The American Journal of Pathology, Vol. 185, No. 1, January 2015

ajp.amjpathol.org

NEUROBIOLOGY

Prostaglandin E2 Receptor Subtype 2 Regulation of Scavenger Receptor CD36 Modulates Microglial Ab42 Phagocytosis Xianwu Li, Erica Melief, Nadia Postupna, Kathleen S. Montine, C. Dirk Keene, and Thomas J. Montine From the Department of Pathology, University of Washington, Seattle, Washington Accepted for publication September 18, 2014. Address correspondence to Xianwu Li, Ph.D., Department of Pathology, University of Washington, Box 359645, Seattle, WA 98104. E-mail: [email protected].

Recent studies underline the potential relevance of microglial innate immune activation in Alzheimer disease. Primary mouse microglia that lack prostaglandin E2 receptor subtype 2 (EP2) show decreased innate immune-mediated neurotoxicity and increased amyloid b (Ab) peptide phagocytosis, features that were replicated in vivo. Here, we tested the hypothesis that scavenger receptor CD36 is an effector of EP2regulated Ab phagocytosis. CD36 expression was 143-fold greater in mouse primary microglia than in primary astrocytes. Three different means of suppressing EP2 signaling increased and an agonist of EP2 decreased CD36 expression in primary wild-type microglia. Activation of Toll-like receptor (TLR) 3, TLR4, and TLR7, but not TLR2 or TLR9, reduced primary microglial CD36 transcription and cell surface CD36 protein and reduced Ab42 phagocytosis as well. At each step, the effects of innate immune activation on CD36 were reversed by at least 50% by an EP2 antagonist, and this partial rescue of microglia Ab42 phagocytosis was largely mediated by CD36 activity. Finally, we showed in hippocampus of wild-type mice that innate immune activation suppressed CD36 expression by an EP2-dependent mechanism. Taken together with results of others that found brain clearance of Ab peptides and behavioral improvements mediated by CD36 in mice, regulation of CD36-mediated Ab phagocytosis by suppression of EP2 signaling may provide a new approach to suppressing some aspects of Alzheimer disease pathogenesis. (Am J Pathol 2015, 185: 230e239; http://dx.doi.org/10.1016/j.ajpath.2014.09.016)

Observational studies in patients and experimental studies that use a variety of model systems have concluded that affected regions of the brain in Alzheimer disease (AD) experience a proinflammatory, pro-oxidative state.1 Indeed, recent studies have highlighted several genes involved in the innate immune response whose variants are associated with increased risk of AD, especially the TREM2 gene.2e4 TREM2 encodes the triggering receptor expressed on myeloid cells-2, a protein expressed by myeloid lineage cells, including microglia in brain, which can function as a receptor for phagocytosis,5 modulate the innate immune response in brain,6,7 and diminish immune response through several signaling pathways, including suppression of Toll-like receptor (TLR) signaling.8,9 The prostaglandin (PG) pathway, a cascade initiated by enzyme-catalyzed synthesis of PGH2 from arachidonic acid by the cyclooxygenases (COXs), COX-1 or COX-2, is an essential component of the innate immune response. Multiple studies Copyright ª 2015 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.ajpath.2014.09.016

have reproducibly shown that nonselective COX or COX2eselective inhibition by nonsteroidal anti-inflammatory drugs (NSAIDs) can suppress amyloid b (Ab) accumulation and associated behavioral changes in transgenic mouse models of AD10 and can reduce the risk of AD dementia in people by approximately 60%.11 Together these results suggest that NSAIDs may effectively suppress some aspect of what is now appreciated to be early steps in AD pathogenesis.12 These compelling experimental model and human data formed the rationale for multiple clinical trials. Two clinical trials targeted what is now understood to be relatively advanced AD: one with NSAIDs in AD dementia and one with NSAIDs in mild cognitive impairment, a diagnostic group enriched for prodromal AD; both failed.13,14 A third trial targeted early-stage Supported by NIH grants ES016754, ES007032, AG005136, and NS062684; and the Nancy and Buster Alvord Endowment. Disclosures: None declared.

EP2-Suppressed Phagocytosis via CD36 AD,15 similar to the epidemiologic cohorts, but was terminated because of toxicity, likely related to perturbation of prostacyclin and thromboxane metabolism.16e18 Despite these disappointing therapeutic outcomes, some data suggest pharmacodynamic effects of NSAIDs in the central nervous system of patients with AD.19,20 We and others have sought to disentangle the potentially beneficial effects of NSAIDs in early AD pathogenesis from toxic effects by focusing on specific PG receptors distal in the signaling cascade rather than proximal COX inhibition.21e23 PGE2 is the principal proinflammatory PG, and increased PGE2 concentrations in cerebrospinal fluid have been found in patients with AD,24 particularly early in the disease process.25 In addition, the inducible form of PGE2 synthase is increased in AD brain and is a key component in Ab-mediated neurotoxicity in a mouse model of AD.26 We, therefore, have focused on PGE2 and its functionally antagonistic receptor subtypes, EP1 to EP4, which are widely expressed in central nervous system cells, including microglia, astrocytes, and neurons.21 Primary microglia from mice homozygous deficient for the EP2 gene (PTGER2; EP2/) have the highly desirable dual phenotype of suppressed TLR-activated microglia-mediated neurotoxicity that depends on the adaptor protein DOCK2 and enhanced microglial phagocytosis of Ab that is DOCK2 independent.27e29 These studies show that microglial EP2 is necessary for lipopolysaccharide-induced microglia-mediated neurotoxicity in microgliaeneuron cocultures through regulation of inducible nitric oxide synthase and COX-2 and subsequent neurotoxic cytokine production, including IL-1b production,27 and indicate EP2-dependent neurotoxicity in EP2/ microglia, with reduced paracrine neurotoxicity in response to aggregated Ab in neuronemicroglia cocultures.28 Recently, these findings were confirmed in mice with conditional deletion of EP2 in myeloid lineage cells and in bone marrow transplant experiments.30,31 Microglial phagocytosis is mediated largely by a class of surface proteins called scavenger receptors. CD36, a type B scavenger receptor originally identified as a receptor for longchain fatty acids and oxidized low-density lipoprotein, is a microglial binding site for Ab,32 facilitates intracellular nucleation of Ab peptides into fibrils,33 and mediates some of the biological effects of Ab peptides, including innate immune activation and oxidative stress.34e36 CD36 is expressed on microglia in AD brains.34 CD36 overexpression in human brain correlates with b-amyloid deposition but not with AD; it is undetectable in healthy brains without Ab deposition.37 We are unaware of any data to link the specific PG receptors with CD36 function. Here, we tested the hypothesis that EP2 suppression of Ab phagocytosis is mediated, at least in part, by CD36.

