INTRODUCTION
Malaria is a highly prevalent infectious disease caused by parasites of several
Plasmodium spp., the most deadly of which,
Plasmodium falciparum, prevails in Africa. In individuals living in areas where malaria is endemic, it is usually uncomplicated and resolves with time even in the absence of treatment with antimalarial drugs. However, in about 1% of cases, almost exclusively among young children, malaria becomes severe and life threatening, resulting in nearly 700,000 deaths each year in Africa alone (
1). Among the most severe complications of
P. falciparum infection in humans is human cerebral malaria (HCM) with a case fatality rate of 15 to 20% in African children despite effective antimalarial chemotherapy (
2,
3). HCM takes a second toll on African children, leaving survivors at high risk of debilitating neurological defects (
4). At present, we have no effective adjunctive therapies for HCM, and developing such therapies in combination with antimalarial drugs would have a large impact on improving global public health.
Currently, our understanding of the pathogenesis of HCM is far from complete and relies heavily on the analysis of histopathology of brain tissue from children who died from HCM (
5,
6). Although HCM is a clinically heterogeneous disease, the commonly accepted definition of HCM centers around neurological symptoms, ultimately unarousable coma, with the presence of infected red blood cells (iRBCs) in the peripheral circulation system with no other apparent causes of coma (
7). Recently, the correct diagnosis of HCM was greatly improved by the use of retinal exams to identify histological features of HCM, correcting what was estimated to be 25 to 30% misdiagnosed cases (
8). Sequestration of iRBCs on the brain vascular endothelium is a defining feature of HCM (
5). Other common features of the brain histopathology in clinically well-characterized HCM patients include brain microhemorrhages associated with axonal and myelin damage, disruption of the blood-brain barrier (BBB), and brain swelling (
5,
6). Systemic activation of the endothelium has also been reported in HCM patients and appears to correlate with disease severity (
9). HCM is also characterized by the production of high levels of proinflammatory cytokines and chemokines that have been correlated with HCM pathogenesis (
10,
11). The accumulation of both monocytes with phagocytosed hemozoin (
5) and platelets (
12), as well as a small number of intravascular leukocytes, including CD8
+ T cells, has also been observed in brain sections of HCM patients (
5,
12). As both the host immune response and sequestration of iRBCs appear to contribute to the pathogenesis of HCM, successful therapies may be ones that target both the host immune response and the parasite.
The mouse model of CM, experimental CM (ECM), recapitulates many characteristics of HCM and therefore may be a useful tool to identify candidates for adjunctive therapy in the human disease (
13,
14). Infection of susceptible C57BL/6 mice with
Plasmodium berghei ANKA (
PbA) parasites results in death of up to 100% of mice usually within 6 to 14 days postinfection (p.i.) following clear signs of neurological damage, including paralysis, ataxia, convulsions, and coma (
13). Strains of mice resistant to
PbA-induced ECM do not show clinical signs of neurological damage but die 2 to 3 weeks after infection due to anemia caused by hyperparasitemia (
13). Examination of the brains of mice with late-stage ECM show many of the features common to HCM, including accumulation of iRBCs along venular endothelium, microhemorrhages, breakdown of the BBB, and brain swelling (
7,
13). Although the degree of sequestration of iRBCs in the brains in ECM appears in general to be less than that in HCM (
14), the presence of iRBCs in the brains of infected mice has been shown to be necessary for the development of ECM (
15–17). ECM is also associated with a marked accumulation of various immune cells in the brains of infected animals, including T cells, monocytes, neutrophils, and NK cells. In particular, recent data provided evidence that the accumulation of CD8
+ T cells in the brains of infected animals and their production of granzyme B and perforin are required for the development of ECM (
15,
18,
19). In 2013, Howland et al. (
20) provided evidence that parasite-specific CD8
+ T cells interact with parasite antigens cross-presented on major histocompatibility complex (MHC) class I molecules on the brain endothelium in ECM. Recently, Pai et al. (
21) used two-photon intravital microscopy to visualize leukocyte behavior in the brains of
PbA-infected mice during ECM. They showed that monocytes accumulated in the brain 1 or 2 days prior to the onset of neurological symptoms and showed decreased rolling speeds due to activation of the endothelium as disease severity increased. Adoptive transfer experiments showed that the behavior of monocytes was dependent on the recruitment of CD8
+ T cells to the brain. Proinflammatory cytokines also appear to play a critical role in ECM, particularly gamma interferon (IFN-γ), tumor necrosis factor (TNF), and lymphotoxin α (
7). Indeed, it was possible to induce ECM in ECM-resistant BALB/c mice by inducing proinflammatory cytokines by treatment with the Toll-like receptor agonist, CpG, during
PbA infections (
22). Taken together, these results support a model for the pathogenesis of ECM in which infection induces inflammatory cytokines that activate endothelial cells to process and present antigens from iRBCs that accumulated on the activated brain endothelium via MHC class I molecules, marking these cells as targets of parasite-specific CD8
+ T cells (
23).
