Skip to main content
Dryad

Immune responses in mice lacking STAT-1 in microglia

Cite this dataset

Harris, Tajie; Cowan, Maureen (2022). Immune responses in mice lacking STAT-1 in microglia [Dataset]. Dryad. https://doi.org/10.5061/dryad.fttdz08w2

Abstract

Toxoplasma gondii is a ubiquitous intracellular protozoan parasite that establishes a life-long chronic infection largely restricted to the central nervous system (CNS). Constant immune pressure, notably IFN-γ-STAT1 signaling, is required for preventing fatal pathology during T. gondii infection. Here, we report that abrogation of STAT1 signaling in microglia, the resident immune cells of the CNS, is sufficient to induce a loss of parasite control in the CNS and susceptibility to toxoplasmic encephalitis during the early stages of chronic infection. Using a microglia-specific genetic labeling and targeting system that discriminates microglia from blood-derived myeloid cells that infiltrate the brain during infection, we find that, contrary to previous in vitro reports, microglia do not express inducible nitric-oxide synthase (iNOS) during T. gondii infection in vivo. Instead, transcriptomic analyses of microglia reveal that STAT1 regulates both (i) a transcriptional shift from homeostatic to “disease-associated microglia” (DAM) phenotype conserved across several neuroinflammatory models, including T. gondii infection, and (ii) the expression of anti-parasitic cytosolic molecules that are required for eliminating T. gondii in a cell-intrinsic manner. Further, genetic deletion of Stat1 from microglia during T. gondii challenge leads to fatal pathology despite largely equivalent or enhanced immune effector functions displayed by brain-infiltrating immune populations. Finally, we show that microglial STAT1-deficiency results in the overrepresentation of the highly replicative, lytic tachyzoite form of T. gondii, relative to its quiescent, semi-dormant bradyzoite form typical of chronic CNS infection. Our data suggest an overall protective role of CNS-resident microglia against T. gondii infection, illuminating (i) general mechanisms of CNS-specific immunity to infection (ii) and a clear role for IFN-STAT1 signaling in regulating a microglial activation phenotype observed across diverse neuroinflammatory disease states.

Methods

Mice. CX3CR1CreERT2 (#020940) and ROSA26Ai6/Ai6 (#007906) mouse lines were originally purchased from Jackson Laboratories and cross-bred to generate CX3CR1CreERT2 x ZsGreenfl/stop/fl mice used as controls. These control mice were subsequently cross-bred with STAT1fl/fl mice (provided by Lothar Hennighausen, NIH) to generate STAT1fl/fl x CX3CR1CreERT2 x ROSA26Ai6/Ai6 (MGSTAT1∆) mice. Age- and sex-matched mice were intraperitoneally administered tamoxifen (4 mg per 20 g body weight, Sigma-Aldrich) between 4-7 weeks of age for 5 consecutive days to induce STAT1 deletion. Four weeks following tamoxifen treatment, mice were challenged with the type II T. gondii strain Me49 or proceeded with naïve experiments. Parasite was passaged through CBA/J and Swiss Webster mice (Jackson Laboratories), and mice used in infection experiments were intraperitoneally challenged with 10 Me49 cysts from CBA/J brain homogenate. Stat1 excision was confirmed by qPCR, flow cytometry, or immunohistochemistry. All mice were housed in University of Virginia specific pathogen-free facilities with a 12h light/dark cycle, with ambient temperature between 68 and 72 F, and 30-60% humidity. Mice used in experiments were euthanized by CO2 asphyxiation if they showed weight loss greater than 20% of their baseline, pre-recorded weight. All procedures were approved and conducted in accordance with the University of Virginia Institutional Animal Care and Use Committee approval of protocol 3968.

Sholl Analysis. Sholl analysis was used to analyze microglial morphological complexity as a readout for their activation state, in accordance with the protocol published in Norris et al., 2014 [40]. In brief, confocal photomicrographs of naive WT and MGSTAT1∆ brains with ZsGreen+ microglia were captured and analyzed in Fiji with the Sholl Analysis plugin software. Images were processed in binary pixels, microglia were manually selected, and shells were inserted in 5 µm concentric circles, starting from 10 µm outside the center of the soma, and ending at the limit of the longest arborization for each analyzed microglia. The number of dendritic intersection points was plotted for each distance away from the soma, and data was analyzed via 2-way ANOVA with Sidak’s multiple comparison test.    

Tissue Processing. Mice were given an overdose of ketamine/xylazine and transcardially perfused with 30 mL of ice-cold 1X PBS. Brains and spleen were collected and placed in cold complete RPMI media (cRPMI) (10% FBS, 1% sodium pyruvate, 1% non-essential amino acids, 1% penicillin/streptomycin, and 0.1% 2-ME). Brains were minced with a razor blade, passed through an 18G needle, and enzymatically digested with 0.227 mg/mL collagenase/dispase and 50U/mL DNase (Roche) for 45 minutes at 37C. If brain samples were used to quantify parasite burden or for gene expression analysis, aliquots were removed and frozen for downstream analysis prior to addition of digestion enzymes. If brain cells were being used for downstream RNA sequencing for WT vs. MGSTAT1∆ analysis, Actinomycin D (Sigma-Aldrich) was added to the digestion buffer at a concentration of 45 µm during incubation and 3 µm during washes to inhibit upregulation of immediate early activation genes associated with subsequent FACs sorting. All brain samples were resuspended in 20 mL of 40% percoll and spun for 25 minutes at 650 xg to remove myelin. Following myelin removal, samples were washed with and subsequently resuspended in cold cRPMI. Blood collected for flow cytometric analysis was isolated from the heart prior to transcardial perfusion and transferred into 1x PBS + EDTA (Thermo Fisher Scientific). Blood was then processed with RBC lysis and resuspended in cold cRPMI prior to staining and fixation for flow cytometry. For peritoneal lavage experiments, 5 mL of cold 1X PBS was injected through the membrane encasing the peritoneal cavity via a 26G needle and withdrawn with a 22G needle. Lavage fluid was washed and suspended in cRPMI.

