increased microglial activation, anti-inflammatory cytokines interleukin-10 (IL-10) and transforming growth factor β (TGF-β) appeared to be expressed earlier in prion-infected CD14−/− mice. In contrast, IL-1β expression appeared to be reduced in the CD14−/− mice in the early stage of infection. Double immunofluorescence staining demonstrated that CD11b- and Iba1-positive microglia mainly produced the anti-inflammatory cytokines, suggesting anti-inflammatory status of microglia in the CD14−/− mice in the early stage of infection. These results imply that CD14 plays a role in the disease progression by suppressing anti-inflammatory responses in the brain in the early stage of infection.

INTRODUCTION

Prion diseases are fatal neurodegenerative disorders including scrapie in sheep and goats, bovine spongiform encephalopathy in cattle, chronic wasting disease in cervids, and Creutzfeldt-Jakob disease in humans. These diseases are characterized by the deposition of disease-specific prion protein (PrPSc), vacuolation of neurons and neuropil, and astrocytosis in the central nervous system (CNS). Despite little recruitment of adaptive immune cells into the CNS in prion diseases, microglial activation has been observed close to depositions of PrPSc before neuronal degeneration occurs (1, 2).

Microglia, the resident macrophages of the CNS, survey the environment under physiological conditions. Microbial infections, injury, and neurodegenerative conditions induce microglial activation, characterized by changes in shape, gene expression, and function including cytokine production, phagocytic activity, and antigen presentation (3, 4). Activated microglia are neurotoxic in that they produce proinflammatory mediators such as interleukin-1β (IL-1β), IL-6, tumor necrosis factor alpha (TNF-α), nitric oxide, and reactive oxygen species (5). Alternatively, activated microglia are neuroprotective in that they block proinflammatory responses and produce anti-inflammatory cytokines IL-10, transforming growth factor β (TGF-β), and neurotrophic factors (6).

Previous studies have suggested the significance of a proinflammatory environment in the CNS in the progression of prion diseases. Schultz et al. demonstrated that IL-1β receptor I-deficient mice delayed the onset of prion diseases with attenuated PrPSc deposition and astrocytosis (7). A protective role of anti-inflammatory cytokines in prion diseases has also been reported: the incubation time in IL-10-deficient mice was greatly shortened (8). Boche et al. showed that inhibition of TGF-β activity in ME7 prion-infected mice induced severe neuronal inflammation and acute neuronal death (9). Taken together, these facts suggest the necessity for the inflammatory environment in the brains of prion-infected mice to be further elucidated for a better understanding of the mechanism of neurodegeneration in prion diseases.

It is well established that PrPSc accumulation precedes neurodegeneration and clinical manifestations of prion diseases. To analyze the host reaction in prion infection in the brain, particularly during the early stages after intracerebral inoculation of prions, we compared the gene expression of prion- and mock-infected mouse brains by cDNA microarray analysis from 60 to 90 days postinoculation (dpi) (M. Horiuchi and C.-H. Song, unpublished observations). Focusing on genes that have been reported to be expressed by microglia, we found that several innate-immunity-related genes, including CD14, and complement factors were upregulated as described in previous studies (10, 11). CD14, a glycosylphosphatidylinositol (GPI)-anchored protein, is well known as a lipopolysaccharide (LPS) receptor. Although CD14 itself cannot induce cellular signaling because of lack of cytoplasmic domains, binding of pathogen-associated molecular patterns to CD14 clusters transmembrane proteins including Toll-like receptor 2 (TLR2) and TLR4 to induce gene expression of proinflammatory mediators (12, 13). Upregulation of CD14 expression associated with deposits of aggregated proteins has been reported in the brains of mouse models and human patients with neurodegenerative disorders including Alzheimer's disease (AD), Parkinson's disease/dementia with Lewy bodies, and amyotrophic lateral sclerosis (14). A polymorphism in the promoter region of the CD14 gene that affects the expression of CD14 has been reported to significantly increase the risk of Parkinson's disease in females (15, 16). Depletion of CD14 in an AD mouse model showed a reduced amyloid β (Aβ) plaque burden with altered microglial activation (17). These findings suggest the involvement of CD14 in the pathogenesis of neurodegenerative disorders.

In the present study, we examined if CD14 influences the neuropathology and neuroinflammatory conditions in prion diseases. When CD14 knockout (CD14−/−) mice were inoculated intracerebrally with Chandler and Obihiro prion strains, the survival time was significantly prolonged compared to that of wild-type (WT) mice, suggesting that CD14 is involved in the progression of the disease. Our data also suggest that an increased microglial activation in CD14−/− mice accompanied by anti-inflammatory cytokine production may contribute to the prolonged survival time of the CD14−/− mice.

MATERIALS AND METHODS

Antibodies.

Anti-mouse prion protein (PrP) monoclonal antibodies (MAbs) 31C6 and 132 were prepared as described previously (18). Anti-Iba1 rabbit polyclonal antibodies were purchased from Wako (product no. 019-20001). Anti-glial fibrillary acidic protein (anti-GFAP) rabbit polyclonal antibodies were from Dako (product no. Z033401). Unconjugated and Alexa Fluor 488-conjugated anti-mouse CD11b rat MAb (clone M1/70), anti-mouse CD14 rat MAb (clone Sal 14-2), and anti-mouse F4/80 rat MAb (clone BM8) were from BioLegend. Anti-NeuN mouse MAb (clone A60) was from Millipore. Anti-mouse CD45 rat MAb (clone 13/2.3) was from Funakoshi. Anti-GFAP mouse MAb (clone GF5), anti-IL-10 rat MAb (clone JES5-2A5), anti-mouse CD68 rat MAb (clone FA-11), anti-TGF-β rabbit polyclonal antibodies (product no. 66043), and anti-IL-1β rabbit polyclonal antibodies (product no. 9722) were from Abcam. ECL horseradish peroxidase-labeled anti-mouse IgG was from Amersham Biosciences (product no. NA9310V). All Alexa Fluor-labeled antibodies were from Life Technologies.

Mice and prion inoculation.

C57BL/6J mice were purchased from Japan Clea Inc. CD14 knockout (CD14−/−) mice of congenic strain B6.129S-Cd14<tm1Frm>/J were purchased from Jackson Laboratories and were further maintained by inbreeding. All animal procedures were approved by the Institutional Animal Care and Use Committee of the Graduate School of Veterinary Medicine, Hokkaido University. Six-week-old female WT and CD14−/− mice were inoculated intracerebrally with 20 μl of 2.5% brain homogenates of the Chandler- or Obihiro-infected C57BL/6J mice. The inoculation was carried out whenever the 6-week-old female CD14−/− mice were available. The exact same aliquots of 2.5% brain homogenates were used for each inoculation. At the clinical stage, mice were observed every day, and the clinical endpoint of disease was defined as recumbency with severe emaciation within two consecutive days.

Quantitative RT-PCR.