The evolutionarily conserved serine/threonine kinase mammalian target of rapamycin (mTOR) plays a central role in regulating the outcome of antigen recognition in the adaptive immune system (
24). mTOR functions at a central node of several evolutionarily conserved pathways that regulate stress responses, metabolism, autophagy, and survival. By integrating these pathways with immune cell receptor signaling pathways, mTOR serves to regulate immune responses (
25). Targeting mTOR by rapamycin is proving to be an effective means of suppressing immune responses primarily due to the ability of rapamycin to inhibit effector T cell differentiation and promote regulatory T cell (Treg) differentiation (
25). In addition, rapamycin has been shown to inhibit parasite growth
in vitro through its interaction with the single
Plasmodium falciparum FK506 binding protein
PfFKBP35, and consequently,
PfFKBP35 is considered a promising target for antimalarial drugs (reviewed in reference
26). Although
Plasmodium parasite genes do not encode an mTOR homolog, the mTOR ATP-competitive kinase inhibitors, Torins, have been recently shown to inhibit parasite growth (
27), possibly through their inhibition of parasite phosphoinositide 3 kinases that are members of the mTOR family.
Here we provide evidence that rapamycin treatment administered as late as 4 days p.i. protects mice from ECM. The most striking effect of rapamycin on disease progression was the prevention of the breakdown of the BBB and brain hemorrhaging and the reduction in the numbers of T cells and iRBCs that accumulate in the brain. Rapamycin markedly altered transcriptional profiles in the brains of infected mice, and analysis of these transcriptional changes predicted that rapamycin inhibited leukocyte trafficking to and proliferation in the brain. Remarkably, rapamycin treatment is protective against ECM, despite significantly increasing immune inflammation both peripherally and in the brain. Rapamycin's effect on parasite growth is complex
in vivo, functioning to elevate peripheral parasitemia and decrease parasite loads in the brain. Recent studies suggest that several additional metabolic pathways that are activated in T cells following antigen recognition are also required to direct the resulting response (
25). The results presented here open a new avenue for the development of adjunctive therapies for HCM by targeting metabolic pathways that regulate immune responses and possibly parasite growth.
DISCUSSION
HCM, a common form of severe malaria, imposes a heavy health burden in sub-Saharan Africa in childhood mortality and among survivors in long-term neurological deficits. At present, we have no adjunctive therapies for HCM, and the development of such therapies would benefit greatly from a clearer understanding of the parasite and host mechanisms that underlie the pathology of HCM. Although our understanding of such mechanisms is far from complete, it seems likely that HCM pathology may have multiple causes with contributions from both the parasite and the host, particularly the host's immune response. HCM and ECM in mice share a number of features, including sequestration of iRBCs in the brain microvasculature, breakdown of the BBB, and elevated levels of proinflammatory cytokines (
7,
13). It is well established that in ECM, CD8
+ T cells play a critical role in the pathogenesis of the infection (
13), whereas in HCM, the functions of leukocytes observed in the brain vasculature (
5,
12) remain uncharacterized.
Here we explored the effect of the mTOR inhibitor rapamycin on the progression of ECM in mice.