Flow cytometry. Following generation of a single-cell suspension, cells were plated in a 96-well plate and incubated for 10 minutes in 50 μL Fc block (1 μg/mL 2.4G2 Ab (BioXCell), 0.1% rat γ-globulin (Jackson ImmunoResearch) at room temperature. Cells were incubated in primary antibodies at a concentration of 1:200, and AF-780 viability dye (eBioscience) at a concentration of 1:800 for 30 minutes at 4°C. Antibody clones used for experiments included: MHC II (M5/114.15.2), CD11b (M1/70), Ly6C (HK1.4), iNOS (CXNFT), CD45 (30-F11), CD3e (145-2C11), CD4 (GK1.5), CD8a (53-6.7), and IFN-γ (XMG1.2) (Thermo Fisher Scientific). After staining for surface markers, cells were washed and fixed overnight in 2% PFA at 4°C, before being washed and intracellularly stained, if quantifying cytosolic protein. For intracellular cytokine staining (IFN-γ), initial single cell suspensions were incubated with Brefeldin A (Selleckchem) for 5 hours at 37°C prior to blocking and staining. For intracellular staining, cells were permeabilized with Permeabilization Buffer (eBioscience) and stained for 30 minutes at room temperature. Cells were washed with FACS buffer and transferred into 5 mL FACS tubes, then were analyzed on a Gallios flow cytometer (Beckman-Coulter). Flow cytometry data was analyzed using FlowJo.

Cell Sorting / Enrichment. For RNA sequencing and analysis of microglial Stat1 relative expression to validate excision, brains were processed into a single cell suspension, as described above. Cells were then magnetically labeled with CD11b-conjugated beads diluted in MACS buffer for 15 minutes, per manufacturer’s instructions. Following a wash with 2 mL of MACS buffer, samples were spun at 1500 RPM for 5 minutes and resuspended in 600 uL of MACS buffer. Myeloid cells were then positively selected for using anti-CD11b-conjugated magnetic beads enrichment (Miltenyi). Cells were resuspended and lysed in Trizol for RT-qPCR analysis, or incubated for 10 minutes in 50 μL Fc block (1 μg/mL 2.4G2 Ab (BioXCell), 0.1% rat γ globulin (Jackson ImmunoResearch) at room temperature for RNA-sequencing. Cells were stained with the following antibodies (Thermo Fisher Scientific) for 30 minutes at 4°C: CD11b-Percp Cy5.5 (Cat. #45-0112-82), MHCII-eFluor 450 (Cat. # 48-5321-82), CD45-APC (Cat. #17-0451-81), Ly6C-PE Cy7 (Cat. #12-5932-80), CD3e-PE Cy7 (Cat. #25-0031-81), NK1.1-PE Cy7 (Cat. #25-5941-81), CD19-PE Cy7 (Cat. #25-0193-81). Live cells were analyzed and sorted using a BD Aria flow cytometer at the University of Virginia Flow Cytometry Core facility. Cells were sorted based on ZsGreen and dump gating (CD3e- NK1.1- CD19- Ly6C-) directly into Trizol (Invitrogen) for RNA extraction and RNA sequencing. For MGSTAT1∆ mice, MHC IIneg microglia were gated in order to positively select for STAT1-deficient cells.

Quantitative RT-PCR. For tissue-level analysis, one-fourth of a mouse brain was placed in 1 mL Trizol (Ambion), mechanically homogenized using 1 mm zirconia/silica beads (Biospec) for 30 seconds using a Mini-BeadBeater 16 (BioSpec). For gene expression analysis of magnetically-enriched cells, cells were homogenized in Trizol by pipetting. RNA was extracted from Trizol according to manufacturer’s instructions (Invitrogen). High Capacity Reverse Transcription Kit (Applied Biosystems) was used to generate cDNA. Quantitative PCR was performed using 2X Taq-based Master Mix (Bioline) and TaqMan gene expression assays (Applied Biosystems), or custom primers (Integrated DNA Technologies), run on a CFX384 Real-Time System thermocycler (Bio-Rad Laboratories).  Murine Hprt and T. gondii Act1 were used for normalization for analyzing host and parasite gene expression, respectively, and relative expression is reported as 2(−ΔΔCT). The following Thermo Fisher mouse gene probes were used: Stat1 (Mm00439518_m1), Hprt (Mm00446968_m1), Ifng (Mm01168134_m1), Nos2 (Mm00440502_m1), Il6 (Mm00446190_m1), Icam1 (Mm00516023_m1), Vcam1 (Mm01320970_m1), Tnfa (Mm00443258_m1), Ccl2 (Mm00441242_m1), Ccl5 (Mm01302427_m1), Cxcl9 (Mm00434946_m1), Cxcl10 (Mm00445235_m1). Custom primers for used for analyzing T. gondii genomic DNA and gene expression were used and are provided in Supplementary Table 1.

Funding

National Institute of Neurological Disorders and Stroke, Award: R01NS112516

National Institute of Neurological Disorders and Stroke, Award: R56NS106028