Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Aug 17.
Published in final edited form as: Pharmacogenomics J. 2008 Oct 28;9(2):116–126. doi: 10.1038/tpj.2008.15

The brain expression of genes involved in inflammatory response, the ribosome, and learning and memory is altered by centrally injected lipopolysaccharide in mice

Robert H Bonow 1,#, Saba Aïd 1,#, Yongqing Zhang 2, Kevin G Becker 2, Francesca Bosetti 1,*
PMCID: PMC2728029  NIHMSID: NIHMS74627  PMID: 18957951

Abstract

Neuroinflammation plays a role in the progression of several neurodegenerative disorders. We used a lipopolysaccharide (LPS) model of neuroinflammation to characterize the gene expression changes underlying the inflammatory and behavioral effects of neuroinflammation. A single intracerebroventricular injection of LPS (5 µg) was administered into the lateral ventricle of mice and, 24 hours later, we examined gene expression in the cerebral cortex and hippocampus using microarray technology. Gene Ontology (GO) terms for inflammation and the ribosome were significantly enriched by LPS, whereas GO terms associated with learning and memory had decreased expression. We detected 224 changed transcripts in the cerebral cortex and 170 in the hippocampus. Expression of Egr1 (also known as Zif268) and Arc, two genes associated with learning and memory, was significantly lower in the cortex, but not hippocampus, of LPS-treated animals. Overall, altered expression of these genes may underlie some of the inflammatory and behavioral effects of neuroinflammation.

Keywords: LPS, neuroinflammation, microarray, Arc, Egr1, cerebral cortex, hippocampus

Introduction

Inflammation plays an important role in the body’s defense against infection, but over-activation of the inflammatory cascade can have detrimental effects. In the central nervous system, inflammation is thought to be associated with normal aging1 and to contribute to the progression of a number of pathological conditions, including Alzheimer’s disease2, Parkinson’s disease3, and amyotrophic lateral sclerosis4.

Lipopolysaccharide (LPS) is a component of the outer membrane of Gram negative bacteria, and is commonly used to induce inflammation in the periphery and in the central nervous system. It stimulates the innate immune system through interactions with CD14 and toll-like receptor 4 (TLR4)5, 6, which are expressed by microglia, the primary immunocompetent cells of the brain. Upon exposure to LPS, microglia become activated and begin producing pro-inflammatory mediators such as cytokines, chemokines, prostanoids, and reactive oxygen species7.

While LPS is not directly toxic to mature neurons, which do not express TLR48, neurons can be indirectly damaged by LPS-activated microglia. A number of studies in vitro have shown that exposing primary neuron/glia mixed cultures to LPS leads to neuronal death, whereas no significant cell loss is observed in neuron-enriched cultures9, 10. Acute administration of LPS directly to the brain recapitulates these results, and leads to neuron death in the hippocampus11. Moreover, behavioral studies have shown that object recognition12 and spatial memory13 are both impaired in animals treated with single doses of LPS. In line with these cognitive deficits, long-term potentiation (LTP) is largely blocked following acute LPS injections12, 14.

In an effort to characterize the transcriptional changes that underlie these physiological and behavioral effects, we examined changes in gene expression in the mouse hippocampus and cerebral cortex 24 hours after a single intracerebroventricular (i.c.v.) injection of LPS using microarray technology.

Results

We began our analysis by filtering the data to include only probes with a reliable signal. A total of 9,557 annotated genes, RIKEN sequences, and expressed sequence tags passed the filtration step. Next, the raw intensity values from this dataset were normalized with a Z-Transformation15. The original data file and the filtered, normalized results are available online in the Gene Expression Omnibus (GEO Accession GSE12284). Using the calculated Z-Scores, we conducted a principal component analysis to reduce the dimensionality of the data and to enable visualization of the relationships between gene expression profiles, as shown in Figure 1a. The first principal component (PC1) separated the samples by brain region, and the second (PC2) did so on the basis of treatment. The first and second principal components accounted for 53.2% and 16.1% of total variance, respectively, indicating that brain region and treatment are responsible for the majority of the differences in gene expression between the samples. To quantify the expression changes for each probe, we compared the Z-Scores between the vehicle and LPS treatment groups using the Z-Ratio statistic15. Positive Z-Ratios indicate increased expression relative to control, and negative Z-Ratios indicate reduced expression. A plot comparing hippocampus and cortex Z-Ratios revealed a significant correlation between the changes in each brain region (r2 = 0.42, P < 0.0001; Figure 1b).

Figure 1.

Figure 1

Global gene expression patterns. A) Principal component analysis separates the individual samples into four distinct groups using each sample’s gene expression profile. The hippocampus and cortex show unique patterns of gene expression at baseline (vehicle) and in response to LPS. The first principal component (PC1) separates samples by brain region, and the second (PC2) does so on the basis of treatment. Each point represents one independent tissue sample; eight animals (LPS, n = 4; vehicle, n = 4) are represented by two points each, one for cortex and one for hippocampus. B) LPS injection causes similar transcriptional changes in both brain regions. The changes in expression for all 9,557 transcripts that passed the filtration step are plotted as Z-Ratios for both hippocampus and cerebral cortex. There is a significant correlation between regions (r2 = 0.411, P < 0.0001).

To provide a functional interpretation of these effects, we conducted a gene set analysis with the PAGE algorithm using Gene Ontology (GO) terms and curated Molecular Signatures Database (MSigDB) gene sets1618. GO terms are gene lists categorized by cellular localization, functional roles, and biological processes16 that are not necessarily co-regulated. By contrast, curated MSigDB gene sets are lists of genes that have been shown to be commonly affected in human diseases or by various experimental conditions17. Our results revealed that a number of pro-inflammatory gene sets were enriched in both regions following LPS administration (Table 1). Other gene sets with increased expression included GO Terms for ribosomal proteins and actin cytoskeletal reorganization, as well as an MSigDB set previously shown to be increased in the aged mouse neocortex19. A number of GO Terms associated with nervous system function showed significantly reduced expression in animals treated with LPS. In addition, we found that LPS caused the downregulation of a set of genes that has been shown to have decreased expression in the hippocampus of patients with Alzheimer’s disease20.

Table 1. Gene sets affected by LPS.