Figure 10 depicts our current model for the effect of rapamycin on ECM. Rapamycin is an attractive candidate for therapy, as it has proven to be an effective means of suppressing immune responses (
25). Moreover, rapamycin has been shown to inhibit the growth of
P. falciparum in vitro through its binding to the parasite homolog of the mammalian FK506 binding protein (
26). The effectiveness of rapamycin as an immunosuppressant is likely due to its ability to inhibit effector T cell differentiation and to inhibit effector T cell metabolism and thus function (
25). Rapamycin treatment of mice during the first 3 days of infection was recently shown to increase survival in ECM with a concomitant decrease in the accumulation of CD8
+ and CD4
+ T cells in the brain (
38). We observed that treatment with rapamycin as late as day 4 p.i. prevented ECM in mice. Treated mice showed none of the signs of pathology of ECM, including breakdown of the BBB, brain hemorrhaging, and neurological symptoms. Treatment with rapamycin resulted in a dramatic decrease in the number of CD8
+ T cells that accumulated in the brains of infected mice as well as the number of iRBCs in the brain vasculature. CD8
+ T cells have been established to play a critical role in ECM (
13). Recent studies provided evidence that CD8
+ T cells engage parasite-derived peptides presented on MHC class I molecules on brain endothelium and in a perforin-dependent process damage the endothelium (
20). On the basis of these observations, we propose that rapamycin blocks the differentiation of CD8
+ effector T cells in lymphoid organs and their migration to the brain, and in the absence of CD8
+ effector T cells in the brain, ECM does not develop.
Our comparison of the gene transcription profiles of uninfected mice,
PbA-infected mice, and
PbA-infected, rapamycin-treated mice provided several novel insights into the molecular and cellular mechanisms underlying ECM. Perhaps most informative was the analysis of genes that were discordantly regulated in the brain; that is genes that were expressed at higher levels in
PbA-infected mice compared to uninfected mice but at lower levels in
PbA-infected, rapamycin-treated mice compared to untreated mice or vice versa. Analysis of the discordantly regulated genes showed that many such genes in the right cerebrum were involved in networks that regulate cellular chemotaxis and invasion and the proliferation of lymphocytes. Notably,
PbA-induced upregulation of
Gzmb, which encodes granzyme B and plays an essential role in the development of ECM during
PbA infection in C57BL/6 mice (
15), was significantly reversed with rapamycin treatment. These findings provide further evidence linking the recruitment of CD8
+ effector T cells to the brain with neuropathology, which may occur from endothelial damage by CD8
+ T cell-mediated cytotoxicity.
Remarkably, rapamycin treatment protected the brains of
PbA-infected mice despite inducing significant increases in inflammation both peripherally and in the brain. This conclusion was supported by our analyses of both cytokines and chemokines in peripheral blood and changes in gene transcription in the spleen and in the brain. The analyses of cytokines in serum provided evidence for large increases in inflammatory cytokines and a decrease in the anti-inflammatory cytokine IL-10. Comparisons of changes in gene transcription in the brains of
PbA-infected mice compared to uninfected mice and in
PbA-infected, rapamycin-treated mice showed that several inflammatory pathways were upregulated upon infection but even further upregulated upon rapamycin treatment. These findings were unexpected, since many studies have implicated inflammation as an integral aspect of CM pathogenesis. In fact, many features of severe malaria have been considered to be similar to those of sepsis (
39), a condition of overwhelming inflammation. Intriguingly, our transcription analysis predicted that among the inflammatory genes, only genes related to the interferon response would be activated in the brains of rapamycin-treated,
PbA-infected mice. Although downstream mediators of inflammation such as
Tnf,
Il1b, and
Il6 were predicted to be activated in the spleen after rapamycin treatment, similar activation of
Tnf,
Il1b, and
Il6 was not observed at any of the brain sites. These results suggest that the inflammatory cascade in the brains of
PbA-infected mice might be truncated or diminished locally by rapamycin treatment. In support of this,
Hmox1, which is induced in response to oxidative stress and protects against ECM (
37), was unexpectedly downregulated in the brains, but not the spleens, of
PbA-infected mice treated with rapamycin, suggesting that rapamycin protects from ECM by limiting oxidative stress events proximal to the induction of
Hmox1. One could speculate that decreased recruitment of leukocytes to the brain conferred by rapamycin treatment reduces cytotoxic CD8
+ T cell-mediated end-organ damage and therefore limits inflammation and oxidative stress locally. Thus, limited end-organ inflammation could provide a possible explanation for why ECM was not observed in rapamycin-treated mice despite increased systemic inflammation. However, it remains to be seen whether this is a direct effect of rapamycin or simply a consequence of decreased leukocyte recruitment to the brain during
PbA infection. Rapamycin treatment may uncouple the
PbA-induced host inflammatory response, which may not in itself be necessary for the development of ECM, from the CD8
+ T cell-mediated response, which is required for ECM pathogenesis.