Total RNA was extracted from the thalamus cut at a thickness of the coronal section with 5 mm around the level of bregma −1.82 mm (as indicated in The Mouse Brain in Stereotaxic Coordinates, 2nd ed. [19]) using TRIzol reagent (Life Technologies). First-strand cDNA was synthesized from 2 μg of the total RNA using a First-Strand cDNA synthesis kit (Amersham Biosciences) according to the manufacturer's instructions. The amplification reaction mixtures contained template cDNA, 1× predesigned TaqMan Gene Expression Assays for mouse Cd14 (Mm00438094_g1) and for mouse ACTB (catalog no. 4352933E), and 1× TaqMan Fast Universal PCR Master Mix. TaqMan assays were carried out using an ABI 7900HT Fast Real-Time PCR system (Applied Biosystems). The amplification profiles were analyzed using a threshold cycle relative quantification method and were normalized with the expression of mouse ACTB gene as described previously (20).

Bioassay.

Brain homogenates from 2 mice at each time point were prepared in sterile phosphate-buffered saline (PBS) to 1% (wt/vol). Twenty microliters of brain homogenates were inoculated intracerebrally into 6-week-old Tga20 mice. To obtain an infectivity-incubation time standard curve, 10-fold serial diluted brain homogenates used as the inocula of the Chandler and Obihiro strains were also injected into Tga 20 mice. The 50% lethal doses (LD50) of the original 10% homogenates were estimated to be 109.3 and 109.5 LD50/g brain tissue for the Chandler and Obihiro strains, respectively. In the Chandler infection, the standard curve for the incubation periods (χ) was fitted by the approximations of concentrations of original brain homogenates (y), determined as follows: y = e23.026−0.425χ for the periods of up to 68 days, and y = e16.118−0.332χ for the periods after 69 days. In the Obihiro infection, the approximation was y = e20.723−0.263χ for the periods of up to107 days and y = e7.092−0.129χ for the periods after 108 days.

PrP-res detection by immunoblotting.

For protease-resistant prion protein (PrP-res) detection, at 60, 90, and 120 dpi and at the terminal stage of the disease, brains were harvested and were homogenized in sterile PBS to prepare 10% (wt/vol) brain homogenates. The homogenates were treated with proteinase K (PK) and were subjected to SDS-PAGE and immunoblotting as described previously (20).

PrPSc-specific immunofluorescence staining.

The PrPSc-specific staining of frozen sections was performed by a method described previously (21) with some modifications. Briefly, the brains were embedded in OCT compound (Sakura Finetek, Japan) and were cut at a thickness of 10 μm. The samples were fixed with 4% paraformaldehyde (PFA) for 10 min at room temperature (rt). The slides were treated with 0.1% Triton X-100 for 10 min at rt and then with 5 M guanidine thiocyanate for 10 min at rt. After washing with PBS, the sections were blocked with 5% fetal bovine serum (FBS) in PBS at 37°C for 30 min. The sections were then incubated with MAb132 at 3 μg/ml at 37°C for 45 min. For the secondary antibody reaction, the sections were incubated with Alexa Fluor 488-labeled anti-mouse IgG (Fab)′2 fragment of goat IgG at rt for 1 h. The samples were counterstained with 4′6-diamidino-2-phenylindole (DAPI), mounted with ProLong Gold antifade reagent (Invitrogen), and then observed with a laser scanning confocal microscope (LSM) (Carl Zeiss International).

Histopathological analysis.

The histopathological analysis was carried out as described previously (22). Briefly, brains were harvested at 90 and 120 dpi and at the terminal stage of the disease and fixed with 10% phosphate-buffered formalin. The brains were embedded in paraffin and cut at the level of bregma −1.82 mm. The samples were stained with hematoxylin and eosin (H&E).

Immunohistochemistry.

For the antigen retrieval of Iba1 detection in paraffin-embedded tissue, the slides were treated with microwaves at 600 W for 10 min in 0.01 M citrate buffer solution, pH 6.0. After washing with PBS, the slides were blocked with normal goat serum (Nichirei) and incubated overnight with primary antibody diluted at 1:1,000 in normal goat serum at 4°C. The slides were treated with 0.3% hydrogen peroxide in methanol to block endogenous peroxidase activity at rt for 10 min. Biotin-labeled anti-rabbit IgG goat polyclonal antibodies (Nichirei) were incubated for 10 min at rt. Peroxidase-labeled streptavidin (Nichirei) was reacted for 5 min at rt. The immunoreactivity was visualized with Impact DAB (Vector). For quantitative analysis of Iba1-positive microglia, cells with more than 10 μm2 of Iba1 immunoreactivity were counted by Image J (U.S. National Institutes of Health, Bethesda, MD, USA; http://rsb.info.nih.gov/ij/).

For the immunohistochemistry of the microglial markers and cytokines in frozen tissue, brains were embedded in the OCT compound (Sakura Finetek, Japan) and were cut at a thickness of 10 μm. Sections were fixed with 4% PFA for 10 min at rt. For the antigen retrieval, samples were treated with 0.1% Tween 20 in PBS for 15 min at rt. The slides were blocked with 2% bovine serum albumin for 15 min at rt and incubated overnight with primary antibody at 4°C. Then, slides were incubated with secondary antibodies and DAPI for 1 h at rt. Finally, slides were mounted with ProLong Gold antifade reagent (Invitrogen) and observed with LSM700 (Zeiss). For the detection of CD14, CD45, and CD68, the antibodies were diluted at 1:800. For CD11b, TGF-β, and IL-1β, the antibodies were diluted at 1:200. For IL-10, the antibody was diluted at 1:100. For GFAP and F4/80, the antibodies were diluted at 1:1,000. For secondary antibodies, all Alexa Fluor-labeled antibodies were diluted at 1:1,000.

RESULTS

Expression of CD14 is increased in microglia and accompanied by progression of prion diseases.

Previous DNA microarray analyses have suggested that gene expression of CD14 was upregulated in prion-infected mouse brains in the early stage of infection (10, 11) (M. H. and C.-H. Song, unpublished). To confirm the gene expression of CD14, we performed a quantitative RT-PCR (Fig. 1A). The CD14 gene expression in the thalamus of the Chandler-infected mice was 2.5 ± 0.8, 3.6 ± 0.8, and 3.6 ± 0.6 higher than that in the thalamus of mock-infected mice, at 75, 90, and 120 dpi, respectively, confirming the upregulation of CD14 expression. We also performed immunofluorescence staining of frozen sections of the Chandler- and Obihiro-infected mice brains to confirm the protein expression of CD14 (Fig. 1B), and CD14-positive cells were most frequently detected in the cerebral peduncle at the observed time points (Fig. 1B). The CD14-positive cells were scattered in corpus callosum, internal capsules, and cerebral peduncles at 60 dpi, spreading more widely in the cerebral peduncles and periventricle areas at 90 dpi, and the spreading had expanded to the thalamus at 120 dpi (Fig. 1B and C). A few CD14-positive cells were also detected in the internal capsules and cerebral peduncles of mock-infected mouse brains, suggesting that these cells may be residents in these areas (Fig. 1C). To confirm the CD14 expression in the microglia, double staining of CD14 with CD11b for microglia and of GFAP for astrocytes was performed (Fig. 1D). Most of the CD14 immunoreactivity was detected in the CD11b-positive microglia, but it was not detected in the GFAP-positive astrocytes.

Fig 1.