Selected gene sets affected by LPS. We used the Parametric Analysis of Gene Set Enrichment (PAGE) algorithm to find Gene Ontology (GO) terms and curated Molecular Signatures Database (MSigDB) gene sets whose expression was altered during acute neuroinflammation. Significant changes were observed for a number of gene sets associated with inflammation and nervous system function in both brain regions. The PAGE Z-Score reflects the size of a gene set and difference between the average Z-Ratio within the set and the average Z-Ratio for the entire array; Z-Scores with a magnitude greater than ± 1.96 are considered statistically significant with P < 0.05.

PAGE Z-Score

Gene Set Size on Array Hippocampus Cortex
Inflammatory Response1 77 14.88 12.28
Chemokine Activity1 22 9.96 7.00
Leukocyte Chemotaxis1 7 7.96 6.71
Cytokine Activity1 55 7.93 5.06
Ribosome1 124 8.58 4.08
Aged Mouse Neocortex, Up2 45 4.53 5.45
Neutrophil Chemotaxis1 9 6.27 3.43
Actin Cytoskeleton Reorganization1 7 4.76 3.56
Alzheimer's Disease, Down2 672 −4.29 0.31
Learning and or Memory1 19 −2.33 −2.26
Synaptic Transmission1 89 −2.83 −2.27
Neuron Migration1 31 −2.88 −3.43
1

Gene Ontology Term

2

Curated MSigDB Gene Set

We next sought to identify individual genes that were affected by LPS. Using strict criteria, including a Z-Ratio cutoff of ± 1.5 and a P-value < 0.05, we detected significant changes for 170 and 224 transcripts in the hippocampus and cerebral cortex, respectively (Table 2 and Table 3). The complete lists of affected genes are available as supplemental data (Tables S1 and S2). Sixty-four were significantly changed in both brain regions, and all of these were changed in the same direction on the microarray.

Table 2. Genes affected by LPS in hippocampus.

Forty-seven of the 170 genes that were significantly affected in the hippocampus. Many of the upregulated genes are involved in either immune or stress responses, whereas the list of downregulated genes is populated by transcripts that are involved in generalized nervous system function or learning and memory. These 47 were chosen based on the relevance of their biological roles; the complete list is available as supplemental data.

Z-Ratio Symbol GenBank Accession Biological Role
Inflammatory Response
23.59   Saa3 NM_011315 Monocyte recruitment
22.35   Lcn2 NM_008491.1 Antimicrobial activity
13.22   Ifitm3 NM_025378.1 Induced by interferon signaling
13.21   Ifitm1 NM_026820.2 Induced by interferon signaling
13.16   C3 NM_009778.1 Complement system
9.39   Ccl12 NM_011331.1 Chemotaxis
8.38   Ly6a NM_010738.2 Lymphocyte proliferation
8.28   B2m NM_009735.2 MHC antigen presentation
5.49   H2-Q2 NM_010392 MHC antigen presentation
4.88   Emr1 NM_010130.1 Generation of Treg cells
4.46   Stat1 NM_009283.2 JAK/STAT signaling
3.73   Vcam1 NM_011693.2 Lymphocyte and leukocyte migration
3.22   Tnfrsf1a NM_011609.2 TNF receptor
3.10   Psme1 NM_011189.1 Antigen processing
3.01   Stat3 NM_011486.2 JAK/STAT signaling
2.60   Clu NM_013492.1 Tissue protection during inflammation
2.57   Slp NM_011413 Complement system
Stress Response
9.80   Mt1 NM_013602.2 Metal detoxification and antioxidant defense
7.45   Apod NM_007470.1 Antioxidant defense
6.92   Mt2 NM_008630.1 Metal detoxification and antioxidant defense
6.00   S100a6 NM_011313.1 Stress response
5.06   Rbm3 NM_016809 RNA binding; stress response
3.82   Hspb1 NM_013560.1 Stress response
2.95   Gpx1 NM_008160.1 Antioxidant defense
Cytoskeleton
4.95   Vim NM_011701.3 Component of cytoskeleton
Lipid Metabolism
2.92   Gpd1 NM_010271.2 Glycerophospholipid metabolism
2.24   Pnpla2 NM_025802.1 Lipid metabolism
Ribosome
2.74   Rps18 NM_011296.1 Ribosomal protein
2.62   Rps5 NM_009095 Ribosomal protein
2.48   Rpl37a NM_009084.2 Ribosomal protein
2.31   Rpl15 NM_025586.1 Ribosomal protein
Learning, Memory, and Neurotransmission
2.40   Grina NM_023168.2 Glutamate binding; NMDA receptor
2.36   Rasgrf1 NM_011245.1 Learning and memory
General Nervous System
2.56   Ptpre NM_011212.2 Antagonism of Kv channels
2.32   Hpca NM_010471.2 Neuroprotective, Ca2+ binding
2.02   Ctsb NM_007798.1 CNS maintenance

Inflammatory Response
−4.54   Selpl NM_009151.2 Lymphocyte tethering & rolling
Stress Response
−2.01   Adcy5 XM_358801.1 Regulates stress resistance
Learning, Memory, and Neurotransmission
−1.59   Grm5 XM_149971.3 Glu receptor, metabotropic
−1.84   Arc NM_018790.1 Learning and memory
−1.84   Nrxn3 NM_172544.1 Synaptic function
−2.08   Slc6a9 NM_008135.1 Glycine neurotransmission
−2.50   Gabra1 NM_010250.2 GABA neurotransmission
−3.73   Ptn NM_008973.1 Modulation of LTP threshold
General Nervous System
−1.82   Neurod2 NM_010895 Neural development
−2.53   Ephb1 NM_173447.2 Neurogenesis & precursor migration
−2.77   Cntn1 NM_007727.1 Axon guidance

Table 3. Genes affected by LPS in cerebral cortex.

Forty-nine of the 224 genes that were significantly affected in the cerebral cortex. As in the hippocampus, the list of transcripts whose expression was increased by LPS includes mostly genes that are involved in either the immune or stress responses, while many of those with reduced expression are involved in nervous system function or learning and memory. These 49 genes were chosen on the basis of their biological roles; the complete list is available as supplemental data.