The results presented here also show that rapamycin treatment protects against ECM despite significantly increasing peripheral parasitemia. However, of perhaps greater importance, rapamycin treatment reduced parasite sequestration in the brains of infected mice which may be critical to ECM pathogenesis. In children with severe malaria, total parasite biomass, quantified by the serum concentration of
P. falciparum histidine-rich protein 2 (
PfHRP2), was shown to be higher in fatal cases than in nonfatal cases despite both groups having equivalent peripheral parasitemias (
40). Using the plasma concentrations of
PfHRP2 to quantify total, circulating, and sequestered parasite biomass, Cunnington et al. (
41) recently showed that the sequestered biomass tended to be higher in children with HCM than in children with uncomplicated malaria, suggesting that sequestration of parasites in the brain, not total body parasitemia, may be critical to HCM pathogenesis (
11). The mechanism by which rapamycin treatment enhanced parasite growth is also of potential interest. The observation that rapamycin treatment had no effect on parasite growth in RAG [KO] mice lacking an adaptive immune system suggests that rapamycin does not act directly on parasites but rather functions to relieve an immune mechanism that normally controls parasite growth. Understanding the nature of this mechanism may provide new targets for antimalarial drugs.
The demonstration that inhibiting mTOR-controlled metabolic pathways by treatment with rapamycin prevented the development of ECM opens up a new avenue toward developing adjunctive therapies for HCM by targeting the metabolism of the host immune cells. Recent studies suggest that several additional metabolic pathways are activated in T cells upon antigen recognition and are required for directing the resulting response (reviewed in reference
25). These pathways involve the transcription factors MYC, which drives cell growth and apoptosis and regulates glycolytic metabolism, and H1Fα, which regulates metabolism under hypoxic conditions as well as the serine/threonine kinase 5′ AMP-activated protein kinase (AMPK), which senses AMP/ATP ratios in cells to regulate cellular functions. Each of these pathways has critical and selective roles in defining T cell function and fate. Depending on the immune mechanisms at play in CM, inhibitors of these pathways may be more effective than rapamycin in controlling disease. For example, treatment with rapamycin inhibits the generation of effector CD8
+ T cells, requiring that rapamycin be administered before day 5 p.i., a time when the clinical symptoms of ECM are generally not apparent. It may be that inhibitors of metabolic pathways that are required for continued effector functions of T cells already present in the brains during CM could be delivered much later when neurological symptoms appear. Although best studied in T cells, metabolic pathways controlled by mTOR regulate diverse immune cell types that may play roles in HCM, including B cells, NK cells, neutrophils, and mast cells. Searches for inhibitors of cellular metabolism that block critical late immune cell function in CM may provide highly effective adjunctive therapies for HCM.
MATERIALS AND METHODS
Ethics statement.
All experiments were approved by the National Institute of Allergy and Infectious Diseases Animal Care and Use Committee.
Animals and malaria infections.