Fig 1

Expression of CD14 in the brains of prion-infected WT mice. (A) Gene expression of CD14. Total RNA was extracted from the thalamus of Chandler- and mock-infected WT mice at 75, 90, and 120 dpi and subjected to quantitative RT-PCR. Expression of the CD14 gene in the thalamus of the mock-infected mice at 75 dpi was defined as 1, and relative expressions are shown. *, P < 0.05; **, P < 0.01 (Student's t test). (B) Immunofluorescence staining of CD14 in the cerebral peduncle. The brains of Chandler-, Obihiro-, and mock-infected mice were harvested at 60, 90, and 120 dpi. Frozen sections at the level of bregma −1.82 mm were subjected to immunofluorescence staining of CD14 (red). Blue, nuclear counterstaining with DAPI. Bars, 50 μm. (C) Summary of the distribution of CD14 in the brains of Chandler-, Obihiro-, and mock-infected mice. Illustrations show the level of bregma −1.82 mm as indicated in The Mouse Brain in Stereotaxic Coordinates, 2nd ed. (19). (D) Double immunofluorescence staining of CD14 (green) with CD11b (left, red) or GFAP (right, red). Representative figures from the cerebral peduncles of the Chandler-infected mice at 120 dpi are shown. Blue, nuclei. Bars, 20 μm.

CD14 influences the progression of prion diseases.

To examine the effect of the lack of CD14 on the neuropathogenesis of prion diseases, we inoculated brain homogenates of the Chandler or Obihiro strain-infected mice into CD14−/− mice (Fig. 2). The CD14−/− mice infected either with the Chandler strain or with the Obihiro strain survived longer than the WT mice. The mean survival time of the Chandler strain-infected CD14−/− mice (161.7 ± 3.7 days, n = 11) was significantly longer than that of the WT mice (153.8 ± 3.7 days, n = 12, P < 0.01, Student's t test) (Fig. 2A). The mean survival time of the Obihiro strain-infected CD14−/− mice (172.3 ± 4.8 days, n = 10) was also significantly longer than that of WT mice (157.9 ± 7.8 days, n = 11, P < 0.01, Student's t test) (Fig. 2B). These results suggest that the lack of CD14 decelerates the progression of the disease.

Fig 2.

Fig 2

Survival times of WT and CD14−/− mice infected with the Chandler or Obihiro strain. (A) Survival curves of the Chandler-infected WT and CD14−/− mice. (B) Survival curves of the Obihiro-infected WT and CD14−/− mice.

To determine if knockout of CD14 influences the accumulation of PrPSc, we analyzed the PrP-res accumulation in the brains by immunoblotting (Fig. 3A and B). The intensity of the PrPSc signals in the brains of CD14−/− mice infected with either the Chandler or the Obihiro strain was slightly reduced at 90 and 120 dpi and became comparable to that seen in the WT mice at the terminal stage.

Fig 3.

Fig 3

PrPSc accumulation in the brains of prion-infected WT and CD14−/− mice. (A) Detection of PrP-res by immunoblotting. Brains were harvested at 90 and 120 dpi and at the terminal stage. Brain tissue equivalents loaded were 500, 350, and 250 μg for the 90 and 120 dpi time points and the terminal stage of the disease, respectively. Two mice were used in each group. M, molecular mass marker. (B) Quantitative analysis of PrP-res. The results of immunoblotting were quantified using Multi Gauge ver 3.0 (Fuji Film). Each blot used 10 ng of rMoPrP for the normalization of the transfer efficiency, and the graphs show relative values (arbitrary units [AU]) to rMoPrP (average of the results from 2 mice). term, terminal stage of the disease. (C) Immunofluorescence staining of PrPSc. Frozen sections were prepared from the brains of WT and CD14−/− mice harvested at 60, 90, and 120 dpi and subjected to PrPSc-specific staining with MAb 132. Representative images from the thalamus are shown (representing the regions of the boxed areas in the corresponding images in panel D). Bars, 50 μm. Green, PrPSc. Blue, nuclei. (D) Summary of PrPSc distribution detected by PrPSc-specific staining.

We have reported that MAb132, recognizing amino acids 119 to 127 of mouse PrP, in combination with pretreatment of cells with 5 M guanidine thiocyanate, is useful for the PrPSc-specific immunofluorescence staining of prion-infected cell cultures (21). It is well established that PrPSc includes protease-sensitive and protease-resistant PrPSc (23, 24). The protease-sensitive PrPSc that cannot be detected by immunoblotting using proteinase K-treated samples could be detected by this PrPSc-specific immunofluorescence staining because of the omission of the protease treatment process. In the present study, we applied this method to frozen brain sections for the detection of PrPSc (Fig. 3C and D). Signals of PrP could be detected from the brains of prion-infected mice, but signals from the brains of uninfected mice remained at background levels under the same conditions (data not shown), demonstrating that this method can be applied to detect PrPSc from frozen brain sections. At 60 dpi, bright punctate staining of PrPSc was detected frequently in the thalamus and occasionally detected in the cerebral cortex of the WT mice infected with the Chandler strain. The PrPSc was also detected in parts of the thalamus of CD14−/− mice, but the occurrence was less frequent than in the same areas of WT mice (Fig. 3C and D). At 90 dpi, PrPSc was distributed more widely; PrPSc could be detected in the hippocampus as well as in the thalamus and cerebral cortex of WT and CD14−/− mice infected with the Chandler strain (Fig. 3D). However, the PrPSc staining in some brain areas of CD14−/− mice seemed to be still weaker than in the WT mice (Fig. 3C and D). There were no marked differences in the PrPSc distribution between the WT and CD14−/− mice at 120 dpi. Delay of the PrPSc accumulation was also observed in the brains of the Obihiro-infected CD14−/− mice. At 60 dpi, PrPSc was detected in the hippocampus and thalamus of WT mice but not in these areas of the CD14−/− mice. At 90 dpi, PrPSc was spread into the hypothalamus and amygdala of WT mice, but the presence of PrPSc in the CD14−/− mice was restricted to the hippocampus and thalamus.

We also analyzed the prion infectivity in the brains of WT and CD14−/− mice using Tga20 mice that overexpress mouse PrP and thus are highly susceptible to mouse-adapted prions (25). Prion infectivity in the brains of both of the Chandler-infected CD14−/− mice at 60 dpi was significantly lower than that in both of the WT mice (Fig. 4). Prion infectivity in the brains of the Obihiro-infected mice also appeared to be lower than those from WT mice; the infectivity in the brain of one CD14−/− mouse (no. 2) was significantly lower than that of one WT mouse (no.1). However, in the later stages, there were no significant differences in the prion infectivity of WT and CD14−/− mice in the Chandler or Obihiro infection (Fig. 4).

Fig 4.

Fig 4

Prion infectivity. (A, B) Prion infectivity in the brains of WT and CD14−/− mice infected by the Chandler (A) or Obihiro (B) strain. The infectivity was measured by bioassay using Tga 20 mice. *, P < 0.05; **, P < 0.001 (analysis of variance [ANOVA] followed by Dunnett's post hoc test). (C, D) Survival times of Tga 20 mice used for bioassay. Survival time for each Tga 20 mouse inoculated with brain homogenates from the Chandler-infected (C) or Obihiro-infected (D) WT or CD14−/− mice is shown. The difference in the Chandler-infected CD14−/− mice at 90 dpi was due to the small standard deviation of mouse no. 1: three of four Tga20 mice in this group reached the terminal stage on the same day.