Materials and Methods Materials Dulbecco’s modified Eagle’s medium/F-12 medium and fetal bovine serum were purchased from Hyclone

The American Journal of Pathology

-

ajp.amjpathol.org

Laboratories (Logan, UT). Papain and DNase I were from Worthington Biochemical (Lakewood, NJ). Butaprost, NS-398, 17-phenyl trinor PGE2, CAY10598, ZK118182, 17-phenyl trinor PGF2a, carbaprostacyclin, U-46619, AH 6809, and CD36 blocking antibody (Clone JC63.1) were from Cayman Chemical Company (Ann Arbor, MI). Dibutyryl cAMP, forskolin, tribromoethanol (Avertin), and polyinosinic-polycytidylic acid (PIC; TLR3 ligand) were from Sigma-Aldrich (St. Louis, MO). Pam3CSK4 (TLR2 ligand), loxoribine (TLR7 ligand), and CpG (TLR9 ligand) were from Invivogen (San Diego, CA). Lipopolysaccharide (TLR4 ligand) was purchased from Calbiochem (La Jolla, CA). pHrodo fluorogenic dye was from Invitrogen (Carlsbad, CA). b Amyloid 1-42 (Ab42) was from American Peptide Company (Sunnyvale, CA).

Animals C57BL/6 mice and CD36/ (B6.129S1-Cd36tm1Mfe/J) mice were purchased from The Jackson Laboratory (Bar Harbor, ME). EP2/ mice were a gift from Dr. Richard Breyer (Vanderbilt University, Nashville, TN). The animals were maintained in a specific pathogen-free environment. All procedures were approved by the University of Washington Institutional Animal Care and Use Committee.

Stereotactic Intracerebroventricular Injection Ten-week-old male mice were anesthetized with isoflurane inhalation and placed in a stereotactic instrument. After the skull was exposed through skin incision, a cranial window (2 to 3 mm) on the left lateral ventricle was opened by drilling with a small burr bit. Five microliter of reagent was injected over a period of 5 minutes by using a Hamilton microsyringe; injections were 200 ng of AH 6809, 50 ng of PIC, AH 6809 plus PIC, or vehicle. After injection, the mice were placed on an isothermal pad and continuously observed until recovery. Twenty-four hours after injection, the mice were sacrificed by i.p. injection of tribromoethanol and perfused with ice-cold phosphatebuffered saline, and brain tissues were rapidly harvested for analysis.

Cell Culture and Exposure Primary microglia and astrocytes were isolated from brains of newborn mice and cultured as previously described.38,39 Medium concentrations of PGE2 were measured by ELISA exactly as described previously.38 Cells were incubated with the compounds described at the indicated concentrations and times. We note that, although EP antagonist AH 6809 has nearly equal affinity for EP1 and DP1 in human tissue,40 it has been shown to be selective for EP2 in mouse tissue.41 EP agonist butaprost was used at 20 mmol/L, based on previous studies that showed effects up to 100 mmol/L.42,43

231

Li et al

Quantitative Real-Time PCR Total RNA was extracted from primary microglia, astrocytes, or brain tissues by using RNeasy Mini Kit (Qiagen, Valencia, CA). Reverse transcription was performed with Omniscript RT Kit (Qiagen). TaqMan probes and primers (Mm01135198_m1 for CD36, fMm99999915_g1 for GAPDH) were purchased from Applied Biosystems (Carlsbad, CA). PCR were performed on an Applied Biosystems ViiA 7 Real-Time PCR System by using the relative quantitative method.

Flow Cytometric Analysis of Cell Surface CD36 in Primary Microglia Microglia were preincubated with 10 mg/mL of anti-mouse CD16/CD32 antibody (BD Biosciences, San Jose, CA) for 5 minutes on ice to block Fc receptoremediated binding. The cells were then incubated with 5 mg/mL of phycoerythrinconjugated CD36 or mouse IgA control (Santa Cruz Biotechnology, Inc., Santa Cruz, CA) for 30 minutes on ice in the dark. Thereafter, microglia were washed twice with phosphate-buffered saline and resuspended in 4% paraformaldehyde for fixation. The fluorescence intensity was determined by BD FACScan flow cytometry.

Ab42 Phagocytosis in Primary Microglia Microglia were incubated with 5 mmol/L pHrodo-labeled Ab42 for 2 hours at 37 C. The nonspecific signal was determined by incubation of microglia with the same concentration of labeled Ab42 for 2 hours at 4 C. Thereafter, microglia were washed three times in phosphate-buffered saline and fixed in 4% paraformaldehyde. The samples were analyzed by LSRII flow cytometry (BD Biosciences) with a 561-nm excitation laser.

CD36 Expression in CHO Cells Human CD36 was expressed in Chinese hamster ovary (CHO) cells as described by others.44 Briefly, cells were maintained in Ham’s F-12 medium that contained 10% fetal bovine serum and penicillin/streptomycin. Human full-length CD36 cDNA was obtained from GE Healthcare (Lafayette, CO) and cloned into the expression vector pIRES2-EGFP at EcoRI and XbaI sites. The recombinant vector or empty vector was transfected into CHO cells by Lipofectamine 2000 according to the manufacturer’s protocol. The expression of CD36 was confirmed by quantitative PCR with Taqman expression assay. Two days after transfection, cells were collected for phagocytosis assay and inhibition with sulfosuccinimidyl oleate (SSO) as described by others.45

Statistical Analysis Statistical analyses were performed with GraphPad Prism 5 (GraphPad Inc., San Diego, CA) with a set to 0.05. All P values

232

presented in the text or figures were corrected for multiple comparisons when appropriate with the method of Bonferroni.

Results Microglia Express Much More CD36 than Astrocytes Microglia, and less so astrocytes, are cellular effectors of innate immune response in the cerebral cortex. CD36 expression in primary wild-type (WT) cultures of microglia and astrocytes prepared from the same five mice was determined by quantitative real-time PCR with each sample normalized to endogenous control GAPDH. CD36 expression in microglia was on average 143-fold greater in microglial than in astrocytes (P < 0.0001). For this reason, subsequent experiments focused on microglia.

Down-Regulation of CD36 Expression by Specific TLR Activation Because many endogenous activators of innate immune response, including Ab,46e49 act through TLRs and because one function of TREM2 is to regulate TLR signaling,8 we measured primary WT mouse microglia CD36 expression after exposure for 18 hours to specific TLR activators that are known to activate microglia.38 Activators of TLR3, TLR4, and TLR7, but not TLR2 or TLR9, suppressed microglia expression of CD36 (Figure 1A). A seven-point time series (Figure 1B) showed no change in CD36 expression after 2 hours of PIC exposure (1.03  0.05 CD36 mRNA compared with time 0), but after 4 hours of exposure CD36 mRNA was reduced to 0.74  0.03 and continued to decrease linearly to 0.16  0.03 at 18 hours (R2 Z 0.95, P < 0.0001). A similar time course for medium PGE2 was approximately complementary with 23  2 pg/mL at time 0 and increased to 2199  64 pg/mL at 18 hours (P < 0.0001). Other scavenger receptors also were altered by PIC incubation for 18 hours: microglial expression of SR-A1 was increased 3.1-  0.7-fold (P < 0.01) and SR-B1 was decreased 0.25-  0.06-fold (P < 0.001) compared with vehicle-exposed cultures.