C57BL/6 and C57BL/6-[KO]RAG-1 female mice (7 to 10 weeks old) were obtained from the Jackson Laboratory. Mice were infected with either
PbA or
PbNK65 (New York line) by injecting 1 × 10
6 PbA- or
PbNK65-iRBCs obtained from infected C57BL/6 mice intraperitoneally (i.p.). Hemoglobin levels in blood samples taken from the tail vein (<10 µl/day) were determined using a HemoCue Hb201+ (HemoCue AB, Angelholm, Sweden). Peripheral parasitemia was determined in blood either by Wright-Giemsa-stained whole-blood smears or by flow cytometry as described below. Infected mice were monitored for the progression of experimental cerebral malaria (ECM) using a 10-point clinical scoring system that rates mice as symptomless (a score of 0), to moribund (a score of 10), as previously described (
42). According to our animal protocol, mice with a clinical score of 6 or greater and severely anemic mice with a hemoglobin level below 2.5 g/dl were euthanized.
Rapamycin and artesunate treatment.
For rapamycin treatment, a stock solution of rapamycin (catalog no. R0395; Sigma Aldrich) was prepared by dissolving rapamycin in pure ethanol (25 mg/ml). For treatment of mice, which weighed approximately 20 g in these studies, the stock rapamycin solution was diluted in a solution of 5% polyethylene glycol 400 (Sigma), 4% ethanol, and 5% Tween 80 for a final concentration of rapamycin of 1 mg/ml. Mice were injected intraperitoneally with 1 mg/kg rapamycin every day starting on day 1, 4, or 5 p.i., unless otherwise noted. For artesunate treatment, 60 mg of artesunate (catalog no. A3731; Sigma) was dissolved in 1 ml of 5% sodium bicarbonate, to which 4 ml of 5% dextrose was added for a final concentration of 12 mg/ml. One hundred milligrams per kilogram was administered i.p. to mice on the specified days.
Flow cytometry of brain and spleen leukocytes.
Mice were anesthetized with ketamine/xylazine 6 days p.i. and transcardially perfused with ice-cold PBS, and the brains and spleens were removed. The brains were dissected, minced, and digested with 1 mg/ml collagenase D for 30 min at 37°C. After the tissue was passed through 70-µm nylon mesh, homogenates were placed on a 90%−60%−40% discontinuous Percoll gradient and centrifuged for 18 min at 1,000 ×
g, and the cells at the 40%−60% interface containing mostly leukocytes were collected for analysis. The spleens were minced and forced through a 70-µm nylon mesh, and the cell suspension was incubated for 10 min in a solution of ammonium-chloride-potassium (ACK; Lonza) to lyse RBCs. The cells were washed and resuspended in RPMI 1640 with 10% heat-inactivated fetal bovine serum (FBS). The cells from both brain and spleen were stained in fluorescence-activated cell sorting (FACS) buffer (PBS plus 1% FBS). The following fluorescent-dye-conjugated antibodies specific for the following cell surface markers were used for staining: brilliant violet 421-conjugated NK1.1 (BV421–NK1.1) (BioLegend), BV605–CD4 (BioLegend), BV785–CD8 (BioLegend), phycoerythrin-conjugated Ly6G (PE–Ly6G) (BD Pharmingen), phycoerythrin-and-Cy7-conjugated CD3 (PE-Cy7–CD3) (eBioscience), allophycocyanin-conjugated Ly6C (APC–Ly6C) (BD Pharmingen), Alexa Fluor 700-conjugated CD44 (AF700–CD44) (eBioscience), APC-Cy7–CD45.2 (BD Pharmingen), and LIVE/DEAD (Aqua; Invitrogen). Gating of subsets is depicted in
Fig. S1 in the supplemental material. Cell acquisition data were obtained on a BD LSRII flow cytometer. Data were analyzed with FlowJo software (Tree Star Technologies).
Assessment of BBB integrity.