We also performed a histopathological analysis of the brains of WT and CD14−/− mice infected with the Chandler strain (Fig. 5). At 90 dpi, slight vacuolar degeneration of neuropil and neurons was only occasionally observed in the thalamus of Chandler-infected WT and CD14−/− mice. Vacuolar degeneration was widely observed throughout the brains after 120 dpi; however, there was no apparent difference in the severity and distribution of vacuolar degeneration of the WT and CD14−/− mice infected with the Chandler strain.

Fig 5.

Fig 5

Histopathological analysis of the brains of prion-infected WT and CD14−/− mice. Representative images from the thalamus, the hippocampus, and the cerebral peduncle of the Chandler-infected mice are shown. Bar, 50 μm.

Microglial activation.

Depletion of CD14 in a mouse model of Alzheimer's disease (AD) was reported to result in a reduction in the number of microglia (17). To assess if depletion of CD14 altered microglial activation in prion diseases, we analyzed the expression of an activated microglial marker, Iba1, by immunohistochemistry (Fig. 6). At 60 dpi, some microglia of CD14−/− mice had more protrusions than those of WT mice. Morphological differences in Iba1-positive microglia became more prominent at 90 dpi; microglia in CD14−/− mice had a larger cytoplasm and more branched protrusions than those of WT mice. At 120 dpi, Iba1-positive microglia in WT mice had a smaller cytoplasm and fewer protrusions; however, microglia in CD14−/− mice still had relatively larger cytoplasm with many protrusions. In the terminal stage, there were no obvious differences in the morphology of microglia of WT and CD14−/− mice.

Fig 6.

Fig 6

Expression of an activated microglial marker, Iba1, in the brains of prion-infected WT and CD14−/− mice. (A) Immunohistochemistry for Iba1. Representative images from the thalamus of the Chandler-infected mice are shown. Higher magnifications of the areas indicated by boxes are shown in the corresponding right panels. Bars, 20 μm. Term, terminal stage. (B) Quantitative analysis of Iba1-positive microglia. Numbers of Iba1-positive cells in the hippocampus and the thalamus (1.5 × 10−1 mm2 tissue section) were determined by Image J. Data are for 2 mice from each group. T, terminal stage; Hp, hippocampus; Th, thalamus.

The quantitative analysis of Iba1-positive microglia showed that in the hippocampus and thalamus of the Chandler-infected WT and CD14−/− mice the numbers of microglia increased from 60 to 120 dpi and then showed no further change or slightly decreased (Fig. 6B). Similar changes were observed in the Obihiro-infected WT and CD14−/− mice. In the Chandler-infected mice, there was little difference in the numbers of Iba1-positive microglia between the WT and CD14−/− mice by 90 dpi. At 120 dpi, Iba1-positive microglia increased more in the thalamus of CD14−/− mice than in WT mice. In the terminal stage, numbers of the Iba1-positive microglia decreased both in WT and CD14−/− mice, although those in the thalamus of CD14−/− mice were still slightly larger than those of WT mice. In the Obihiro-infected mice, there were no differences in the numbers of Iba1-positive microglia of the WT and CD14−/− mice at 60 dpi. At 90 and 120 dpi, numbers of Iba1-positive microglia in the thalamus of CD14−/− mice appeared to be larger than in the thalamus of WT mice. In the terminal stage, there were no marked differences in the numbers of Iba1-positive microglia of WT and CD14−/− mice.

To further characterize the differences in microglial activation of prion-infected WT and CD14−/− mice, we performed immunofluorescence staining for other microglial markers (Fig. 7). Expression of CD11b, a commonly used microglial marker, was elevated in the brains of WT and CD14−/− mice from 90 to 120 dpi, and the expression of CD11b in CD14−/− mice was more intense than that in WT mice at each time point. A marker for macrophages and monocytes, F4/80, is also frequently used as a microglial marker. Similar to CD11b, F4/80 immunoreactivity was detected more in CD14−/− mice than in WT mice at 90 and 120 dpi. Similar changes were also observed in the immunofluorescence staining for CD68, a phagocytic marker of macrophages and microglia: CD68-positive cells increased time dependently both in WT and in CD14−/− mice, and CD68 immunoreactivity was more prominent in CD14−/− mice than in WT mice. We also analyzed CD45, a common leukocyte antigen expressed on all leukocytes. Ramified parenchymal microglia express lower levels of CD45 than peripheral macrophages (26, 27), while the expression of CD45 in microglia may be increased under certain pathological conditions such as HIV encephalitis (28, 29) and Alzheimer's disease (30). For the study here, expression of CD45 was clearly more upregulated in CD14−/− mice than in WT mice at 90 and 120 dpi (Fig. 7).

Fig 7.

Fig 7

Immunofluorescence staining for microglial markers in the brains of prion-infected WT and CD14−/− mice. Frozen blocks of the brains harvested at 90 and 120 dpi were subjected to immunofluorescence staining for microglial markers CD11b, F4/80, CD68, and CD45. Representative images from the thalamus of the Chandler-infected mice are shown. Bars, 50 μm.

Expression of anti-inflammatory cytokines.

Depletion of CD14 in the AD model mouse resulted in a reduced amyloid β (Aβ) plaque burden with the increased gene expression of the anti-inflammatory cytokine IL-10 (17). Similar to the reduced Aβ plaque burden in AD model mice lacking CD14−/−, this study observed that accumulation of PrPSc in prion-infected CD14−/− mice was delayed by 90 dpi (Fig. 3C and D). The knockout of the IL-10 gene has been shown to greatly shorten the survival time of prion-infected mice, suggesting a protective role of IL-10 in the progression of prion diseases (8). Therefore, we analyzed the anti-inflammatory cytokine expression in prion-infected CD14−/− mice to determine if a lack of CD14 modulates the inflammatory response in the brain.

Compared to the mock-infected mice, the prion-infected WT mice showed a weak but detectable IL-10 expression from 60 to 120 dpi. Interestingly, IL-10 immunoreactivity was observed more frequently in the thalamus of CD14−/− mice than in WT mice throughout the time of observation (Fig. 8A), and IL-10 immunoreactivity in CD14−/− mice appeared to increase from 60 to 90 dpi but to decrease from 90 to 120 dpi. The IL-10-positive areas were quantified and statistically analyzed if more than 3 mice were available. At 60 dpi, the areas positive for IL-10 in CD14−/− mice were significantly larger than in WT mice (P < 0.01, Student's t test) (Fig. 8B). The IL-10-positive areas in CD14−/− mice were also larger than those in WT mice at 90 and 120 dpi. We also examined the cell type in the brains expressing IL-10 by double staining (Fig. 8C to F). Most of the IL-10 immunoreactivity was detected in CD11b- or Iba1-positive microglia (Fig. 8C and D) and some in NeuN-positive neurons (Fig. 8E), in the brains of both WT and CD14−/− mice from 60 to 120 dpi. There was no IL-10 immunoreactivity detected in GFAP-positive astrocytes (Fig. 8F).

Fig 8.