Regulation of CD36 Expression by EP2 We next surveyed specific agonists to prostanoid receptors EP, DP, FP, IP, and TP to determine which, if any, specific product of COX might contribute to innate immune-mediated reduction in CD36 expression. Agonists of EP2 or EP4 mimicked the effect of innate immune activation by significantly reducing CD36 expression in primary WT mouse microglia, whereas agonists of EP1 or TP had a functionally antagonistic effect; agonists of FP, IP, and DP had no significant effect on CD36 at known effective concentrations (Figure 2A).39 In contrast, a COX-2eselective inhibitor, an EP2 antagonist, and EP2/ microglia had progressively increasing microglial CD36 expression. Activation of the innate immune response

ajp.amjpathol.org

-

The American Journal of Pathology

EP2-Suppressed Phagocytosis via CD36

Figure 1

Effects of TLR activators on CD36 mRNA expression. A: WT mouse primary microglia were treated with Veh or 1 mg/mL activator of TLR2 (Pam3CSK4), 20 mg/mL TLR3 (PIC), 1 mg/mL TLR4 (lipopolysaccharide), 1 mmol/L TLR7 (loxoribine), or 1 mmol/L TLR9 (CpG) for 18 hours. RNA was extracted for quantitative real-time PCR. CD36 mRNA values in each sample were normalized to endogenous control GAPDH. B: WT mouse primary microglia were treated with 20 mg/mL PIC for the indicated times. CD36 mRNA was assessed by quantitative real-time PCR, and results were normalized to GAPDH. Medium PGE2 was assayed by ELISA. Data are expressed as means  SEM. n Z 3 per group. ***P < 0.001 for Bonferronicorrected repeated pair comparisons with vehicle (A) or with value at time 0 (B). PGE2, prostaglandin E2; PIC, polyinosinic-polycytidylic acid; TLR, Toll-like receptor; Veh, vehicle; WT, wild-type.

with agents specific for TLR3, TLR4, or TLR7 each reproducibly decreased microglial CD36 expression, an effect that was mostly reversed for each TLR activator with an EP2 antagonist (AH 6809) (Figure 2B); although reversal by AH 6809 was greatest in the context of TLR3 activation. A selective antagonist of EP4 is not commercially available.50,51 EP2 and EP4 are stimulatory G-protein-couple receptors, but because EP2 also can activate G-proteineindependent second messenger signaling,52 we tested the effect of directly manipulating cAMP signaling on CD36 expression; dibutyryl cAMP or forskolin decreased CD36 mRNA values by the same magnitude as butaprost alone (Figure 2C). These data show that multiple eicosanoid products of COX exert tonic control over CD36 expression in mouse primary microglia and that among these EP2 or EP4 activation suppresses CD36 expression to an amount matched by direct activation of cAMP second messenger signaling. Finally, results by using an antagonist show that EP2 signaling is responsible for most reduced microglial CD36 expression with TLR-activated innate immune response.

The American Journal of Pathology

-

ajp.amjpathol.org

Figure 2 Effects of eicosanoid receptors with and without innate immune activation on CD36 mRNA expression by mouse primary microglia. RNA was isolated from cultures prepared from wild-type mice (dark bars) or EP2/ mice (light bar) after 18 hours of incubation. Results from quantitative real-time PCR were normalized to endogenous control GAPDH. A: Microglia exposed to vehicle or 5 mmol/L agonists of EP1 (17-phenyl trinor PGE2), 20 mmol/L EP2 (butaprost), 2.5 mmol/L EP4 (CAY10598), 10 mmol/L DP (ZK118182), 10 mmol/L FP (17-phenyl trinor PGF2a), 10 mmol/L IP (carbaprostacyclin), or 5 mmol/L TP (U-46619). B: Microglia exposed to vehicle or 20 mmol/L COX-2eselective inhibitor (NS398), 20 mmol/L EP2 antagonist AH, and TLR activators (PIC for TLR3 at 20 mg/mL, LPS for TLR4 at 1 mg/mL, or loxoribine for TLR7 at 0.5 mmol/L) with or without 20 mmol/L AH. C: Microglia exposed to 20 mmol/L butaprost, 250 mmol/L db cAMP, or 10 mmol/L forskolin. Data are expressed as means  SEM. n Z 3 to 12 per group. *P < 0.05, **P < 0.01, and ***P < 0.001 for Bonferronicorrected repeated pair comparisons with vehicle; yP < 0.05 and yyP < 0.001 for Bonferroni-corrected paired comparisons between two experimental groups. AH, AH 6809; COX-2, cyclooxygenase 2; db cAMP, dibutyryl cAMP; EP2/, homozygous EP2 deficient; LPS, lipopolysaccharide; PGE2, prostaglandin E2; PIC, polyinosinic-polycytidylic acid; TLR, Toll-like receptor; Veh, vehicle.

233

Li et al CD36 expression is transcriptionally regulated, but we nevertheless sought to validate the changes observed in microglia CD36 mRNA expression with changes in CD36 protein. Flow cytometric analysis of primary WT mouse microglia labeled with phycoerythrin-conjugated anti-CD36 antibody; 24 hours of exposure to butaprost, an EP2 agonist, significantly reduced CD36 cell surface protein to values that were proportionate to mRNA reduction (Figure 3). Conversely, exposure to AH 6809 increased CD36 cell surface protein values. These results are consistent with the known transcriptional regulation of CD36 protein and indicate that changes in microglia cell surface CD36 protein follow the same pattern as CD36 mRNA in response to an agonist or antagonist of EP2.

Inhibition of CD36-Dependent Phagocytosis by EP2 Activation Our next series of experiments used primary WT and CD36/ mouse microglia to investigate whether CD36 mediated EP2

Figure 3 Effects of TLR3 activator and prostaglandin EP2 signaling on CD36 protein. WT microglia were treated with 20 mmol/L butaprost, 20 mmol/L AH, 20 mg/mL PIC, or a combination of PIC and AH for 24 hours. CD36 protein on cell surface was assessed by staining with PE-labeled antiCD36 antibody or isotype-matched PE-IgA (isotype control). A: Representative histogram from flow cytometric assay of CD36 immunofluorescence. B: Geometric means of fluorescence intensity was normalized to vehicle. Data are expressed as geometric means of fluorescence intensity  SEMs. n  3 per group. **P < 0.01 and ***P < 0.001 for Bonferroni-corrected repeated paired comparisons with vehicle; yP < 0.05 and yyP < 0.001 for Bonferroni-corrected paired comparisons between two experimental groups. AH, AH 6809; Buta, butaprost; CT, control; EP2, E2 receptor subtype 2; PE, phycoerythrin; PIC, polyinosinic-polycytidylic acid; TLR, Tolllike receptor; WT, wild-type.