Evans blue (20 mg/kg) was injected intravenously on day 6 p.i., and 3 h later, the mice were anesthetized and perfused, and the brains were removed and immediately frozen at −80°C for later processing. EB was quantified by a modified version of the previously described protocol (
43). Briefly, EB was extracted from brains with one perfused brain per 2-ml skirted screw cap tube (Greiner). Seven hundred microliters of
N,
N-dimethylformamide (DMF) (catalog no. D4551; Sigma) was added with three silica beads (2.3 mm) (catalog no. 11079125z; Biospec) per tube and homogenized for 1 min at room temperature (Minibeadbeater-16 model 607; BioSpec Products). This homogenized solution was centrifuged at 16,000 ×
g for 20 min at 4°C. The supernatant was transferred to a separate tube and spun again at the same speed and temperature for 10 min. Two hundred microliters of this supernatant was then quantified in duplicate using a Varioskan Flash fluorometer (620-nm excitation; 695-nm emission; Thermo Scientific). For quantification, a standard curve was generated by using a uninfected perfused brain prepared in the same way and adding EB at a twofold dilution starting at 1 mg/ml to 1 µg/ml.
Quantification of peripheral blood parasitemia by flow cytometry.
Parasitemia was determined by flow cytometry using a modified version of a previously described method (
44). Briefly, blood (approximately 0.6 µl) was obtained from mouse tail veins, fixed with 0.025% aqueous glutaraldehyde solution, washed with 2 ml PBS, resuspended, and stained with the following: the DNA dye Hoechst 33342 (Sigma) (8 µM), the DNA and RNA dye dihydroethidium (diHEt) (10 µg/ml), an APC-conjugated antibody (Ab) specific for CD45.2 (BioLegend), a pan-C57BL/6 lymphocyte marker, and APC-Cy7-conjugated Ab specific for Ter119 (BD Pharmingen), an RBC marker. The cells were analyzed on a BD LSRII flow cytometer equipped with UV (325-nm), violet (407-nm), blue (488-nm), and red (633-nm) lasers. Data were analyzed using FlowJo software (Tree Star Technologies). iRBCs were CD45.2
−, Ter119
+, Hoechst positive, and diHEt
+. Overall parasitemia was calculated as the number of iRBCs/total number of RBCs.
Quantification of parasites localized in the brain.
After anesthetization and intracardiac perfusion of mice as described above, the brains were removed, immediately frozen in liquid nitrogen, and pulverized, and RNA was extracted using a Qiagen RNeasy minikit according to the manufacturer's instructions. Genomic DNA was digested on a column using a RNase-free DNase set (Qiagen), and the elimination of genomic DNA was further confirmed using no reverse transcriptase (no-RT) controls. cDNA was generated using a iScript cDNA synthesis kit (Bio-Rad Laboratories). SYBR green PCR master mix (Bio-Rad) was used to determine the relative expression of parasite 18S rRNA and of three host housekeeping genes, hprt, gapdh, and ppia. The Pb-18S primers and primer sequences were 5′-AAGCATTAAATAAAGCGAATACATCCTTAC-3′ and 5′-GGAGATTGGTTTTGACGTTTATGTG-3′. The mouse hprt, gapdh, and ppia primer sequences were 5′-TGCTCGAGATGTGATGAAGG-3′ and 5′-TCCCCTGTTGACTGGTCATT-3′, 5′-GTGGAGTCATACTGGAACATGTAG-3′ and 5′-AATGGTGAAGGTCGGTGTG-3′, and 5′-TTCACCTTCCCAAAGACCAC-3′ and 5′-CAAACACAAACGGTTCCCAG-3′, respectively.
The geometric means of the threshold cycle (CT) values of housekeeping genes were used as the baseline for comparing the ΔCT value of 18S gene amplification. The changes in gene expression were calculated by comparing the ΔCT values of experimental and control groups using the comparative CT method (2−ΔΔCT).
Brain histology.
Brain samples were fixed in 10% buffered formalin, embedded in paraffin, and sectioned. Sections were stained with hematoxylin and eosin (H&E) for ultrastructural examination and detection of hemorrhages and iRBC hemozoin. For detection of IgG, the slides were sequentially incubated in citrate for 20 min, 2% normal horse serum for 20 min, and biotinylated horse Ab specific for mouse IgG (Vector Labs, Burlingame, CA) for 30 min. Biotinylated horse Ab was detected using a biotin avidin peroxidase complex kit (Vector Labs). The slides were then examined by light microscopy with magnifications between ×5 and ×100, and the microscopic images were evaluated by two independent investigator in a blind manner (blind to the study design) as described previously (
45). For quantitation of hemorrhages, 10 microscopic 40× power fields were examined, and the numbers of hemorrhages were counted and averaged.