Z-Ratio Symbol GenBank Accession Biological Role
Inflammatory Response
21.57   Saa3 NM_011315 Monocyte recruitment
19.05   Lcn2 NM_008491.1 Antimicrobial activity
14.38   Ifitm3 NM_025378.1 Induced by interferon signaling
13.73   Ifitm1 NM_026820.2 Induced by interferon signaling
13.13   Ccl12 NM_011331.1 Chemotaxis
10.04   Ly6a NM_010738.2 Lymphocyte proliferation
9.87   B2m NM_009735.2 MHC antigen presentation
5.89   H2-Q2 NM_010392 MHC antigen presentation
3.20   Psme1 NM_011189.1 Antigen processing
3.07   Vcam1 NM_011693.2 Lymphocyte and leukocyte migration
2.53   Ptk2b NM_172498.1 Leukocyte migration
1.96   Lgmn NM_011175.1 Lysosomal processing
1.89   C1qg NM_007574.1 Complement system
Stress Response
8.88   Mt1 NM_013602.2 Metal detoxification and antioxidant defense
6.00   Apod NM_007470.1 Antioxidant defense
5.86   Mt2 NM_008630.1 Metal detoxification and antioxidant defense
5.75   Rbm3 NM_016809 RNA binding; stress response
4.79   S100a6 NM_011313.1 Stress response
3.47   Hspb8 NM_030704.1 Stress response
2.08   Gpx1 NM_008160.1 Antioxidant defense
Cytoskeleton
5.47   Vim NM_011701.3 Component of cytoskeleton
3.17   Vil2 NM_009510.1 Regulation of cytoskeleton
Lipid Metabolism
2.53   Dbi NM_007830.2 Lipid binding
2.14   Pnpla2 NM_025802.1 Lipid metabolism
Ribosome
2.18   Rps27l NM_026467.1 Ribosomal protein
1.93   Rpl39 NM_026055.1 Ribosomal protein
1.61   Rpl3 NM_013762 Ribosomal protein
1.57   Rps25 NM_024266 Ribosomal protein
1.51   Rpl7a NM_013721 Ribosomal protein

Inflammatory Response
−2.53   Sema4a NM_013658.2 T cell priming & Th1/Th2 response
−4.51   Dusp1 NM_013642.1 Inhibits macrophage inflammatory response
−4.64   Selpl NM_009151.2 Lymphocyte tethering & rolling
Stress Response
−3.03   Hsp105 NM_013559.1 Stress response
Lipid Metabolism
−4.74   Ppap2b NM_080555.1 Lipid metabolism
Learning, Memory, and Neurotransmission
−1.59   Grm5 XM_149971.3 Glu receptor, metabotropic
−1.80   Adcy8 NM_009623.1 Neural plasticity
−1.81   Prkaca NM_008854 Learning and memory
−2.11   Ptn NM_008973.1 Modulates LTP threshold
−2.13   Grasp NM_019518.2 Metabotropic glu receptor trafficking
−2.30   Slc6a9 NM_008135.1 Glycine neurotransmission
−2.39   Cckbr NM_007627.2 DA receptor sensitivity
−2.80   Ntsr2 NM_008747 Fear memory
−5.27   Egr1 NM_007913.2 Learning and memory
−5.92   Arc NM_018790.1 Learning and memory
General Nervous System
−2.35   Mtap1b NM_008634.1 Neurite outgrowth
−2.45   Ephb1 NM_173447.2 Neurogenesis & precursor migration
−2.47   Olig1 NM_016968.2 Oligodendrocyte differentiation
−2.85   Cx3cl1 NM_009142.2 Neuroprotection
−3.15   Plp NM_011123 Protein component of myelin

A number of these individually significant genes recapitulate the results of our gene set analysis; genes encoding ribosomal proteins, acute phase proteins, and stress response proteins showed significantly increased expression, whereas those associated with nervous system function, and particularly neurotransmission, learning, and memory, showed significantly reduced expression in both brain regions. Of particular interest are the genes for Activity regulated cytoskeletal-associated protein (Arc) and Early growth response 1 (Egr1, also known as Zif268), which are intimately involved in learning and memory21, 22. The microarray data indicated that Arc expression was significantly reduced in both the hippocampus (Z-Ratio = −1.84) and cerebral cortex (Z-Ratio = −5.92), whereas Egr1 was affected only in the cortex (Z-Ratio = −5.27).

We then validated our microarray results with real-time PCR using primers and probes for a subset of the genes that were found to be individually significant. These genes were chosen based on their biological relevance and for their wide range of positive and negative Z-Ratios. The expression changes for 77% of the selected genes from the cortex were confirmed by PCR results, and 54% were reproducible in the hippocampus (Table 4). A plot of fold change as assessed by PCR versus microarray Z-Ratio revealed a strongly linear relationship for both brain regions (Figure 2a, b). However, one gene, Cxcl12, was shown by PCR to be increased by 34% in the hippocampus despite having a negative Z-Ratio (Z-Ratio = −3.54). PCR showed a significant reduction in Arc mRNA following LPS in the cortex, but there was no statistically significant difference in the hippocampus (Figure 3a, b). Similarly, Egr1 expression was significantly reduced in the cortex, but not in the hippocampus (Figure 3c, d).

Table 4. Genes validated with real-time PCR.

Results of microarray validation with real-time PCR. We chose to validate a total of fifteen genes, eight of which were affected in both cerebral cortex and hippocampus. LPS-induced gene expression changes determined by real-time PCR are presented as fold-change versus the vehicle group. NS indicates that no significant expression change was observed on the microarray; we did not conduct real-time PCR reactions for the majority of these genes (ND).

Cerebral Cortex Hippocampus


Symbol GenBank Accession Z-Ratio PCR FC Z-Ratio PCR FC
Saa3 NM_011315 21.57 178.27*** 23.59 312.48***
Ccl12 NM_011331 13.13 13.05*** 9.39 10.11***
Apod NM_007470.1 6.00 2.48*** 7.45 3.27***
Gpx1 NM_008160.1 2.08 1.24*** 2.95 1.65***
Grm5 XM_149971.3 −1.59 1.01 −1.58 −1.10
Srf NM_020493 −1.66 −1.05 NS ND
Adcy8 NM_009623.1 −1.80 −1.16*** NS ND
Prkaca NM_008854 −1.81 1.01 NS ND
Neurod2 NM_010895 NS ND −1.82 −1.06
Gabra1 NM_010250.2 NS ND −2.50 −1.04
Ephb1 NM_173447.2 −2.45 −1.31*** −2.53 −1.41**
Mtap1b NM_008634.1 −2.91 −1.13** NS ND
Egr1 NM_007913.2 −5.27 −1.58* NS 1.09
Arc NM_018790.1 −5.92 −1.63** −1.84 −1.27
Cxcl12 NM_013655.2 −6.76 −1.62* −3.53 1.35*
*

P < 0.05

**

P < 0.01

***

P < 0.001.