234

effects on Ab42 phagocytosis. We quantified phagocytosis of Ab42 labeled with a pH-sensitive dye (pHrodo) and under the same conditions that affected CD36 mRNA and cell surface protein values (shown in Figures 2 and 3). Butaprost significantly decreased Ab42 phagocytosis in WT microglia, validating previous experiments from our laboratory that used EP2/ microglia with synthetic Ab42 and human brain-derived Ab peptides (Figure 4A).28,30,53 TLR3 activation from PIC exposure significantly reduced Ab42 phagocytosis by WT microglia, and this immune suppression was significantly but partially reversed by AH 6809, paralleling the effect of PIC on CD36 expression and indicating that EP2 activation accounted for approximately one-half of PIC-suppressed Ab42 phagocytosis. CD36 inhibitor SSO reversed this rescue, consistent with CD36-dependent phagocytosis. Because SSO may have actions in addition to inhibiting CD36, we sought to validate this finding with three different experiments. In the first, we used AH 6809 (coincubated with control IgA) to achieve increased Ab42 phagocytosis (P < 0.01) that paralleled increased CD36 expression (Figure 4B); this prophagocytic effect of AH 6809 was completely blocked by a CD36 neutralizing antibody. Consistent with this, CD36 blocking antibodies did not further inhibit butaprost-treated cells. In the second set of experiments, we compared phagocytosis in microglia prepared from WT and CD36/ mice (Figure 4C). CD36/ microglia had reduced Ab42 phagocytosis compared with WT microglia (P < 0.001). Importantly, increased Ab42 phagocytosis seen with exposure to AH 6809 was ablated in CD36/ microglia, whereas butaprost had no significant effect on Ab42 phagocytosis in these cells. In combination, results from WT microglia with the use of SSO or a CD36neutralizing antibody and from CD36/ microglia support the conclusion that the effects of EP2 activation on Ab42 phagocytosis are largely mediated by CD36. Finally, we transfected CHO cells with human CD36 as described by others.44 Vector-transfected CHO cell uptake of Ab42 was defined as 100%. Expression of human CD36 significantly increased uptake of Ab42 by CHO cells to 182%  6% of vehicle-transfected control (P < 0.001), similar to what has been reported previously by others.44 Also validating the results of others,45 exposure to SSO by using the same method as described above for mouse primary microglia completely inhibited Ab42 uptake by human CD36expressing CHO cells to 102%  9% of vehicle-transfected control; Ab42 uptake by vehicle-transfected CHO cells was not significantly altered by SSO (94%  8% of vectortransfected cells without SSO).

Reduced CD36 Expression by PIC Largely Depends on EP2 in Mouse Brain Finally, we investigated whether EP2 regulated CD36 expression in a relevant region of WT mouse brain. Hippocampal CD36 mRNA values were quantified 24 hours after 50 ng intracerebroventricular injection of PIC into the

ajp.amjpathol.org

-

The American Journal of Pathology

EP2-Suppressed Phagocytosis via CD36 left lateral ventricle with or without two different doses of the EP2 antagonist AH 6809 (200 or 50 ng) (Figure 5). Our results showed that direct exposure to TLR3 activator PIC significantly reduced ipsilateral hippocampal CD36 expression to one-half of vehicle (0.51  0.09; P < 0.05 by Bonferroni-corrected posttest) and that this was reversed by simultaneous exposure to AH 6809. Indeed, two-way analysis of variance had P < 0.01 for an interaction term between PIC and AH 6809, and Bonferroni-corrected

posttests were not significant for comparison of PIC with PIC plus 200 ng of AH 6809. Similar results were achieved when using 50 ng (0.86  0.21) rather than 200 ng of AH 6809 (0.81  0.22).

Discussion Considerable data from observational studies of human brain and experimental models support the hypothesis that microglial innate immune activation is a feature and possible contributor to AD pathogenesis, a hypothesis recently reinforced by genetic association of AD dementia with several genes that regulate innate immunity, especially TREM2. Results from these studies are further buttressed by compelling epidemiologic and experimental model data that support inhibition of the PG cascade as an effective therapeutic approach early in the pathogenesis of AD before clinical expression or behavioral impairments in transgenic mouse models. Enthusiasm over these observational and experimental data are tempered by failed trials that used NSAIDs to suppress innate immunity in subjects with mild cognitive impairment or AD dementia and the termination of an AD prevention trial because of toxicity. Although the clinical trial results clearly are disappointing, they do not necessarily indict the innate immunity hypothesis for AD but rather may reflect limitations of intervening too late in the course of AD and/or therapeutic shortcomings of proximal inhibition of the PG cascade in people. Together, these results cautiously animate pursuit of components distal in the PG cascade as a potentially more effective means of suppressing microglial innate immunity while averting unwanted side effects. Previously, with the use of primary mouse microglia, we have shown that suppression of EP2 signaling leads to a therapeutically desirable dual phenotype of reduced innate Figure 4

Regulation of mouse primary microglia phagocytosis of Ab42 by prostaglandin EP2 receptor signaling. After exposure to agents, phagocytosis was determined by incubation with pHrodo-labeled Ab42 for 2 hours at 37 C. Fluorescence intensities were determined by flow cytometry, and geometric means of fluorescence intensity were normalized to vehicleexposed WT cells. A: WT microglia were pretreated with 20 mmol/L EP2 agonist butaprost, 20 mg/mL TLR3 activator PIC, or 20 mg/mL PIC plus 20 mmol/L EP2 antagonist AH 6809 for 24 hours and then incubated with pHrodo-labeled Ab42. Cultures treated with PIC þ AH 6809 were also incubated with 500 mmol/L CD36 inhibitor, SSO, for an additional hour before phagocytosis assay. B: WT microglia were treated with 20 mmol/L AH 6809 or 20 mmol/L agonist butaprost for 24 hours, followed by incubation with 10 mg/mL anti-CD36 IgA or mouse control IgA for 1 hour, and then phagocytosis was determined. C: Primary microglia from WT or CD36/ mice were treated with 20 mmol/L EP2 antagonist AH 6809 or 20 mmol/L agonist butaprost for 24 hours, and then phagocytosis was determined. Data are expressed as means  SEM. n Z 3 per group (A and C); n Z 3 or 4 per group (B). *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001 for Bonferroni-corrected repeated paired comparisons with vehicle; y P < 0.05 and yyP < 0.01 for Bonferroni-corrected paired comparisons between two experimental groups. Ab42, b-amyloid 1-42; EP2, E2 receptor subtype 2; PIC, polyinosinic-polycytidylic acid; SSO, sulfosuccinimidyl oleate; TLR, Toll-like receptor; WT, wild-type.