Fig 8

Expression of IL-10 in the brains of WT and CD14−/− mice infected with the Chandler and Obihiro strains. (A) Immunofluorescence staining of IL-10. Frozen blocks of the brains harvested at 60, 90, and 120 dpi were subjected to immunofluorescence staining. Representative images from the thalamus at each time point are shown. Bars, 50 μm. Red, IL-10; blue, nuclei. (B) Quantitative analysis of IL-10-positive areas in the thalamus of Chandler-infected mice. The areas positive for IL-10 (μm2/0.1 mm2) were quantified using Imaris ver 7.6.1 (Bitplane). The numbers of mice used for the analysis were 3 for both WT and CD14−/− mice at 60 dpi, 2 for both WT and CD 14−/− mice at 90 dpi, 2 for WT mice at 120 dpi, and 1 CD14−/− mouse at 120 dpi. *, P < 0.01, Student's t test. (C to F) Double immunofluorescence staining of IL-10 with CD11b (C), Iba1 (D), NeuN (E), and GFAP (F). Representative images from the thalamus of the Chandler-infected mice are shown. Green, immunoreactivities for these markers; red, IL-10; blue, nuclei. Arrows show double-positive cells. Bars, 10 μm.

We also analyzed another anti-inflammatory cytokine, TGF-β (Fig. 9A), which has been reported to play a role in the suppression of progression of prion diseases (9). At 60 dpi, TGF-β immunoreactivity was observed more frequently in CD14−/− mice than in WT mice infected with the Chandler or Obihiro strains. Although no significant difference was observed in TGF-β-positive areas between WT and CD14−/− mice at 60 dpi (P = 0.053, Student's t test), the areas in two of three samples from the CD14−/− mice were larger than in all samples from WT mice (Fig. 9B), suggesting this tendency. However, little difference in the TGF-β immunoreactivity was observed between WT and CD14−/− mice after 90 dpi (Fig. 9A and B). However, there was little difference in the TGF-β immunoreactivity of WT and CD14−/− mice after 90 dpi. Double staining showed that TGF-β immunoreactivity was detected mostly in CD11b- and Iba1-positive microglia (Fig. 9C and D) and only occasionally in NeuN-positive neurons (Fig. 9E). Very little TGF-β immunoreactivity was detected in GFAP-positive astrocytes (Fig. 9F).

Fig 9.

Fig 9

Expression of TGF-β in the brains of WT and CD14−/− mice infected with the Chandler and Obihiro strains. (A) Immunofluorescence staining of TGF-β at 60, 90, and 120 dpi. Representative images from the thalamus are shown for each time point. Bars, 50 μm. Red, TGF-β; blue, nuclei. (B) Quantitative analysis of TGF-β-positive areas in the thalamus of Chandler-infected mice. The method and mice used for the quantification are the same as in the legend for Fig. 8. (C to F) Double immunofluorescence staining of TGF-β with CD11b (C), Iba1 (D), NeuN (E), and GFAP (F). Representative figures from the thalamus of the Chandler-infected mice are shown. Green, immunoreactivities for these markers; red, TGF-β; blue, nuclei. Arrows show double-positive cells. Bars, 10 μm.

Expression of proinflammatory cytokine.

We also assessed if a lack of CD14 influenced the expression of proinflammatory cytokines in the brains of prion-infected mice. Here, we focused on IL-1β, since a lack of IL-1β receptor signaling has been reported to delay the progression of prion diseases (7). Different from the situation in anti-inflammatory cytokines, more immunoreactivity of IL-1β was detected in WT mice than in CD14−/− mice at 60 and 90 dpi, particularly in the corpus callosum, internal capsules, and cerebral peduncles (Fig. 10A). Quantitative analysis showed that IL-1β-positive areas in the internal capsule of WT mice were significantly larger than the same areas of CD14−/− mice at 60 dpi (P < 0.05, Welch's t test) (Fig. 10B). Also, IL-1β-positive areas of WT mice tended to be larger than those of CD14−/− mice at 90 and 120 dpi (Fig. 10B), although statistical analyses could not be carried out due to the limited number of animals (n = 2). The differences in the expression of IL-1β of WT and CD14−/− mice appeared to be less prominent in the thalamus throughout the time of the observations (data not shown).

Fig 10.

Fig 10

Expression of IL-1β in the brains of prion-infected WT and CD14−/− mice. (A) Immunofluorescence staining of IL-1β in the internal capsule of mice infected with Chandler or Obihiro strains at 60, 90, and 120 dpi. Bars, 50 μm; red, IL-1β; blue, nuclei. (B) Quantitative analysis of IL-1β-positive areas in the thalamus of Chandler-infected mice. The method and mice used for the quantification are the same as in the legend for Fig. 8. *, P < 0.01, Welch's t test.

DISCUSSION

Prolonged survival of prion-infected CD14−/− mice and delayed PrPSc deposition in the brains suggest that CD14 plays a role in acceleration of disease progression after prion infection. Similarly, depletion of CD14 reduced Aβ deposition in a mouse model of AD (17), suggesting that CD14 could also play a role in other neurodegenerative disorders. However, microglial activation is different in prion-infected and AD model mice lacking CD14; microglia in CD14−/− mice infected with prions were more strongly activated than in WT mice, particularly in the early stage of the disease, whereas there is reduced microglial activation in AD model mice lacking CD14 (17).

There is a line of evidence that microglial activation causes detrimental effects in prion diseases. For instance, there is a report that a blockade of the colony-stimulating factor 1 receptor (CSF1R) signaling pathway reduced microglial proliferation in the brains of prion-infected mice and slowed the disease progression (31). Prion-infected mice deficient for CD40 ligand showed shortened incubation periods with increased microglial activation (32). Furthermore, prion-infected CXCR3-deficient mice survived longer than WT mice with reduced microglial activation, although prion propagation and PrPSc accumulation were accelerated (33). Here, in contrast, we showed that microglia were more activated in prion-infected CD14−/− mice than in WT mice but that prion-infected CD14−/− mice survived longer than WT mice. This result implies that microglial activation in the CD14−/− mice could offer a protective effect in the disease progression after prion infection. Particularly, the relationship between the increased microglial activation and delayed PrPSc accumulation in CD14−/− mice is intriguing. Priller et al. reported that during the course of prion infection, first the resident microglia were activated and thereafter bone marrow-derived microglia colonized the brain of the prion-infected mice, and more than 50% of the microglia were replaced by bone marrow-derived microglia by the onset of the disease (34). Thus, it is possible that the origin and activation state of microglia differ in the early and in the late stages of prion diseases. Here, we showed that the increased activation of microglia in prion-infected CD14−/− is concomitant with the upregulation of the anti-inflammatory cytokine production such as IL-10 and the delayed PrPSc accumulation. The precise phenotype of the microglia at this time point remains to be elucidated, but it is suggested that microglia activated in the early stage of prion infection in the CD14−/− mice possess a neuroprotective potential.