Figure 2.

Figure 2

Quantitative real-time PCR validation of the microarray data. a–b) Gene expression changes measured by real-time PCR correlate strongly with changes found by microarray analysis in both cerebral cortex (r2 = 0.949, P < 0.0001) and hippocampus (r2 = 0.955, P < 0.0001). Because the Z-Ratio statistic incorporates log-normalized Z-Scores, the logarithm of the fold change as assessed by PCR must be used in calculating the linear regression. The strong correlation between PCR results and microarray data indicate that the expression changes observed using the microarray are real and reproducible with more sensitive methods.

Figure 3.

Figure 3

Expression changes for Arc and Egr1 as determined by Quantitative real-time PCR. a–d) Real-time PCR confirmed significant downregulation of Arc and Egr1 transcripts in the cerebral cortex of LPS-treated mice (n = 8) when compared to vehicle control (n = 7), but no significant change was found in the hippocampus. *P < 0.05, **P < 0.01.

Discussion

The goal of this study was to characterize the changes in gene expression that occur in the mouse hippocampus and cerebral cortex in response to direct activation of innate immunity by centrally injected LPS. While previous studies have described the effects of peripheral LPS administration on the whole mouse brain transcriptome23, 24, and one has described the gene expression changes that occur in the hippocampus after a central injection of LPS25, ours is the first to compare the gene expression profiles of these two brain regions during acute neuroinflammation. Our results reveal that while the two brain regions have unique gene expression profiles at baseline, the expression changes induced by LPS are similar in both hippocampus and cerebral cortex. This finding suggests that, on the whole, both regions respond similarly to acute neuroinflammation.

A functional interpretation of these responses was provided by surveying the effects of LPS on predefined sets of genes from the Gene Ontology (GO) Project and Molecular Signatures Database16, 17. Several GO terms associated with inflammation, including Inflammatory Response, Chemokine Activity, Cytokine Activity, Neutrophil Chemotaxis, and Leukocyte Chemotaxis were highly upregulated in both hippocampus and cortex following LPS treatment. In both regions we also detected significant enrichment of a set of genes that was previously shown to have increased expression in the aged mouse neocortex19, further supporting the concept that brain inflammation increases during normal aging1. Overall, the results of the gene set analysis in the hippocampus and cortex were strikingly similar, and are indicative of a marked inflammatory response.

Expression of mRNA encoding ribosomal proteins was also increased in both regions at the level of gene sets and individual genes, which suggests that the brain is working to increase its capacity for protein synthesis. In support of this observation, a previous study in rats reported that chronic infusion of LPS caused ultrastructural changes in neurons consistent with impaired cytosolic protein synthesis. The authors also noted deep invaginations in nuclear membranes that were filled with polyribosomes, which they interpreted as an attempt by neurons to compensate for impaired cytosolic translation26. The increased transcription of ribosomal proteins that we observed would support this conclusion. However, since activated microglia are known to contain endoplasmic reticulum rich in ribosomes26, 27, it is possible that the increase we found is at least in part due to transcriptional changes that occur in microglia as they become activated. Further work will be required to determine the individual contributions of neurons and glia to this effect.

The GO terms Learning and/or Memory, Synaptic Transmission, and Neuron Migration were decreased in both hippocampus and cerebral cortex, which suggests that neural dysfunction occurs in both regions as a consequence of acute neuroinflammation. Indeed, LPS-induced neuroinflammation has been shown to adversely affect neurogenesis, neurite outgrowth, and LTP12, 2830. These are important processes in the adult nervous system, and their disruption likely contributes to the impaired cognitive performance of rodents treated with LPS.

The Ephrin receptor B1 (EphB1) is involved in the differentiation and migration of neural precursors in the adult hippocampus and in the morphogenesis of neurites and dendritic spines31, 32. We observed significantly reduced expression of this transcript in both brain regions (cerebral cortex: Z-Ratio = −2.45, PCR fold change = −1.31; hippocampus: Z-Ratio = −2.53, PCR fold change = −1.41), and this may contribute to the impaired neurogenesis and to the reduced number or dendritic spines observed during neuroinflammation28, 30, 33.

Despite the overall similarities in transcriptional changes in hippocampus and cerebral cortex, the plasticity-associated immediate-early genes Arc and Egr1 were found to be decreased only in the cerebral cortex upon PCR validation. Arc protein has been shown to regulate actin polymerization and AMPA receptor trafficking at synapses, and Egr1 is a transcription factor that regulates the expression of a number of plasticity-associated target genes, including Arc21, 3437. Both genes are rapidly transcribed postsynaptically following stimuli that produce behavioral learning or LTP, and transgenic animals lacking either gene exhibit impaired memory and weakened LTP21, 22, 3840. Given the importance of Arc and Egr1 in memory, it is plausible that their reduced expression underlies the memory deficits observed in neuroinflammation. In support of this concept, an antisense-mediated knockdown of Arc protein translation has been shown to impair the maintenance of LTP, but not to affect its induction41. Hennigan and colleagues (2007) obtained similar results using an acute peripheral injection of LPS to induce neuroinflammation; the authors observed a significant impairment in LTP maintenance, but no change in LTP induction12. In both studies, the deficits in LTP were evident within one hour of induction. Reduced Egr1 expression may also contribute to inflammation-induced memory impairment; both LPS-treated rats and Egr1−/− mice perform similarly on tests of object recognition and fail to show preference for novel objects 24 hours after training12, 38. These striking similarities suggest that impaired Arc and Egr1 expression may contribute to the memory problems that occur as a consequence of neuroinflammation. However, because immediate-early genes such as Arc and Egr1 can serve as markers for neural activity21, 40, 42, it is also plausible that the reduction we observed is a consequence of diminished neural activity in the cerebral cortex. Indeed, a study by Vereker et al. (2000), using the same paradigm as Hennigan but with twice the dose of LPS, characterized significant deficits in both the induction and maintenance of LTP, which they attributed to lowered glutamate release14. These results point to decreased excitatory neurotransmission, which would also produce the reductions in Arc and Egr1 transcription that we observed. Since these models of neuroinflammation used peripheral injections of LPS in rats and we used centrally injected LPS in mice, it is difficult to directly compare the dosing regimens; further studies will be required to determine if glutamate release is affected in our model as well. A possible effect of the anesthetic used before euthanizing the animals should also be considered, as general anesthesia has been shown to interfere with the expression of neuronal immediate-early genes4346. Additional work should be conducted to address this possibility.