The American Journal of Pathology

-

ajp.amjpathol.org

235

CD36 mRNA (fold of Veh)

Li et al Vehicle PIC 1.00 0.75

** 0.50 0.25 0.00 No AH 6809

Plus AH 6809

Figure 5 Modulation of CD36 expression in mouse hippocampus by PIC and prostaglandin E2 receptor subtype 2 antagonist AH 6809. Wild-type mice received intracerebroventricular injection into the left lateral ventricle with vehicle, 50 ng PIC, 200 ng AH 6809, or 50 ng PIC plus 200 ng AH 6809. Ipsilateral hippocampus was isolated after 24 hours, mRNA values of CD36 were determined by quantitative real-time PCR, and results were normalized to endogenous control GAPDH. Data are expressed as means  SEM. n Z 6 mice per group. Two-way analysis of variance (df 1,1,1, 20) had **P < 0.01 for PIC and interaction between PIC and AH 6809 but was insignificant for AH 6809 alone. Bonferroni-corrected posttests were significant only for the pair with no AH 6809 (**P < 0.01) but were insignificant for the pair with AH 6809. PIC, polyinosinic-polycytidylic acid; Veh, vehicle.

immune-mediated neurotoxicity and enhanced Ab phagocytosis, including neuritic plaque material.28 Subsequently, we and others have validated these findings in macrophages and in vivo in mouse brain by using homozygous deficient mice, conditional deletion mice, and bone marrow transplant mice. Although we have made some progress in understanding the molecular mechanisms of EP2 regulation of microglia-mediated neurotoxicity, the mechanisms by which EP2 signaling regulates Ab42 phagocytosis remained unclear. Here, we showed that scavenger receptor CD36, a key regulator of microglia Ab42 phagocytosis, is closely regulated by EP2 signaling in primary WT mouse microglia both without further activation or in the context of TLR activators. We focused our work on primary microglia because they express the majority of CD36 among phagocytosis-competent cells in the central nervous system. Primary WT microglial expression of CD36 was increased by an EP2 antagonist and decreased by an EP2 agonist; the latter was associated with proportionately reduced CD36 cell surface protein and Ab42 phagocytosis. We showed that specific TLR activators led to decreased microglia CD36 transcription and that this translated into reduced cell surface CD36 protein and reduced Ab42 phagocytosis. At each step in this process, we showed that the effects of innate immune activation on CD36 could be reversed by at least half by suppressing EP2 signaling and that the effects of EP2 activation on Ab42 phagocytosis are largely mediated by CD36. Together, our results from primary microglia cultures indicated tonic control of CD36 expression by EP2 signaling, and that most innate immune-mediated suppression of Ab42 phagocytosis depended on EP2, and that this EP2 effect was CD36 dependent.

236

Current data support reduced clearance as the dominant mechanism for Ab accumulation in the most common forms of AD (reviewed in Wildsmith et al54). Although Ab may be transported out of brain, it also may be cleared by microglial phagocytosis. Indeed, several lines of evidence point to Ab phagocytosis as a potentially important process in AD. These include the increased genetic risk of AD with TREM2 variants, reduced expression of the phagocytosis-related protein beclin 1 in diseased brain regions and isolated microglia from patients with AD,55,56 and reduced CD36 expression on leukocytes in patients with AD dementia or mild cognitive impairment.57 It is important to note that, although we have focused on its phagocytic function, the CD36/Ab complex is pleiotropic and interacts with several intracellular molecules, such as p130Cas, Pyk2, and Paxillin, and initiates signaling pathways that ultimately lead to F-actin polymerization and cytoskeleton reorganization.58 Our results from mouse microglia confirmed those from other cell types that have shown decreased expression of CD36 after TLR3 or TLR4 activation.59,60 We expanded this survey to include TLR7, activation of which by an miRNA can propagate neurodegeneration.61 Our data also showed that the innate immune-mediated decrease in CD36 was mostly, but not fully, reversed by an EP2 antagonist, suggesting that some other proinflammatory factor(s) also inhibit microglial CD36 expression. One possibility is tumor necrosis factor-a,62,63 although this is complicated because tumor necrosis factor-a secretion depends in part on EP2.38 Our data also suggest EP4 as another possibility. However, we were limited in pursuing this further because of lack of a selective EP4 antagonist. In contrast to innate immune activation effects on CD36 expression, a variety of antiinflammatory mediators increase CD36 expression, including IL-4, IL-13, and peroxisome proliferator-activated receptor g agonist.64e67 Blocking CD36 with an antibody reduces cytokine and nitric oxide when microglia are incubated with PrP protein.68 Although difficult to classify broadly as either proinflammatory or anti-inflammatory, we found that an agonist of EP1 or TP also increased CD36 expression in microglia. Functional antagonism is observed commonly among eicosanoid receptors.21 Our results point to a dominant effect of EP2-mediated suppression of CD36 expression in the context of microglial innate immune activation, perhaps related to its higher binding affinity for PGE2 than for EP1.21 Our in vivo experiments showed that CD36 expression in WT mouse hippocampus followed the same mechanisms as microglia. Specifically, activation of innate immune response with PIC significantly decreased CD36 expression, although the magnitude was somewhat less than that observed in primary microglia. The reasons for this difference in magnitude of effect by PIC in vivo versus in vitro are not clear from our experiments but may be related to the influence of other cell types and clearance of the inflammagen. Regardless, our results with a highly selective activator of TLR3 are consistent with those from AD

ajp.amjpathol.org

-

The American Journal of Pathology

EP2-Suppressed Phagocytosis via CD36 transgenic mice that also observed reduced CD36 expression in the brain,63 a finding confounded by the multiple actions of Ab peptides. Most importantly, coadministration of AH 6809 largely reversed the effect of PIC, so much so that there was no significant difference between hippocampal CD36 mRNA values in the PIC alone group versus the PIC plus AH 6809 group. We did not attempt to duplicate behavioral experiments in mice because several other laboratories already have investigated CD36 in the context of mouse behavior and models of immune activation or AD. Indeed, CD36deficient mice show significant learning impairment as assessed by radial arm maze.69 In contrast, increased CD36 expression by peroxisome proliferator-activated receptor g/ retinoid X receptor a agonists increased microglial Ab phagocytosis and improved cognitive performance in an AD transgenic mouse.67 Similarly, increased CD36 expression after brain microinjection of IL-4/IL-13 into another AD transgenic mouse was associated with increased Ab clearance and improved cognitive performance.66

4.

5.

6.

7.

8.

9.

Conclusions In summary, our data are consistent with an innate immune hypothesis for AD in which age, inherited deficits, or environmental factors impair the clearance of Ab peptides either by reduced transport and/or by reduced phagocytosis. This promotes accumulation of Ab peptides and other debris that function as endogenous ligands of TLRs and other pattern recognition receptors that activate an innate immune response; this in turn further lowers phagocytic activity. Our results are consistent with others in supporting a key role for CD36 in Ab phagocytosis by microglia and highlight EP2 antagonists as an effective means to suppress this toxic reinforcing cycle. Combined with results of others, our data suggest that EP2 antagonists have the potential as a therapeutic approach to suppressing key aspects of AD pathogenesis.

10.

11.

12.

13.

Acknowledgments We thank Samantha Rice for assistance with animal husbandry and Aimee Schantz and Carol Arnold for managerial support. EP2/ mice were a gift from Dr. Richard Breyer (Vanderbilt University, Nashville, TN).

14.

15.