Although an accelerating role of CD14 in the progression of prion diseases is suggested by the current study, the mechanism by which CD14 works in the process of the disease needs to be further elucidated. One possible involvement of CD14 indicated by the results of the immunofluorescence staining for anti-inflammatory cytokines is that it downregulates alternative activation of microglia directly or indirectly. Induction of a proinflammatory response is another possible function of CD14 in prion diseases. CD14 is a GPI-anchoring protein and thus lacks an intracellular domain, but it works together with the TLR4 homodimer to induce cellular signaling involved in proinflammatory responses (12). Therefore, a lack of CD14 affects the signaling through the CD14/TLR4 complex, which could be a cause of the longer survival of prion-infected CD14−/− mice. However, a previous study showed that TLR4-signaling mutant mice, C3H/HeJ (Tlr4Lps-d) mice, possessing a single amino acid mutation in the cytoplasmic domain, were highly susceptible to prion infection, suggesting that TLR4 signaling can interfere with the disease progression (35). This is seemingly contradictory to the results of our study here. However, TLR4 is not the only counterpart of CD14: CD14 is reported to form clusters with a TLR2-TLR6 heterodimer (36). Ablation of myeloid differentiation factor 88 (MyD88), an obligate signal transducer adaptor protein for TLRs, did not affect the neuropathology of prion diseases after intracerebral inoculation of prions, indicating that TLR signaling through MyD88, resulting in NF-κb activation, is not involved in prion neuropathogenesis (37). However, as TLRs can utilize both MyD88-dependent and -independent pathways (38), TLR2 also utilizes the Toll/interleukin 1 receptor domain containing adaptor protein (39). Thus, it would seem possible that CD14 can transduce cellular signaling through an association with TLRs other than TLR4 and MyD88-independent pathways and be able to modulate the inflammatory milieu, which may influence the pathogenesis of prion diseases.

In addition to reduced deposition of pathogenic agents, another similarity of CD14−/− mice in prion infection and the AD model is the upregulation of the anti-inflammatory cytokine IL-10 (17). This makes it conceivable that the phenotype of the microglia shifted to an anti-inflammatory, alternative activation status, even though the TNF-α gene expression was also more upregulated in the AD model mice with the CD14−/− background than in those with the WT background (17). Among anti-inflammatory cytokines, both IL-10 and TGF-β have been reported to be involved in the pathogenesis of prion diseases. Thackray et al. reported that survival times of IL-10-deficient mice was remarkably short (8), although the mouse genetic background greatly influences the effect of IL-10 on prion diseases (40), suggesting a significant suppressive role of this cytokine in the disease progression. Inhibition of TGF-β activity facilitated cerebral inflammation and acute neuronal death in ME7-infected mice (9). IL-10 has been reported to have a neuroprotective role by blocking caspase-3-like activity (41), and TGF-β also promotes neuronal survival by upregulating anti-apoptotic Bcl-2 family proteins (42). Therefore, an earlier expression of these anti-inflammatory cytokines may partially contribute to the prolonged survival of CD14−/− mice. Furthermore, both IL-10 and TGF-β provide anti-inflammatory effects by downregulating proinflammatory mediators such as IL-1β, IL-6, TNF-α, and NO (4345). Indeed, expression of IL-1β in some brain regions of CD14−/− mice appeared to be lower than in WT mice (Fig. 10). However, it is also possible that the lower expression of IL-1β here resulted from a lack of TLR-CD14 signaling (46).

In AD model mice, bone marrow-derived microglia infiltrated from the peripheral circulation have protective potential against Aβ deposition and cognitive impairment in the early stage of the disease (47, 48), although it is controversial whether the protective effect is due to a clearance of Aβ by microglia (49, 50). However, it is also possible that at a later stage, increased Aβ production may overwhelm the microglial activity, or the proinflammatory milieu may decrease the phagocytic activity of microglia (51), or microglia may change the activation status to a more proinflammatory and neurotoxic phenotype (52). PrPSc is believed to be produced in intracellular organelles such as early and recycling endosomes as well as on the plasma membrane (5355). This would seem to exclude microglia from any role in the clearance of PrPSc itself by phagocytosis in the early stage of infection, although microglia remove neurons damaged by PrPSc accumulation (1, 56). Thus, the delayed PrPSc accumulation in the early stage of the disease in CD14−/− mice may imply the existence of a brain niche with accelerated microglial activation that may provide anti-prion propagation conditions, particularly at the early stage of the disease.

In this study, we showed that the increased activation of microglia accompanied by the altered expression of anti-inflammatory cytokines is possibly involved in the prolonged survival times and delayed deposition of PrPSc in prion-infected CD14−/− mice. This suggests that a CD14-dependent signaling pathway in microglia plays some role in the acceleration of the disease progression. Further analyses of the activation states of microglia and microglial functions would provide a better understanding of the roles of microglia in prion pathogenesis.

ACKNOWLEDGMENTS

This work was supported by a Grant-in-Aid for Science Research (A) (grant no. 23248050), a Grant for Challenging Exploratory Research (grant no. 23658233), a Grant-in-Aid for Young Researcher (B) (grant no. 23780303), and a grant from the global COE program (F-001) and a grant from the Program of Founding Research Centers for Emerging and Reemerging Infectious Diseases, from the Ministry of Education, Culture, Sports, Science, and Technology, Japan. This work was also supported by a grant for TSE research (H23-Shokuhin-Ippan-005) and a grant from the Research on Measures for Intractable Diseases program (grant no. H23-Nanchi-Ippan-013) from the Ministry of Health, Labor and Welfare of Japan. This work was also supported in part by a Grant-in-Aid from the BSE Control Project of the Ministry of Agriculture, Forestry and Fisheries of Japan.

We thank Zensho Co., Ltd., for use of the biosafety level 3 (BSL3) facility.