Rosi and colleagues previously linked chronic inflammation to increased Arc expression in the dorsal rat hippocampus after chronic i.c.v. infusion of low doses of LPS47. Though the combined in situ hybridization and cell counting technique used in their study is notably different from the microarray and real-time PCR approach that we employed, the contrast between their results and ours suggest that acute and chronic neuroinflammation may have divergent effects on Arc, and possibly Egr1, transcription. Expression of these two genes has been shown to be lowered in association with β-amyloid plaques in AD patients, in animal models of AD, and in aged mice48, 49. Our results indicate that the inflammatory component of Alzheimer’s and normal aging may be responsible for this effect. In line with this hypothesis, treatment with the non-steroidal anti-inflammatory drug indomethacin restores memory performance and LTP in animal models of AD50. However, it is not known whether anti-inflammatory treatment also restores Arc and Egr1 expression to normal levels. Future studies should investigate this possibility.

The results of this study indicate that, on the whole, gene expression profiles of the hippocampus and cerebral cortex are similarly affected by acute neuroinflammation. However, despite the overall similarity between the two brain regions, we observed reduced transcription of plasticity-associated genes in the cerebral cortex, but not hippocampus. The gene expression changes that we have characterized contribute to the understanding of brain inflammation, and may aid in the development of new therapies for diseases with a neuroinflammatory component.

Materials and methods

Animals and Tissue Processing

All animal procedures were conducted under an NICHD-approved animal protocol in accordance with NIH guidelines for the care and use of laboratory animals. Twelve to 15 week-old mice bred on a C57BL/6 and SV129/Ola genetic background were anesthetized with intraperitoneal (i.p.) ketamine (100 mg/kg) and xylazine (10 mg/kg) and placed on a stereotactic table (Kopf Instruments, Tujunga, CA, USA). Using a 33 gauge needle (World Precision Instruments, Sarasota, FL, USA) attached to a microsyringe pump (Stoelting, Wood Dale, IL, USA), sterile phosphate buffered saline (PBS) vehicle (5 µL) or LPS (E. coli serotype 127:B8; 5 µg in 5 µL sterile PBS) was injected into the lateral ventricle (−2.3 mm dorsal/ventral, −1.0 mm lateral, and −0.5 mm anterior/posterior from bregma) at a rate of 1.0 µL/min, as previously described11, 33, 51. Twenty-four hours after surgery, animals were euthanized with an overdose of sodium pentobarbital (100 mg/kg, i.p.) and immediately decapitated. . While other studies have shown a peak in inflammatory markers such as IL-1β or iNOS at 6 hours after systemic LPS injection 52, 53, the dose of LPS used in our study, when injected intracerebroventricularly, produced a peak in most inflammatory markers examined at 24 h in a preliminary time course pilot study. This time point has been shown by us and others to produce significant levels of neuroinflammatory and oxidative stress markers when measured at 24 h after i.c.v. injection of LPS 51, 5456. The hippocampus and cerebral cortex from both hemispheres were rapidly dissected, flash frozen in 2-methylbutane at −60°C, and stored at −80°C. Total RNA was extracted from hippocampus and cortex samples using Qiagen RNeasy Lipid Tissue Mini and Midi kits, respectively (Qiagen, Valencia, CA, USA). RNA quality was assessed by measuring the A260/280 ratio. RNA integrity was verified by visualization of the 28S and 18S ribosomal rRNA bands, with no presence of smear, using a denaturing agarose gel.

Microarray

Four samples from each brain region and treatment group were used. A 0.5 µg aliquot of total RNA from each sample was labeled using the Illumina TotalPrep RNA Amplification Kit (Ambion; Austin, TX, USA). RNA was first converted into single-stranded cDNA using reverse transcription with an oligo-dT primer containing the T7 RNA polymerase promoter, and then the single-stranded cDNA was copied to produce double-stranded cDNA molecules. The double-stranded cDNA was used in an overnight in vitro transcription reaction where single-stranded RNA (cRNA) was generated and labeled by incorporating biotin-16-UTP. A total of 0.75 µg of biotin-labeled cRNA was hybridized for 16 hours to Illumina's Sentrix MouseRef-8 Expression BeadChips (Illumina, San Diego, CA, USA). Each BeadChip has 24,000 well-annotated RefSeq transcripts per sample with approximately 30-fold redundancy. The arrays were washed and blocked. Biotinylated cRNA was detected with streptavidin-Cy3 and quantitated using Illumina's BeadStation 500X Genetic Analysis Systems scanner. The image data was extracted using BeadStudio software (Illumina).

Data Analysis

The expression data were filtered to include only probes with a consistent signal on each chip; the cutoff value was established at P < 0.02. The resulting dataset was next analyzed with DIANE 6.0, a spreadsheet based microarray analysis program. An overview of DIANE can be found online at http://www.grc.nia.nih.gov/branches/rrb/dna/diane_software.pdf. Using DIANE, the background was subtracted from the raw signal intensity of each gene, and the results were normalized with a Z-Score transformation15. Z-normalized data were then analyzed with principal component analysis (PCA). To determine the gene expression changes caused by LPS, Z-Scores for the vehicle and LPS treatment groups were compared using the Z-Ratio statistic15:

ZRatio=ZScoreLPSZScoreVehicleσ[ZScoreLPSZScoreVehicle].

Gene set analysis was performed using the Parametric Analysis of Gene-set Enrichment (PAGE) algorithm18 to detect Gene Ontology (GO) Terms16 and curated Molecular Signatures Database (MSigDB) gene sets17 affected by LPS. Only MSigDB gene sets with more than 30% representation in the filtered dataset were included in our analysis. Expression changes for individual genes were considered significant if they met four criteria: Z-Ratio above 1.5 or below −1.5; false detection rate (FDR)57 of less than 0.30; a P-value statistic for Z-Score replicability below 0.05; and mean background-corrected signal intensity greater than zero.