Cytokine measurements.
Blood samples were collected on day 6 p.i., and sera were stored at −80°C until analyzed for IL-6, IL-10, IL-12p70, MIP-1α, MCP-1, RANTES, IFN-γ, tumor necrosis factor alpha (TNF-α), IL-1β, CXCL1, TARC, and TCA using the Q-Plex array mouse cytokine kit (Quansys Biosciences) according to the manufacturer's instructions.
Microarray chip processing and data analysis.
C57BL/6 mice were infected with PbA or mock infected with saline vehicle and treated with either saline or rapamycin beginning on day 1 p.i. (4 conditions; 4 mice for each condition). On day 6 p.i., the mice were anesthetized and perfused with saline, and samples from 4 tissues (spleen, right cerebrum, cerebellum, and olfactory bulb; 64 samples total) were immediately frozen in liquid nitrogen. RNA was isolated. For each sample, labeled target was combined with 2× hybridization buffer, 3 nM B2 control oligonucleotide (catalog no. 900457; Affymetrix), 20× hybridization control stock (Affymetrix), and dimethyl sulfoxide (DMSO) making a final volume of 150 µl for the individual hybridizations to the Affymetrix GeneChip mouse gene 2.0 ST array containing the C57BL/6 mouse genome. The hybridization cocktail, including the components listed above, was denatured for 5 min at 99°C and then transferred to a 45°C heat block for an additional 5 min before transferring 130 µl of the cocktail onto the chip. The hybridization was carried out at a constant temperature of 45°C for approximately 40 h using an Affymetrix 640 hybridization oven. Upon completion of the hybridization step, each sample was removed from the chip and archived. Each chip was filled with approximately 160 µl of wash buffer A and then processed on the fluidics station 450. The reagents for the stain mixture consisted of 2× morpholineethanesulfonic acid (MES) stain buffer, 50 mg/ml of bovine serum albumin (BSA), 1 mg/ml of streptavidin phycoerythrin and water to make up a total volume of 600 µl for each stain. A holding buffer was added to make up a total volume of 800 µl for storage and scanning. Upon completion of the fluidics process, each sample was scanned using the Affymetrix GeneChip 3000 7Gplus scanner, and an expression console (Affymetrix version 1.3) was used to convert the data files to intensity (cel) files. The quality analysis was performed according to the “Quality Assessment of Exon and Gene Arrays” (Affymetrix revision 1.1). cel files representing individual samples were normalized using robust multiarray average (RMA) normalization followed by median normalization. Filtering was performed to remove any probe with mean log2 expression of all samples below 5.0 or log2 standard deviation of all samples below 1. Sample quality control was performed using principal component analysis (PCA) and sample-wise density plots in R. No outliers were identified in any of the aforementioned quality control methods used.
An empirical Bayes modified three-way repeated measure analysis of variance (ANOVA) was computed between the different treatment conditions using the limma package library in R to obtain false discovery rate (FDR)-adjusted P values and fold changes. The values for the probes were considered statistically significant if their FDR-adjusted P values were <0.05 and their absolute fold change was >1.5 except where otherwise noted. Gene symbols, log fold change ratios, P values, and false discovery rates from the empirical Bayes ANOVA were imported into Ingenuity Pathway Analysis (IPA) (Qiagen) to determine pathway enrichment scores and perform upstream regulator and regulator effect analyses. Network diagrams were exported from IPA, and heatmaps were generated with the pheatmap package and gplots libraries in R.
Statistical analysis.
Statistical analyses of nonmicroarray data were computed using the latest versions of GraphPad Prism 6. Most comparisons are unpaired Mann-Whitney tests with Bonferroni's adjustments for multiple comparisons applied when appropriate. Student's
t test was computed for the qPCR data to allow for easier fold change calculation. One-way ANOVA with Tukey posthoc adjustments for multiple comparisons was used for the log
10-transformed cell count data in
Fig. 4. All survival curves are Kaplan-Meier curves with any log rank tests for any comparisons among curves.