References 1. Galasko D, Montine TJ: Biomarkers of oxidative damage and inflammation in Alzheimer’s disease. Biomark Med 2010, 4:27e36 2. Lambert JC, Ibrahim-Verbaas CA, Harold D, Naj AC, Sims R, Bellenguez C, et al: Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet 2013, 45: 1452e1458 3. Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, Cruchaga C, Sassi C, Kauwe JS, Younkin S, Hazrati L,

The American Journal of Pathology

-

ajp.amjpathol.org

16.

17.

Collinge J, Pocock J, Lashley T, Williams J, Lambert JC, Amouyel P, Goate A, Rademakers R, Morgan K, Powell J, St George-Hyslop P, Singleton A, Hardy J; Alzheimer Genetic Analysis Group: TREM2 variants in Alzheimer’s disease. N Engl J Med 2013, 368:117e127 Jonsson T, Stefansson H, Steinberg S, Jonsdottir I, Jonsson PV, Snaedal J, Bjornsson S, Huttenlocher J, Levey AI, Lah JJ, Rujescu D, Hampel H, Giegling I, Andreassen OA, Engedal K, Ulstein I, Djurovic S, Ibrahim-Verbaas C, Hofman A, Ikram MA, van Duijn CM, Thorsteinsdottir U, Kong A, Stefansson K: Variant of TREM2 associated with the risk of Alzheimer’s disease. N Engl J Med 2013, 368:107e116 Hsieh CL, Koike M, Spusta SC, Niemi EC, Yenari M, Nakamura MC, Seaman WE: A role for TREM2 ligands in the phagocytosis of apoptotic neuronal cells by microglia. J Neurochem 2009, 109:1144e1156 Forabosco P, Ramasamy A, Trabzuni D, Walker R, Smith C, Bras J, Levine AP, Hardy J, Pocock JM, Guerreiro R, Weale ME, Ryten M: Insights into TREM2 biology by network analysis of human brain gene expression data. Neurobiol Aging 2013, 34:2699e2714 Sieber MW, Jaenisch N, Brehm M, Guenther M, Linnartz-Gerlach B, Neumann H, Witte OW, Frahm C: Attenuated inflammatory response in triggering receptor expressed on myeloid cells 2 (TREM2) knockout mice following stroke. PLoS One 2013, 8:e52982 Ito H, Hamerman JA: TREM-2, triggering receptor expressed on myeloid cell-2, negatively regulates TLR responses in dendritic cells. Eur J Immunol 2012, 42:176e185 Sun M, Zhu M, Chen K, Nie X, Deng Q, Hazlett LD, Wu Y, Li M, Wu M, Huang X: TREM-2 promotes host resistance against Pseudomonas aeruginosa infection by suppressing corneal inflammation via a PI3K/Akt signaling pathway. Invest Ophthalmol Vis Sci 2013, 54:3451e3462 Lim GP, Yang F, Chu T, Chen P, Beech W, Teter B, Tran T, Ubeda O, Ashe KH, Frautschy SA, Cole GM: Ibuprofen suppresses plaque pathology and inflammation in a mouse model for Alzheimer’s disease. J Neurosci 2000, 20:5709e5714 Szekely CA, Zandi PP: Non-steroidal anti-inflammatory drugs and Alzheimer’s disease: the epidemiological evidence. CNS Neurol Disord Drug Targets 2010, 9:132e139 Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM, Iwatsubo T, Jack CR Jr, Kaye J, Montine TJ, Park DC, Reiman EM, Rowe CC, Siemers E, Stern Y, Yaffe K, Carrillo MC, Thies B, Morrison-Bogorad M, Wagster MV, Phelps CH: Toward defining the preclinical stages of Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement 2011, 7:280e292 Aisen PS, Schafer KA, Grundman M, Pfeiffer E, Sano M, Davis KL, Farlow MR, Jin S, Thomas RG, Thal LJ; Alzheimer’s Disease Cooperative Study: Effects of rofecoxib or naproxen vs placebo on Alzheimer disease progression: a randomized controlled trial. JAMA 2003, 289:2819e2826 Thal LJ, Ferris SH, Kirby L, Block GA, Lines CR, Yuen E, Assaid C, Nessly ML, Norman BA, Baranak CC, Reines SA; Rofecoxib Protocol 078 study group: A randomized, double-blind, study of rofecoxib in patients with mild cognitive impairment. Neuropsychopharmacology 2005, 30:1204e1215 ADAPT Research Group, Martin BK, Szekely C, Brandt J, Piantadosi S, Breitner JC, Craft S, Evans D, Green R, Mullan M: Cognitive function over time in the Alzheimer’s Disease Anti-inflammatory Prevention Trial (ADAPT): results of a randomized, controlled trial of naproxen and celecoxib. Arch Neurol 2008, 65:896e905 Hoozemans JJ, Veerhuis R, Rozemuller JM, Eikelenboom P: Soothing the inflamed brain: effect of non-steroidal anti-inflammatory drugs on Alzheimer’s disease pathology. CNS Neurol Disord Drug Targets 2011, 10:57e67 Jaturapatporn D, Isaac MG, McCleery J, Tabet N: Aspirin, steroidal and non-steroidal anti-inflammatory drugs for the treatment of Alzheimer’s disease. Cochrane Database Syst Rev 2012, 2:Cd006378