Quantitative real-time Polymerase Chain Reaction

Microarray results were validated by quantitative real-time PCR (qRT-PCR), as previously described51, 58. In addition to the samples used for the microarray, we added 3 and 4 mice for the vehicle (total n=7) and LPS treatment group (total n=8), respectively. The following TaqMan Gene Expression Assays (Applied Biosystems, Foster City, CA, USA) were used: Adcy8 (Mm00507722_m1), Apod (Mm00431817_m1), Arc (Mm00479619_g1), Ccl12 (Mm01617100_m1), Cxcl12 (Mm00445552_m1), Egr1 (Mm00656724_m1), Ephb1 (Mm00557961_m1), Gabra1 (Mm00439040_m1), Gpx1 (Mm00656767_g1), Grm5 (Mm00690332_m1), Mtap1b (Mm00485261_m1), Neurod2 (Mm00440465_g1), Prkaca (Mm00660092_m1), Saa3 (Mm00441203_m1), and Srf (Mm00491032_m1). Pgk1 (Mm00435617_m1) was shown not to change by comparing threshold cycle (CT) values for both vehicle and LPS treatment groups, and was therefore used as an endogenous control. Statistical analysis was performed by calculating the difference in threshold cycle (ΔCT) between a given target gene and Pgk1, and then by comparing the ΔCT values between vehicle (n = 7) and LPS (n = 8) treatment groups with student’s t test. The relative expression of target genes was computed using the ΔΔCT method59.

Supplementary Material

Table S1
Table S2

Acknowledgements

We thank Drs. Christopher Toscano and Sang-Ho Choi for helpful discussion and Dr. Marcel Van Der Brug for statistical help. This work was entirely supported by the Intramural Research Program of the National Institutes of Health, National Institute on Aging.

Footnotes

Duality of interest

The authors declare no duality of interest.