Footnotes

Published ahead of print 2 October 2013

REFERENCES

  • 1.Williams A, Lucassen PJ, Ritchie D, Bruce M. 1997. PrP deposition, microglial activation, and neuronal apoptosis in murine scrapie. Exp. Neurol. 144:433–438 [DOI] [PubMed] [Google Scholar]
  • 2.Giese A, Brown DR, Groschup MH, Feldmann C, Haist I, Kretzschmar HA. 1998. Role of microglia in neuronal cell death in prion disease. Brain Pathol. 8:449–457 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. 2011. Physiology of microglia. Physiol. Rev. 91:461–553 [DOI] [PubMed] [Google Scholar]
  • 4.Ransohoff RM, Perry VH. 2009. Microglial physiology: unique stimuli, specialized responses. Annu. Rev. Immunol. 27:119–145 [DOI] [PubMed] [Google Scholar]
  • 5.Block ML, Zecca L, Hong JS. 2007. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat. Rev. Neurosci. 8:57–69 [DOI] [PubMed] [Google Scholar]
  • 6.Neumann H, Kotter MR, Franklin RJ. 2009. Debris clearance by microglia: an essential link between degeneration and regeneration. Brain 132:288–295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Schultz J, Schwarz A, Neidhold S, Burwinkel M, Riemer C, Simon D, Kopf M, Otto M, Baier M. 2004. Role of interleukin-1 in prion disease-associated astrocyte activation. Am. J. Pathol. 165:671–678 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Thackray AM, McKenzie AN, Klein MA, Lauder A, Bujdoso R. 2004. Accelerated prion disease in the absence of interleukin-10. J. Virol. 78:13697–13707 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Boche D, Cunningham C, Docagne F, Scott H, Perry VH. 2006. TGFbeta1 regulates the inflammatory response during chronic neurodegeneration. Neurobiol. Dis. 22:638–650 [DOI] [PubMed] [Google Scholar]
  • 10.Hwang D, Lee IY, Yoo H, Gehlenborg N, Cho JH, Petritis B, Baxter D, Pitstick R, Young R, Spicer D, Price ND, Hohmann JG, Dearmond SJ, Carlson GA, Hood LE. 2009. A systems approach to prion disease. Mol. Syst. Biol. 5:252. 10.1038/msb.2009.10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Xiang W, Windl O, Wunsch G, Dugas M, Kohlmann A, Dierkes N, Westner IM, Kretzschmar HA. 2004. Identification of differentially expressed genes in scrapie-infected mouse brains by using global gene expression technology. J. Virol. 78:11051–11060 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chow JC, Young DW, Golenbock DT, Christ WJ, Gusovsky F. 1999. Toll-like receptor-4 mediates lipopolysaccharide-induced signal transduction. J. Biol. Chem. 274:10689–10692 [DOI] [PubMed] [Google Scholar]
  • 13.Kirschning CJ, Wesche H, Merrill Ayres T, Rothe M. 1998. Human toll-like receptor 2 confers responsiveness to bacterial lipopolysaccharide. J. Exp. Med. 188:2091–2097 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Letiembre M, Liu Y, Walter S, Hao W, Pfander T, Wrede A, Schulz-Schaeffer W, Fassbender K. 2009. Screening of innate immune receptors in neurodegenerative diseases: a similar pattern. Neurobiol. Aging 30:759–768 [DOI] [PubMed] [Google Scholar]
  • 15.Baldini M, Lohman IC, Halonen M, Erickson RP, Holt PG, Martinez FD. 1999. A Polymorphism* in the 5′ flanking region of the CD14 gene is associated with circulating soluble CD14 levels and with total serum immunoglobulin E. Am. J. Respir. Cell Mol. Biol. 20:976–983 [DOI] [PubMed] [Google Scholar]
  • 16.Lin JJ, Chen CH, Yueh KC, Chang CY, Lin SZ. 2006. A CD14 monocyte receptor polymorphism and genetic susceptibility to Parkinson's disease for females. Parkinsonism Relat. Disord. 12:9–13 [DOI] [PubMed] [Google Scholar]
  • 17.Reed-Geaghan EG, Reed QW, Cramer PE, Landreth GE. 2010. Deletion of CD14 attenuates Alzheimer's disease pathology by influencing the brain's inflammatory milieu. J. Neurosci. 30:15369–15373 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kim CL, Umetani A, Matsui T, Ishiguro N, Shinagawa M, Horiuchi M. 2004. Antigenic characterization of an abnormal isoform of prion protein using a new diverse panel of monoclonal antibodies. Virology 320:40–51 [DOI] [PubMed] [Google Scholar]
  • 19.Paxinos G, Franklin KBJ. 2001. The mouse brain in stereotaxic coordinates, 2nd ed. Academic Press, San Diego, CA [Google Scholar]
  • 20.Uryu M, Karino A, Kamihara Y, Horiuchi M. 2007. Characterization of prion susceptibility in Neuro2a mouse neuroblastoma cell subclones. Microbiol. Immunol. 51:661–669 [DOI] [PubMed] [Google Scholar]
  • 21.Yamasaki T, Suzuki A, Shimizu T, Watarai M, Hasebe R, Horiuchi M. 2012. Characterization of intracellular localization of PrP(Sc) in prion-infected cells using a mAb that recognizes the region consisting of aa 119-127 of mouse PrP. J. Gen. Virol. 93:668–680 [DOI] [PubMed] [Google Scholar]
  • 22.Song CH, Furuoka H, Kim CL, Ogino M, Suzuki A, Hasebe R, Horiuchi M. 2008. Effect of intraventricular infusion of anti-prion protein monoclonal antibodies on disease progression in prion-infected mice. J. Gen. Virol. 89:1533–1544 [DOI] [PubMed] [Google Scholar]
  • 23.Pastrana MA, Sajnani G, Onisko B, Castilla J, Morales R, Soto C, Requena JR. 2006. Isolation and characterization of a proteinase K-sensitive PrPSc fraction. Biochemistry 45:15710–15717 [DOI] [PubMed] [Google Scholar]
  • 24.Tzaban S, Friedlander G, Schonberger O, Horonchik L, Yedidia Y, Shaked G, Gabizon R, Taraboulos A. 2002. Protease-sensitive scrapie prion protein in aggregates of heterogeneous sizes. Biochemistry 41:12868–12875 [DOI] [PubMed] [Google Scholar]
  • 25.Fischer M, Rulicke T, Raeber A, Sailer A, Moser M, Oesch B, Brandner S, Aguzzi A, Weissmann C. 1996. Prion protein (PrP) with amino-proximal deletions restoring susceptibility of PrP knockout mice to scrapie. EMBO J. 15:1255–1264 [PMC free article] [PubMed] [Google Scholar]
  • 26.Ford AL, Goodsall AL, Hickey WF, Sedgwick JD. 1995. Normal adult ramified microglia separated from other central nervous system macrophages by flow cytometric sorting. Phenotypic differences defined and direct ex vivo antigen presentation to myelin basic protein-reactive CD4+ T cells compared. J. Immunol. 154:4309–4321 [PubMed] [Google Scholar]
  • 27.Becher B, Antel JP. 1996. Comparison of phenotypic and functional properties of immediately ex vivo and cultured human adult microglia. Glia 18:1–10 [DOI] [PubMed] [Google Scholar]
  • 28.Cosenza MA, Zhao ML, Si Q, Lee SC. 2002. Human brain parenchymal microglia express CD14 and CD45 and are productively infected by HIV-1 in HIV-1 encephalitis. Brain Pathol. 12:442–455 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Cosenza-Nashat MA, Kim MO, Zhao ML, Suh HS, Lee SC. 2006. CD45 isoform expression in microglia and inflammatory cells in HIV-1 encephalitis. Brain Pathol. 16:256–265 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Masliah E, Mallory M, Hansen L, Alford M, Albright T, Terry R, Shapiro P, Sundsmo M, Saitoh T. 1991. Immunoreactivity of CD45, a protein phosphotyrosine phosphatase, in Alzheimer's disease. Acta Neuropathol. 83:12–20 [DOI] [PubMed] [Google Scholar]