237

Li et al 18. Montine TJ, Sonnen JA, Milne G, Baker LD, Breitner JC: Elevated ratio of urinary metabolites of thromboxane and prostacyclin is associated with adverse cardiovascular events in ADAPT. PLoS One 2010, 5:e9340 19. Babiloni C, Frisoni GB, Del Percio C, Zanetti O, Bonomini C, Cassetta E, Pasqualetti P, Miniussi C, De Rosas M, Valenzano A, Cibelli G, Eusebi F, Rossini PM: Ibuprofen treatment modifies cortical sources of EEG rhythms in mild Alzheimer’s disease. Clin Neurophysiol 2009, 120:709e718 20. Breitner JC, Baker LD, Montine TJ, Meinert CL, Lyketsos CG, Ashe KH, Brandt J, Craft S, Evans DE, Green RC, Ismail MS, Martin BK, Mullan MJ, Sabbagh M, Tariot PN; ADAPT Research Group: Extended results of the Alzheimer’s disease anti-inflammatory prevention trial. Alzheimers Dement 2011, 7:402e411 21. Cimino PJ, Keene CD, Breyer RM, Montine KS, Montine TJ: Therapeutic targets in prostaglandin E2 signaling for neurologic disease. Curr Med Chem 2008, 15:1863e1869 22. Shi J, Wang Q, Johansson JU, Liang X, Woodling NS, Priyam P, Loui TM, Merchant M, Breyer RM, Montine TJ, Andreasson K: Inflammatory prostaglandin E2 signaling in a mouse model of Alzheimer disease. Ann Neurol 2012, 72:788e798 23. Andreasson K: Emerging roles of PGE2 receptors in models of neurological disease. Prostaglandins Other Lipid Mediat 2010, 91: 104e112 24. Montine TJ, Sidell KR, Crews BC, Markesbery WR, Marnett LJ, Roberts LJ 2nd, Morrow JD: Elevated CSF prostaglandin E2 levels in patients with probable AD. Neurology 1999, 53:1495e1498 25. Combrinck M, Williams J, De Berardinis MA, Warden D, Puopolo M, Smith AD, Minghetti L: Levels of CSF prostaglandin E2, cognitive decline, and survival in Alzheimer’s disease. J Neurol Neurosurg Psychiatry 2006, 77:85e88 26. Akitake Y, Nakatani Y, Kamei D, Hosokawa M, Akatsu H, Uematsu S, Akira S, Kudo I, Hara S, Takahashi M: Microsomal prostaglandin E synthase-1 is induced in alzheimer’s disease and its deletion mitigates alzheimer’s disease-like pathology in a mouse model. J Neurosci Res 2013, 91:909e919 27. Shie FS, Montine KS, Breyer RM, Montine TJ: Microglial EP2 is critical to neurotoxicity from activated cerebral innate immunity. Glia 2005, 52:70e77 28. Shie FS, Breyer RM, Montine TJ: Microglia lacking E Prostanoid Receptor subtype 2 have enhanced Abeta phagocytosis yet lack Abeta-activated neurotoxicity. Am J Pathol 2005, 166:1163e1172 29. Cimino PJ, Sokal I, Leverenz J, Fukui Y, Montine TJ: DOCK2 is a microglial specific regulator of central nervous system innate immunity found in normal and Alzheimer’s disease brain. Am J Pathol 2009, 175:1622e1630 30. Keene CD, Chang RC, Lopez-Yglesias AH, Shalloway BR, Sokal I, Li X, Reed PJ, Keene LM, Montine KS, Breyer RM, Rockhill JK, Montine TJ: Suppressed accumulation of cerebral amyloid {beta} peptides in aged transgenic Alzheimer’s disease mice by transplantation with wild-type or prostaglandin E2 receptor subtype 2-null bone marrow. Am J Pathol 2010, 177:346e354 31. Johansson JU, Pradhan S, Lokteva LA, Woodling NS, Ko N, Brown HD, Wang Q, Loh C, Cekanaviciute E, Buckwalter M, Manning-Bog AB, Andreasson KI: Suppression of inflammation with conditional deletion of the prostaglandin E2 EP2 receptor in macrophages and brain microglia. J Neurosci 2013, 33: 16016e16032 32. Silverstein RL, Febbraio M: CD36, a scavenger receptor involved in immunity, metabolism, angiogenesis, and behavior. Sci Signal 2009, 2:re3 33. Sheedy FJ, Grebe A, Rayner KJ, Kalantari P, Ramkhelawon B, Carpenter SB, Becker CE, Ediriweera HN, Mullick AE, Golenbock DT, Stuart LM, Latz E, Fitzgerald KA, Moore KJ: CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nat Immunol 2013, 14:812e820

238

34. Coraci IS, Husemann J, Berman JW, Hulette C, Dufour JH, Campanella GK, Luster AD, Silverstein SC, El-Khoury JB: CD36, a class B scavenger receptor, is expressed on microglia in Alzheimer’s disease brains and can mediate production of reactive oxygen species in response to beta-amyloid fibrils. Am J Pathol 2002, 160:101e112 35. Park L, Wang G, Zhou P, Zhou J, Pitstick R, Previti ML, Younkin L, Younkin SG, Van Nostrand WE, Cho S, Anrather J, Carlson GA, Iadecola C: Scavenger receptor CD36 is essential for the cerebrovascular oxidative stress and neurovascular dysfunction induced by amyloid-beta. Proc Natl Acad Sci U S A 2011, 108:5063e5068 36. El Khoury JB, Moore KJ, Means TK, Leung J, Terada K, Toft M, Freeman MW, Luster AD: CD36 mediates the innate host response to beta-amyloid. J Exp Med 2003, 197:1657e1666 37. Ricciarelli R, D’Abramo C, Zingg JM, Giliberto L, Markesbery W, Azzi A, Marinari UM, Pronzato MA, Tabaton M: CD36 overexpression in human brain correlates with beta-amyloid deposition but not with Alzheimer’s disease. Free Radic Biol Med 2004, 36: 1018e1024 38. Li X, Cudaback E, Keene CD, Breyer RM, Montine TJ: Suppressed microglial E prostanoid receptor 1 signaling selectively reduces tumor necrosis factor alpha and interleukin 6 secretion from toll-like receptor 3 activation. Glia 2011, 59:569e576 39. Li X, Cudaback E, Breyer RM, Montine KS, Keene CD, Montine TJ: Eicosanoid receptor subtype-mediated opposing regulation of TLRstimulated expression of astrocyte glial-derived neurotrophic factor. FASEB J 2012, 26:3075e3083 40. Abramovitz M, Adam M, Boie Y, Carriere M, Denis D, Godbout C, Lamontagne S, Rochette C, Sawyer N, Tremblay NM, Belley M, Gallant M, Dufresne C, Gareau Y, Ruel R, Juteau H, Labelle M, Ouimet N, Metters KM: The utilization of recombinant prostanoid receptors to determine the affinities and selectivities of prostaglandins and related analogs. Biochim Biophys Acta 2000, 1483:285e293 41. Kiriyama M, Ushikubi F, Kobayashi T, Hirata M, Sugimoto Y, Narumiya S: Ligand binding specificities of the eight types and subtypes of the mouse prostanoid receptors expressed in Chinese hamster ovary cells. Br J Pharmacol 1997, 122:217e224 42. Pooler AM, Arjona AA, Lee RK, Wurtman RJ: Prostaglandin E2 regulates amyloid precursor protein expression via the EP2 receptor in cultured rat microglia. Neurosci Lett 2004, 362:127e130 43. Patrizio M, Colucci M, Levi G: Protein kinase C activation reduces microglial cyclic AMP response to prostaglandin E2 by interfering with EP2 receptors. J Neurochem 2000, 74:400e405 44. Wilkinson K, Boyd JD, Glicksman M, Moore KJ, El Khoury J: A high content drug screen identifies ursolic acid as an inhibitor of amyloid beta protein interactions with its receptor CD36. J Biol Chem 2011, 286:34914e34922 45. Kuda O, Pietka TA, Demianova Z, Kudova E, Cvacka J, Kopecky J, Abumrad NA: Sulfo-N-succinimidyl oleate (SSO) inhibits fatty acid uptake and signaling for intracellular calcium via binding CD36 lysine 164: SSO also inhibits oxidized low density lipoprotein uptake by macrophages. J Biol Chem 2013, 288:15547e15555 46. Reed-Geaghan EG, Savage JC, Hise AG, Landreth GE: CD14 and toll-like receptors 2 and 4 are required for fibrillar A{beta}-stimulated microglial activation. J Neurosci 2009, 29:11982e11992 47. Tang SC, Lathia JD, Selvaraj PK, Jo DG, Mughal MR, Cheng A, Siler DA, Markesbery WR, Arumugam TV, Mattson MP: Toll-like receptor-4 mediates neuronal apoptosis induced by amyloid beta-peptide and the membrane lipid peroxidation product 4-hydroxynonenal. Exp Neurol 2008, 213:114e121 48. Udan ML, Ajit D, Crouse NR, Nichols MR: Toll-like receptors 2 and 4 mediate Abeta(1-42) activation of the innate immune response in a human monocytic cell line. J Neurochem 2008, 104:524e533 49. Lotz M, Ebert S, Esselmann H, Iliev AI, Prinz M, Wiazewicz N, Wiltfang J, Gerber J, Nau R: Amyloid beta peptide 1-40 enhances the action of Toll-like receptor-2 and -4 agonists but antagonizes Tolllike receptor-9-induced inflammation in primary mouse microglial cell cultures. J Neurochem 2005, 94:289e298

ajp.amjpathol.org

-

The American Journal of Pathology

EP2-Suppressed Phagocytosis via CD36 50. Coleman RA, Smith WL, Narumiya S: International Union of Pharmacology classification of prostanoid receptors: properties, distribution, and structure of the receptors and their subtypes. Pharmacol Rev 1994, 46:205e229 51. De Bono DP, Lumley P, Been M, Keery R, Ince SE, Woodings DF: Effect of the specific thromboxane receptor blocking drug AH23848 in patients with angina pectoris. Br Heart J 1986, 56:509e517 52. Jiang J, Dingledine R: Prostaglandin receptor EP2 in the crosshairs of anti-inflammation, anti-cancer, and neuroprotection. Trends Pharmacol Sci 2013, 34:413e423 53. Shie FS, Montine KS, Breyer RM, Montine TJ: Microglial EP2 as a new target to increase amyloid beta phagocytosis and decrease amyloid beta-induced damage to neurons. Brain Pathol 2005, 15: 134e138 54. Wildsmith KR, Holley M, Savage JC, Skerrett R, Landreth GE: Evidence for impaired amyloid beta clearance in Alzheimer’s disease. Alzheimers Res Ther 2013, 5:33 55. Pickford F, Masliah E, Britschgi M, Lucin K, Narasimhan R, Jaeger PA, Small S, Spencer B, Rockenstein E, Levine B, WyssCoray T: The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice. J Clin Invest 2008, 118:2190e2199 56. Lucin KM, O’Brien CE, Bieri G, Czirr E, Mosher KI, Abbey RJ, Mastroeni DF, Rogers J, Spencer B, Masliah E, Wyss-Coray T: Microglial beclin 1 regulates retromer trafficking and phagocytosis and is impaired in Alzheimer’s disease. Neuron 2013, 79:873e886 57. Giunta M, Rigamonti AE, Scarpini E, Galimberti D, Bonomo SM, Venturelli E, Muller EE, Cella SG: The leukocyte expression of CD36 is low in patients with Alzheimer’s disease and mild cognitive impairment. Neurobiol Aging 2007, 28:515e518 58. Stuart LM, Bell SA, Stewart CR, Silver JM, Richard J, Goss JL, Tseng AA, Zhang A, El Khoury JB, Moore KJ: CD36 signals to the actin cytoskeleton and regulates microglial migration via a p130Cas complex. J Biol Chem 2007, 282:27392e27401 59. Zamora C, Canto E, Nieto JC, Angels Ortiz M, Juarez C, Vidal S: Functional consequences of CD36 downregulation by TLR signals. Cytokine 2012, 60:257e265 60. Melkamu T, Squillace D, Kita H, O’Grady SM: Regulation of TLR2 expression and function in human airway epithelial cells. J Membr Biol 2009, 229:101e113

The American Journal of Pathology

-

ajp.amjpathol.org

61. Lehmann SM, Kruger C, Park B, Derkow K, Rosenberger K, Baumgart J, Trimbuch T, Eom G, Hinz M, Kaul D, Habbel P, Kalin R, Franzoni E, Rybak A, Nguyen D, Veh R, Ninnemann O, Peters O, Nitsch R, Heppner FL, Golenbock D, Schott E, Ploegh HL, Wulczyn FG, Lehnardt S: An unconventional role for miRNA: let-7 activates Toll-like receptor 7 and causes neurodegeneration. Nat Neurosci 2012, 15:827e835 62. Boyer JF, Balard P, Authier H, Faucon B, Bernad J, Mazieres B, Davignon JL, Cantagrel A, Pipy B, Constantin A: Tumor necrosis factor alpha and adalimumab differentially regulate CD36 expression in human monocytes. Arthritis Res Ther 2007, 9:R22 63. Hickman SE, Allison EK, El Khoury J: Microglial dysfunction and defective beta-amyloid clearance pathways in aging Alzheimer’s disease mice. J Neurosci 2008, 28:8354e8360 64. Feng J, Han J, Pearce SF, Silverstein RL, Gotto AM Jr, Hajjar DP, Nicholson AC: Induction of CD36 expression by oxidized LDL and IL-4 by a common signaling pathway dependent on protein kinase C and PPAR-gamma. J Lipid Res 2000, 41:688e696 65. Berry A, Balard P, Coste A, Olagnier D, Lagane C, Authier H, Benoit-Vical F, Lepert JC, Seguela JP, Magnaval JF, Chambon P, Metzger D, Desvergne B, Wahli W, Auwerx J, Pipy B: IL-13 induces expression of CD36 in human monocytes through PPARgamma activation. Eur J Immunol 2007, 37:1642e1652 66. Kawahara K, Suenobu M, Yoshida A, Koga K, Hyodo A, Ohtsuka H, Kuniyasu A, Tamamaki N, Sugimoto Y, Nakayama H: Intracerebral microinjection of interleukin-4/interleukin-13 reduces beta-amyloid accumulation in the ipsilateral side and improves cognitive deficits in young amyloid precursor protein 23 mice. Neuroscience 2012, 207: 243e260 67. Yamanaka M, Ishikawa T, Griep A, Axt D, Kummer MP, Heneka MT: PPARgamma/RXRalpha-induced and CD36-mediated microglial amyloid-beta phagocytosis results in cognitive improvement in amyloid precursor protein/presenilin 1 mice. J Neurosci 2012, 32:17321e17331 68. Kouadir M, Yang L, Tan R, Shi F, Lu Y, Zhang S, Yin X, Zhou X, Zhao D: CD36 participates in PrP(106-126)-induced activation of microglia. PLoS One 2012, 7:e30756 69. Abumrad NA, Ajmal M, Pothakos K, Robinson JK: CD36 expression and brain function: does CD36 deficiency impact learning ability? Prostaglandins Other Lipid Mediat 2005, 77:77e83

239

Prostaglandin E2 receptor subtype 2 regulation of scavenger receptor CD36 modulates microglial Aβ42 phagocytosis.

Recent studies underline the potential relevance of microglial innate immune activation in Alzheimer disease. Primary mouse microglia that lack prosta...
1MB Sizes 0 Downloads 6 Views