References

  • 1.Godbout JP, Johnson RW. Age and neuroinflammation: a lifetime of psychoneuroimmune consequences. Neurol Clin. 2006;24(3):521–538. doi: 10.1016/j.ncl.2006.03.010. [DOI] [PubMed] [Google Scholar]
  • 2.Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, et al. Inflammation and Alzheimer's disease. Neurobiol Aging. 2000;21(3):383–421. doi: 10.1016/s0197-4580(00)00124-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hartmann A, Hunot S, Hirsch EC. Inflammation and dopaminergic neuronal loss in Parkinson's disease: a complex matter. Exp Neurol. 2003;184(2):561–564. doi: 10.1016/j.expneurol.2003.08.004. [DOI] [PubMed] [Google Scholar]
  • 4.Boillee S, Vande Velde C, Cleveland DW. ALS: a disease of motor neurons and their nonneuronal neighbors. Neuron. 2006;52(1):39–59. doi: 10.1016/j.neuron.2006.09.018. [DOI] [PubMed] [Google Scholar]
  • 5.Ingalls RR, Heine H, Lien E, Yoshimura A, Golenbock D. Lipopolysaccharide recognition, CD14, and lipopolysaccharide receptors. Infect Dis Clin North Am. 1999;13(2):341–353. doi: 10.1016/s0891-5520(05)70078-7. vii. [DOI] [PubMed] [Google Scholar]
  • 6.Lien E, Means TK, Heine H, Yoshimura A, Kusumoto S, Fukase K, et al. Toll-like receptor 4 imparts ligand-specific recognition of bacterial lipopolysaccharide. J Clin Invest. 2000;105(4):497–504. doi: 10.1172/JCI8541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8(1):57–69. doi: 10.1038/nrn2038. [DOI] [PubMed] [Google Scholar]
  • 8.Lehnardt S, Lachance C, Patrizi S, Lefebvre S, Follett PL, Jensen FE, et al. The toll-like receptor TLR4 is necessary for lipopolysaccharide-induced oligodendrocyte injury in the CNS. J Neurosci. 2002;22(7):2478–2486. doi: 10.1523/JNEUROSCI.22-07-02478.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Boje KM, Arora PK. Microglial-produced nitric oxide and reactive nitrogen oxides mediate neuronal cell death. Brain Res. 1992;587(2):250–256. doi: 10.1016/0006-8993(92)91004-x. [DOI] [PubMed] [Google Scholar]
  • 10.Jeohn GH, Kong LY, Wilson B, Hudson P, Hong JS. Synergistic neurotoxic effects of combined treatments with cytokines in murine primary mixed neuron/glia cultures. J Neuroimmunol. 1998;85(1):1–10. doi: 10.1016/s0165-5728(97)00204-x. [DOI] [PubMed] [Google Scholar]
  • 11.Choi SH, Langenbach R, Bosetti F. Genetic deletion or pharmacological inhibition of cyclooxygenase-1 attenuate lipopolysaccharide-induced inflammatory response and brain injury. FASEB J. 2007 doi: 10.1096/fj.07-9411com. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hennigan A, Trotter C, Kelly AM. Lipopolysaccharide impairs long-term potentiation and recognition memory and increases p75NTR expression in the rat dentate gyrus. Brain Res. 2007;1130(1):158–166. doi: 10.1016/j.brainres.2006.10.066. [DOI] [PubMed] [Google Scholar]
  • 13.Shaw KN, Commins S, O'Mara SM. Lipopolysaccharide causes deficits in spatial learning in the watermaze but not in BDNF expression in the rat dentate gyrus. Behav Brain Res. 2001;124(1):47–54. doi: 10.1016/s0166-4328(01)00232-7. [DOI] [PubMed] [Google Scholar]
  • 14.Vereker E, Campbell V, Roche E, McEntee E, Lynch MA. Lipopolysaccharide inhibits long term potentiation in the rat dentate gyrus by activating caspase-1. J Biol Chem. 2000;275(34):26252–26258. doi: 10.1074/jbc.M002226200. [DOI] [PubMed] [Google Scholar]
  • 15.Cheadle C, Vawter MP, Freed WJ, Becker KG. Analysis of microarray data using Z score transformation. J Mol Diagn. 2003;5(2):73–81. doi: 10.1016/S1525-1578(10)60455-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000;25(1):25–29. doi: 10.1038/75556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(43):15545–15550. doi: 10.1073/pnas.0506580102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kim SY, Volsky DJ. PAGE: parametric analysis of gene set enrichment. BMC Bioinformatics. 2005;6:144. doi: 10.1186/1471-2105-6-144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lee CK, Weindruch R, Prolla TA. Gene-expression profile of the ageing brain in mice. Nat Genet. 2000;25(3):294–297. doi: 10.1038/77046. [DOI] [PubMed] [Google Scholar]
  • 20.Blalock EM, Geddes JW, Chen KC, Porter NM, Markesbery WR, Landfield PW. Incipient Alzheimer's disease: microarray correlation analyses reveal major transcriptional and tumor suppressor responses. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(7):2173–2178. doi: 10.1073/pnas.0308512100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Knapska E, Kaczmarek L. A gene for neuronal plasticity in the mammalian brain: Zif268/Egr-1/NGFI-A/Krox-24/TIS8/ZENK? Prog Neurobiol. 2004;74(4):183–211. doi: 10.1016/j.pneurobio.2004.05.007. [DOI] [PubMed] [Google Scholar]
  • 22.Tzingounis AV, Nicoll RA. Arc/Arg3.1: linking gene expression to synaptic plasticity and memory. Neuron. 2006;52(3):403–407. doi: 10.1016/j.neuron.2006.10.016. [DOI] [PubMed] [Google Scholar]
  • 23.Godbout JP, Chen J, Abraham J, Richwine AF, Berg BM, Kelley KW, et al. Exaggerated neuroinflammation and sickness behavior in aged mice following activation of the peripheral innate immune system. FASEB J. 2005;19(10):1329–1331. doi: 10.1096/fj.05-3776fje. [DOI] [PubMed] [Google Scholar]
  • 24.Mastronardi C, Whelan F, Yildiz OA, Hannestad J, Elashoff D, McCann SM, et al. Caspase 1 deficiency reduces inflammation-induced brain transcription. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(17):7205–7210. doi: 10.1073/pnas.0701366104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lund S, Christensen KV, Hedtjarn M, Mortensen AL, Hagberg H, Falsig J, et al. The dynamics of the LPS triggered inflammatory response of murine microglia under different culture and in vivo conditions. J Neuroimmunol. 2006;180(1–2):71–87. doi: 10.1016/j.jneuroim.2006.07.007. [DOI] [PubMed] [Google Scholar]
  • 26.Hauss-Wegrzyniak B, Vannucchi MG, Wenk GL. Behavioral and ultrastructural changes induced by chronic neuroinflammation in young rats. Brain Res. 2000;859(1):157–166. doi: 10.1016/s0006-8993(00)01999-5. [DOI] [PubMed] [Google Scholar]
  • 27.Casamenti F, Prosperi C, Scali C, Giovannelli L, Colivicchi MA, Faussone-Pellegrini MS, et al. Interleukin-1beta activates forebrain glial cells and increases nitric oxide production and cortical glutamate and GABA release in vivo: implications for Alzheimer's disease. Neuroscience. 1999;91(3):831–842. doi: 10.1016/s0306-4522(98)00680-0. [DOI] [PubMed] [Google Scholar]
  • 28.Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O. Inflammation is detrimental for neurogenesis in adult brain. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(23):13632–13637. doi: 10.1073/pnas.2234031100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hauss-Wegrzyniak B, Lynch MA, Vraniak PD, Wenk GL. Chronic brain inflammation results in cell loss in the entorhinal cortex and impaired LTP in perforant path-granule cell synapses. Exp Neurol. 2002;176(2):336–341. doi: 10.1006/exnr.2002.7966. [DOI] [PubMed] [Google Scholar]
  • 30.Monje ML, Toda H, Palmer TD. Inflammatory blockade restores adult hippocampal neurogenesis. Science. 2003;302(5651):1760–1765. doi: 10.1126/science.1088417. [DOI] [PubMed] [Google Scholar]
  • 31.Chumley MJ, Catchpole T, Silvany RE, Kernie SG, Henkemeyer M. EphB receptors regulate stem/progenitor cell proliferation, migration, and polarity during hippocampal neurogenesis. J Neurosci. 2007;27(49):13481–13490. doi: 10.1523/JNEUROSCI.4158-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Henkemeyer M, Itkis OS, Ngo M, Hickmott PW, Ethell IM. Multiple EphB receptor tyrosine kinases shape dendritic spines in the hippocampus. J Cell Biol. 2003;163(6):1313–1326. doi: 10.1083/jcb.200306033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Milatovic D, Zaja-Milatovic S, Montine KS, Horner PJ, Montine TJ. Pharmacologic suppression of neuronal oxidative damage and dendritic degeneration following direct activation of glial innate immunity in mouse cerebrum. Journal of neurochemistry. 2003;87(6):1518–1526. doi: 10.1046/j.1471-4159.2003.02120.x. [DOI] [PubMed] [Google Scholar]
  • 34.Chowdhury S, Shepherd JD, Okuno H, Lyford G, Petralia RS, Plath N, et al. Arc/Arg3.1 interacts with the endocytic machinery to regulate AMPA receptor trafficking. Neuron. 2006;52(3):445–459. doi: 10.1016/j.neuron.2006.08.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Messaoudi E, Kanhema T, Soule J, Tiron A, Dagyte G, da Silva B, et al. Sustained Arc/Arg3.1 synthesis controls long-term potentiation consolidation through regulation of local actin polymerization in the dentate gyrus in vivo. J Neurosci. 2007;27(39):10445–10455. doi: 10.1523/JNEUROSCI.2883-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.James AB, Conway AM, Morris BJ. Genomic profiling of the neuronal target genes of the plasticity-related transcription factor -- Zif268. Journal of neurochemistry. 2005;95(3):796–810. doi: 10.1111/j.1471-4159.2005.03400.x. [DOI] [PubMed] [Google Scholar]
  • 37.Li L, Carter J, Gao X, Whitehead J, Tourtellotte WG. The neuroplasticity-associated arc gene is a direct transcriptional target of early growth response (Egr) transcription factors. Mol Cell Biol. 2005;25(23):10286–10300. doi: 10.1128/MCB.25.23.10286-10300.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Jones MW, Errington ML, French PJ, Fine A, Bliss TV, Garel S, et al. A requirement for the immediate early gene Zif268 in the expression of late LTP and long-term memories. Nat Neurosci. 2001;4(3):289–296. doi: 10.1038/85138. [DOI] [PubMed] [Google Scholar]
  • 39.Plath N, Ohana O, Dammermann B, Errington ML, Schmitz D, Gross C, et al. Arc/Arg3.1 is essential for the consolidation of synaptic plasticity and memories. Neuron. 2006;52(3):437–444. doi: 10.1016/j.neuron.2006.08.024. [DOI] [PubMed] [Google Scholar]
  • 40.Worley PF, Christy BA, Nakabeppu Y, Bhat RV, Cole AJ, Baraban JM. Constitutive expression of zif268 in neocortex is regulated by synaptic activity. Proceedings of the National Academy of Sciences of the United States of America. 1991;88(12):5106–5110. doi: 10.1073/pnas.88.12.5106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Guzowski JF, Lyford GL, Stevenson GD, Houston FP, McGaugh JL, Worley PF, et al. Inhibition of activity-dependent arc protein expression in the rat hippocampus impairs the maintenance of long-term potentiation and the consolidation of long-term memory. J Neurosci. 2000;20(11):3993–4001. doi: 10.1523/JNEUROSCI.20-11-03993.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Guzowski JF, McNaughton BL, Barnes CA, Worley PF. Environment-specific expression of the immediate-early gene Arc in hippocampal neuronal ensembles. Nat Neurosci. 1999;2(12):1120–1124. doi: 10.1038/16046. [DOI] [PubMed] [Google Scholar]
  • 43.Douglas RM, Dragunow M, Robertson HA. High-frequency discharge of dentate granule cells, but not long-term potentiation, induces c-fos protein. Brain Res. 1988;464(3):259–262. doi: 10.1016/0169-328x(88)90033-2. [DOI] [PubMed] [Google Scholar]
  • 44.Dragunow M, Abraham WC, Goulding M, Mason SE, Robertson HA, Faull RL. Long-term potentiation and the induction of c-fos mRNA and proteins in the dentate gyrus of unanesthetized rats. Neurosci Lett. 1989;101(3):274–280. doi: 10.1016/0304-3940(89)90545-4. [DOI] [PubMed] [Google Scholar]
  • 45.Jeffery KJ, Abraham WC, Dragunow M, Mason SE. Induction of Fos-like immunoreactivity and the maintenance of long-term potentiation in the dentate gyrus of unanesthetized rats. Brain Res Mol Brain Res. 1990;8(4):267–274. doi: 10.1016/0169-328x(90)90039-g. [DOI] [PubMed] [Google Scholar]
  • 46.Worley PF, Bhat RV, Baraban JM, Erickson CA, McNaughton BL, Barnes CA. Thresholds for synaptic activation of transcription factors in hippocampus: correlation with long-term enhancement. J Neurosci. 1993;13(11):4776–4786. doi: 10.1523/JNEUROSCI.13-11-04776.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Rosi S, Ramirez-Amaya V, Vazdarjanova A, Worley PF, Barnes CA, Wenk GL. Neuroinflammation alters the hippocampal pattern of behaviorally induced Arc expression. J Neurosci. 2005;25(3):723–731. doi: 10.1523/JNEUROSCI.4469-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Blalock EM, Chen KC, Sharrow K, Herman JP, Porter NM, Foster TC, et al. Gene microarrays in hippocampal aging: statistical profiling identifies novel processes correlated with cognitive impairment. J Neurosci. 2003;23(9):3807–3819. doi: 10.1523/JNEUROSCI.23-09-03807.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Dickey CA, Loring JF, Montgomery J, Gordon MN, Eastman PS, Morgan D. Selectively reduced expression of synaptic plasticity-related genes in amyloid precursor protein + presenilin-1 transgenic mice. J Neurosci. 2003;23(12):5219–5226. doi: 10.1523/JNEUROSCI.23-12-05219.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Stephan A, Laroche S, Davis S. Learning deficits and dysfunctional synaptic plasticity induced by aggregated amyloid deposits in the dentate gyrus are rescued by chronic treatment with indomethacin. Eur J Neurosci. 2003;17(9):1921–1927. doi: 10.1046/j.1460-9568.2003.02610.x. [DOI] [PubMed] [Google Scholar]
  • 51.Aid S, Langenbach R, Bosetti F. Neuroinflammatory response to lipopolysaccharide is exacerbated in mice genetically deficient in cyclooxygenase-2. J Neuroinflammation. 2008;5:17. doi: 10.1186/1742-2094-5-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Wong ML, Bongiorno PB, Rettori V, McCann SM, Licinio J. Interleukin (IL) 1beta, IL-1 receptor antagonist, IL-10, and IL-13 gene expression in the central nervous system and anterior pituitary during systemic inflammation: pathophysiological implications. Proc Natl Acad Sci U S A. 1997;94(1):227–232. doi: 10.1073/pnas.94.1.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Wong ML, Rettori V, al-Shekhlee A, Bongiorno PB, Canteros G, McCann SM, et al. Inducible nitric oxide synthase gene expression in the brain during systemic inflammation. Nat Med. 1996;2(5):581–584. doi: 10.1038/nm0596-581. [DOI] [PubMed] [Google Scholar]
  • 54.Choi SH, Langenbach R, Bosetti F. Genetic deletion or pharmacological inhibition of cyclooxygenase-1 attenuate lipopolysaccharide-induced inflammatory response and brain injury. FASEB J. 2008;22(5):1491–1501. doi: 10.1096/fj.07-9411com. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Montine TJ, Milatovic D, Gupta RC, Valyi-Nagy T, Morrow JD, Breyer RM. Neuronal oxidative damage from activated innate immunity is EP2 receptor-dependent. Journal of neurochemistry. 2002;83(2):463–470. doi: 10.1046/j.1471-4159.2002.01157.x. [DOI] [PubMed] [Google Scholar]
  • 56.Goralski KB, Abdulla D, Sinal CJ, Arsenault A, Renton KW. Toll-like receptor-4 regulation of hepatic Cyp3a11 metabolism in a mouse model of LPS-induced CNS inflammation. Am J Physiol Gastrointest Liver Physiol. 2005;289(3):G434–G443. doi: 10.1152/ajpgi.00562.2004. [DOI] [PubMed] [Google Scholar]
  • 57.Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(9):5116–5121. doi: 10.1073/pnas.091062498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Choi SH, Langenbach R, Bosetti F. Cyclooxygenase-1 and -2 enzymes differentially regulate the brain upstream NF-kappa B pathway and downstream enzymes involved in prostaglandin biosynthesis. Journal of neurochemistry. 2006;98(3):801–811. doi: 10.1111/j.1471-4159.2006.03926.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–408. doi: 10.1006/meth.2001.1262. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Table S1
Table S2

RESOURCES