  • 31.Gomez-Nicola D, Fransen NL, Suzzi S, Perry VH. 2013. Regulation of microglial proliferation during chronic neurodegeneration. J. Neurosci. 33:2481–2493 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Burwinkel M, Schwarz A, Riemer C, Schultz J, van Landeghem F, Baier M. 2004. Rapid disease development in scrapie-infected mice deficient for CD40 ligand. EMBO Rep. 5:527–531 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Riemer C, Schultz J, Burwinkel M, Schwarz A, Mok SW, Gultner S, Bamme T, Norley S, van Landeghem F, Lu B, Gerard C, Baier M. 2008. Accelerated prion replication in, but prolonged survival times of, prion-infected CXCR3−/− mice. J. Virol. 82:12464–12471 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Priller J, Prinz M, Heikenwalder M, Zeller N, Schwarz P, Heppner FL, Aguzzi A. 2006. Early and rapid engraftment of bone marrow-derived microglia in scrapie. J. Neurosci. 26:11753–11762 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Spinner DS, Cho IS, Park SY, Kim JI, Meeker HC, Ye X, Lafauci G, Kerr DJ, Flory MJ, Kim BS, Kascsak RB, Wisniewski T, Levis WR, Schuller-Levis GB, Carp RI, Park E, Kascsak RJ. 2008. Accelerated prion disease pathogenesis in Toll-like receptor 4 signaling-mutant mice. J. Virol. 82:10701–10708 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Henneke P, Takeuchi O, van Strijp JA, Guttormsen HK, Smith JA, Schromm AB, Espevik TA, Akira S, Nizet V, Kasper DL, Golenbock DT. 2001. Novel engagement of CD14 and multiple toll-like receptors by group B streptococci. J. Immunol. 167:7069–7076 [DOI] [PubMed] [Google Scholar]
  • 37.Prinz M, Heikenwalder M, Schwarz P, Takeda K, Akira S, Aguzzi A. 2003. Prion pathogenesis in the absence of Toll-like receptor signalling. EMBO Rep. 4:195–199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kawai T, Akira S. 2010. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat. Immunol. 11:373–384 [DOI] [PubMed] [Google Scholar]
  • 39.Kenny EF, Talbot S, Gong M, Golenbock DT, Bryant CE, O'Neill LA. 2009. MyD88 adaptor-like is not essential for TLR2 signaling and inhibits signaling by TLR3. J. Immunol. 183:3642–3651 [DOI] [PubMed] [Google Scholar]
  • 40.Tamguney G, Giles K, Glidden DV, Lessard P, Wille H, Tremblay P, Groth DF, Yehiely F, Korth C, Moore RC, Tatzelt J, Rubinstein E, Boucheix C, Yang X, Stanley P, Lisanti MP, Dwek RA, Rudd PM, Moskovitz J, Epstein CJ, Cruz TD, Kuziel WA, Maeda N, Sap J, Ashe KH, Carlson GA, Tesseur I, Wyss-Coray T, Mucke L, Weisgraber KH, Mahley RW, Cohen FE, Prusiner SB. 2008. Genes contributing to prion pathogenesis. J. Gen. Virol. 89:1777–1788 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Bachis A, Colangelo AM, Vicini S, Doe PP, De Bernardi MA, Brooker G, Mocchetti I. 2001. Interleukin-10 prevents glutamate-mediated cerebellar granule cell death by blocking caspase-3-like activity. J. Neurosci. 21:3104–3112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kim ES, Kim RS, Ren RF, Hawver DB, Flanders KC. 1998. Transforming growth factor-beta inhibits apoptosis induced by beta-amyloid peptide fragment 25-35 in cultured neuronal cells. Brain Res. Mol. Brain Res. 62:122–130 [DOI] [PubMed] [Google Scholar]
  • 43.Bogdan C, Nathan C. 1993. Modulation of macrophage function by transforming growth factor beta, interleukin-4, and interleukin-10. Ann. N. Y. Acad. Sci. 685:713–739 [DOI] [PubMed] [Google Scholar]
  • 44.Ledeboer A, Breve JJ, Poole S, Tilders FJ, Van Dam AM. 2000. Interleukin-10, interleukin-4, and transforming growth factor-beta differentially regulate lipopolysaccharide-induced production of pro-inflammatory cytokines and nitric oxide in co-cultures of rat astroglial and microglial cells. Glia 30:134–142 [DOI] [PubMed] [Google Scholar]
  • 45.Seyler I, Appel M, Devissaguet JP, Legrand P, Barratt G. 1997. Modulation of nitric oxide production in RAW 264.7 cells by transforming growth factor-beta and interleukin-10: differential effects on free and encapsulated immunomodulator. J. Leukoc. Biol. 62:374–380 [DOI] [PubMed] [Google Scholar]
  • 46.Fitzgerald KA, Rowe DC, Golenbock DT. 2004. Endotoxin recognition and signal transduction by the TLR4/MD2-complex. Microbes Infect. 6:1361–1367 [DOI] [PubMed] [Google Scholar]
  • 47.El Khoury J, Toft M, Hickman SE, Means TK, Terada K, Geula C, Luster AD. 2007. Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease. Nat. Med. 13:432–438 [DOI] [PubMed] [Google Scholar]
  • 48.Simard AR, Soulet D, Gowing G, Julien JP, Rivest S. 2006. Bone marrow-derived microglia play a critical role in restricting senile plaque formation in Alzheimer's disease. Neuron 49:489–502 [DOI] [PubMed] [Google Scholar]
  • 49.Grathwohl SA, Kalin RE, Bolmont T, Prokop S, Winkelmann G, Kaeser SA, Odenthal J, Radde R, Eldh T, Gandy S, Aguzzi A, Staufenbiel M, Mathews PM, Wolburg H, Heppner FL, Jucker M. 2009. Formation and maintenance of Alzheimer's disease beta-amyloid plaques in the absence of microglia. Nat. Neurosci. 12:1361–1363 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Mildner A, Schlevogt B, Kierdorf K, Bottcher C, Erny D, Kummer MP, Quinn M, Bruck W, Bechmann I, Heneka MT, Priller J, Prinz M. 2011. Distinct and non-redundant roles of microglia and myeloid subsets in mouse models of Alzheimer's disease. J. Neurosci. 31:11159–11171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Koenigsknecht-Talboo J, Landreth GE. 2005. Microglial phagocytosis induced by fibrillar beta-amyloid and IgGs are differentially regulated by proinflammatory cytokines. J. Neurosci. 25:8240–8249 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Jimenez S, Baglietto-Vargas D, Caballero C, Moreno-Gonzalez I, Torres M, Sanchez-Varo R, Ruano D, Vizuete M, Gutierrez A, Vitorica J. 2008. Inflammatory response in the hippocampus of PS1M146L/APP751SL mouse model of Alzheimer's disease: age-dependent switch in the microglial phenotype from alternative to classic. J. Neurosci. 28:11650–11661 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Godsave SF, Wille H, Pierson J, Prusiner SB, Peters PJ. 2013. Plasma membrane invaginations containing clusters of full-length PrP(Sc) are an early form of prion-associated neuropathology in vivo. Neurobiol. Aging 34:1621–1631 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Godsave SF, Wille H, Kujala P, Latawiec D, DeArmond SJ, Serban A, Prusiner SB, Peters PJ. 2008. Cryo-immunogold electron microscopy for prions: toward identification of a conversion site. J. Neurosci. 28:12489–12499 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Jeffrey M, Goodsir CM, Bruce ME, McBride PA, Scott JR, Halliday WG. 1992. Infection specific prion protein (PrP) accumulates on neuronal plasmalemma in scrapie infected mice. Neurosci. Lett. 147:106–109 [DOI] [PubMed] [Google Scholar]
  • 56.Hughes MM, Field RH, Perry VH, Murray CL, Cunningham C. 2010. Microglia in the degenerating brain are capable of phagocytosis of beads and of apoptotic cells, but do not efficiently remove PrPSc, even upon LPS stimulation. Glia 58:2017–2030 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Virology are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES