Introduction
Among peripheral leukocytes invading the injured brain, T cells have been consistently identified as the invading leukocyte subpopulation with the largest impact on secondary neurodegeneration and modulation of the ischemic brain damage (Kleinschnitz et al., 2010; Liesz et al., 2011). T cell subpopulations have the potential to play either a neuroprotective or a deleterious role in post-stroke neuroinflammation. In particular, the pro-inflammatory TH1, TH17 subsets of THELPER cells, and IL-17-producing γδ T cells have been shown to induce secondary neurotoxicity, leading to infarct expansion with worse functional outcome (Gelderblom et al., 2012; Shichita et al., 2009), whereas regulatory T cells (TREG) exert anti-inflammatory and neuroprotective function suppressing an excessive inflammatory reaction to the brain infarct. Recruitment of peripheral immune cells is not limited to the brain parenchyma since an accumulation of T cells is observed in the choroid plexus and the meninges after stroke (Benakis et al., 2016; Llovera et al., 2017). Attempt in blocking the recruitment of peripheral effector T cells diminished neuronal damage in different cerebral ischemic models, resulting in improvement of stroke outcome and suggesting a possible therapeutic target (Liesz et al., 2011; Llovera et al., 2015). Considering the relatively low number of only a few thousand lymphocytes invading the brain after stroke compared to more than 50 times higher cell count of innate immune cells (invading and resident) in the post-stroke brain (Gelderblom et al., 2009), it is surprising to observe such a dramatic effect of a small number of T cells on the neuroinflammatory response to stroke.
Therefore, we hypothesized that T cells have a polarizing effect on microglial function. In turn, microglia – as the most abundant immune cell population in the ischemic brain – could amplify the T cells’ impact on the cerebral immune milieu. Indeed, microglia interact with T cells via either cell-to-cell contact, cytokine-mediated communication, or antigen presentation, leading to activation/polarization of adaptive immune cells entering the brain (Goldmann and Prinz, 2013). T cell-microglia interaction can further influence the neuroinflammatory response in experimental models of multiple sclerosis (Dong and Yong, 2019) and possibly in stroke (Wang et al., 2016). In fact, recent evidence suggests a crosstalk between microglia and T cells as a key determinant of neuronal plasticity during recovery from brain injury (Shi et al., 2021). However, while the influence of microglia/macrophages on T cells has been well studied, it is still unclear how in reverse the T cells influence microglial function, and whether early interaction of T cells with microglia in the acute response to stroke can have an immediate impact on microglia and further change the course of disease progression.
Using morphological analysis, single-cell sequencing and adoptive transfer models of ex vivo differentiated THELPER cell subpopulations, we performed an in-depth analysis of the immunomodulatory effects of T cells on microglial polarization. Better understanding of the T cell-microglia crosstalk holds the potential to use polarized T cells as a therapeutic approach with large impact on the cerebral inflammatory milieu potentiated by resident microglia.
Results
Lymphocytes modulate the activation state of microglia in response to stroke
First, we investigated the effect of lymphocytes on microglial morphology and transcriptome in male
Figure 1.
Lymphocytes influence microglia morphology after stroke.
(A) Schematic of the experimental design: morphological analysis of microglia and transcriptomic profile of sorted microglia were performed in naïve mice or 5 days after stroke in wild-type (WT) and
CD45+CD11b+ myeloid cells were sorted by flow cytometry from naïve mice or 5 days after stroke (pool of 3 mice per condition; Figure 1A and Figure 2A). To better discriminate the transcriptional signature of microglial cells from other CD45+CD11b+ myeloid cells, we performed an unsupervised clustering analysis and identified 15 distinct clusters across conditions (Figure 2—figure supplement 1A). Based on the expression of previously defined markers of homeostatic and reactive microglia per cell cluster (high gene expression of
Figure 2.
Lymphocytes influence microglia transcriptional signature.
(A) CD45+CD11b+ cells were sorted from the ipsilateral hemisphere in naïve mice or 5 days after stroke in wild-type (WT) and
Figure 2—figure supplement 1.
Transcriptomic analysis of microglia isolated from wild-type (WT) and
(A) Uniform manifold approximation and projection (UMAP) plots showing expression of known CD45+CD11b+ myeloid cell marker genes and Louvain-clusters. (B and C) Manifold and clustering of microglia: (B) UMAP plots indicate Louvain-clusters of microglial cells. (C) Dot plots show gene expression distribution of marker genes split by Louvain-cluster, and their grouping into homeostatic (top) and reactive (bottom) microglia.
Figure 2—figure supplement 2.
Microglia single cell trajectory inference in wild-type (WT) and
(A and B) Microglia single cell trajectory inference describes the evolution of microglia activation from naïve to stroke condition. (A) Partition-based graph abstraction graph (PAGA, right) shows cluster connectivity of Louvain-clusters (left) with a threshold of 0.1. Nodes represent subsets, and thicker edges indicate stronger connectedness between subsets. The trajectories path 1 and 2 are selected based on the connectivity (edge width) in the PAGA after the root and end paths were defined based on marker gene expression. Clusters along the paths from root-to-end were merged since these are consecutive. Trajectory path 1 (dark gray): root (cluster 0), path 1 (clusters 6, 1, and 3 merged), and end (cluster 7); trajectory path 2 (black): root (cluster 0), path 2 (cluster 4, 2, and 5 merged), and end (cluster 7). (B) Barplot shows cell frequencies split by condition in trajectories path 1 and path 2. (C and D) Differential microglial gene expression analysis between root and end clusters along trajectory path 2. (C) Heatmaps show scaled expression along path 2 trajectory of microglial genes specifically regulated in
Figure 2—figure supplement 3.
Immune cell infiltration in wild-type (WT) and
(A) Absolute cell count of CD3+ T cells and CD19+ B cells in the ischemic hemisphere (ipsilateral) 5 days after distal occlusion of the middle cerebral artery (dMCAO) in WT mice as quantified by flow cytometry. There is a 14-fold increase of T cells in comparison to B cells at this time point; Student t-test, ***p<0.001. (B) Absolute cell count of CD45highCD11b+ myeloid cells: Ly6G+ Neutrophils, CD11c+MHCII+ dendritic cells, Ly6C+Ly6G– monocytes, F4/80+Ly6G– macrophages in the ischemic hemisphere (ipsilateral) 5 days after dMCAO in WT (red) and
The microglial reaction to stroke causes a gradual shift from the homeostatic transcriptomic profile to a reactive state. In order to capture differences in the microglia transcriptome along its transition phase, we performed single-cell trajectory inference analysis (Figure 2—figure supplement 2). Partition-based graph abstraction (PAGA) revealed two distinct paths with high connectivity from the homeostatic (naïve) microglia cluster (root cluster) to the reactive (stroke) microglia cluster (end cluster; Figure 2—figure supplement 2A). Interestingly, the number of microglial cells in stroke
Because
THELPER cell subpopulations drive the distinct polarization of microglia
To test whether microglial phenotypes can be specifically skewed by the CD4+ T cell subsets of functionally opposing THELPER cell subpopulations, we differentiated TH1 and TREG in vitro (Figure 3—figure supplement 1A) and tested whether these THELPER cells can reprogram the stroke-associated microglia. Differentiated T cells or vehicle were injected into the cisterna magna (CM) of lymphocyte-deficient
Figure 3.
TH1 and regulatory T cells (TREG) cells influence microglia gene expression after stroke.
(A) Naïve CD4 cells were polarized in vitro to TH1 or TREG phenotype (Figure 3—figure supplement 1A). One million cells (TH1 or TREG cells) or vehicle (control, CT) were injected into the cisterna magna (CM) in
Figure 3—figure supplement 1.
T cell polarization in vitro and infarct volumetry in
(A) CD4 naïve T cells were polarized in vitro toward TH1 (top row, CD4+Tbet+) or regulatory T cells (TREG; bottom row, CD4+FoxP3+). (B and C) Infarct volume analysis 5 days after distal occlusion of the middle cerebral artery (dMCAO) as quantified on cresyl violet stained coronal sections (20 μm thick sections taken every 400 μm). Data are represented as mean ± SD. Each dot corresponds to one mouse. Student t-test; NS, non significant.
Figure 4.
Acute post-stroke treatment with engineered T cells overexpressing IL-10 modulates microglial activation and ameliorates functional deficit.
(A and B) Flow cytometry analysis and whole skull-brain coronal sections of 106 eGFP+TH1 cells injected into the cisterna magna (CM) of
Figure 4—figure supplement 1.
Localization of polarized T cell and IL-10 plasmid construct.
(A) Gating strategy of CD4+eGFP+ cells analyzed by flow cytometry in the ipsilateral (red) and contralateral (blue) hemispheres (CD45+CD11b–CD19–CD3+CD4+FITC+) in
Engineered T cells overexpressing IL-10 induce a pro-regenerative transcriptomic profile in microglia
In order to further explore the implication of TREG-microglia interactions to modulate the post-stroke inflammatory environment, we tested the therapeutic potential of the known anti-inflammatory properties of TREG via IL10 on the local microglial immune milieu. We engineered T cells by viral transfection to overexpress the anti-inflammatory cytokine IL-10 (eTc-IL10; Figure 4—figure supplement 1B, C). In a therapeutic approach, we injected eTc-IL10 cells into the CM of WT mice 4 hr after stroke – a translationally relevant time window considering a similar time window for acute therapy with thrombolytics in stroke patients (Figure 4C). We investigated whether eTc-IL10 treatment affected stroke outcome but did not find any difference in infarct volumes between conditions (Figure 4D). This is in accordance with the concept of early ischemic lesion formation in stroke which is not being affected by the delayed immunological mechanisms (Dirnagl et al., 1999). In contrast, mice receiving eTc-IL10 injection in the CM had a significant improvement of functional outcome at 48 hr after stroke as shown by a reduced forelimb asymmetry in comparison to vehicle-treated mice (Figure 4E). This might reflect the implication of inflammatory pathways and specifically cytokine secretion on functional deficits and delayed recovery after stroke in contrast to the early primary lesion development (Filiano et al., 2017; Roth et al., 2020). We then evaluated whether gene expression was altered after stroke upon eTc-IL10 treatment. RNA was isolated from the whole ischemic hemisphere, and neuroinflammatory genes were quantified using the Nanostring platform. Interestingly, we found that several genes associated with a DAM profile were down-regulated in mice treated with eTc-IL10 such as
Discussion
The cellular constituents of the acute neuroinflammatory response to stroke have been well characterized, including microglial activation, leukocyte invasion, and the contribution of different lymphocyte subpopulation (Anrather and Iadecola, 2016). However, the reciprocal interactions of these different immune cell populations remain largely under-investigated in the context of brain injury. A better understanding of the T cell-polarizing effect on microglial function has strong translational implication since T cells may act as a ‘Trojan horse’ with large impact on the cerebral inflammatory milieu potentiated by microglia (Cramer et al., 2018).
Here, we established a mechanistic link between T cells and microglial morphology and transcriptomic signature in the context of stroke. We showed the distinct role of T cell subpopulations on switching microglial polarization state in response to stroke. Our results from transcriptomic analysis suggest that the microglia-polarizing effect of different THELPER cell subpopulations is mainly mediated via their specific cytokine/chemokine secretion pattern. Microglia that were challenged with TH1 cells expressed an up-regulation of genes associated with type I INF signaling – the key cytokine secreted by the TH1 subpopulation. In contrast, TREG cells modulated a gene set in microglia associated with chemotaxis-mediated mechanisms (
An important caveat and potential key reason for the so far still pending success in harnessing the therapeutic function of IL-10 are its short half-life (less than 1 hr) and limited bioactivity after in vivo administration as a recombinant protein (Le et al., 1997; Saxena et al., 2015). Moreover, the systemic IL-10 application can have considerable and unforeseen side-effects due to the potentially divergent function of IL-10 on inflamed and homeostatic tissue, including direct effects on neurons, astrocytes, endothelial cells, and other cellular constituents of physiological brain function (Saraiva et al., 2019). Therefore, we aimed to take a different approach for the localized and sustained production of IL-10 at the inflamed peri-lesional brain parenchyma. For this, we took advantage of the potent capability of T cells to be specifically recruited and accumulated to the ischemic lesion site in order to deliver IL-10 from genetically engineered IL-10-overexpressing T cells (Heindl et al., 2021; Llovera et al., 2017). We demonstrated that IL-10 overexpression by this approach substantially modulated microglia gene expression by down-regulation of microglial gene signature associated with phagocytosis of synapses correlating with functional recovery after stroke. Interestingly, eTc-IL10 cells did not exclusively invade the injured brain but were also located in the meningeal compartment and could additionally contribute to functional recovery by resolving inflammation at these border structures or providing IL-10 to the brain parenchyma along CSF flow. This concept is in accordance with previous observations of meningeal immune cell accumulation after stroke (Benakis et al., 2018) and that meningeal T cell-derived cytokines may enter the brain via CSF flow and paravascular spaces (Iliff et al., 2012).
An important finding in this study was the observation that IL-10 overexpression in T cells modulated microglial genes involved in the complement pathway, phagocytosis, and synaptic pruning and was associated with a better functional outcome after stroke. Complement factors are localized to developing CNS synapses during periods of active synapse elimination and are required for normal brain wiring (Schafer et al., 2012). Inactive synapses tagged with complement proteins such as C1q may be eliminated by microglial cells. Likewise in the mature brain, early synapse loss is a hallmark of several neurodegenerative diseases (Stephan et al., 2012). Indeed, complement proteins are profoundly up-regulated in many CNS diseases prior to signs of neuron loss, suggesting mechanisms of complement-mediated synapse elimination regulated by microglia potentially driving disease progression (Stephan et al., 2012) and stroke recovery. It is therefore conceivable that T cells overexpressing IL-10 down-regulate the complement system in microglia and prevent excessive elimination of synapse and consequently protect against neuronal dysfunction. This is particularly of interest because microglia effector function has not only been associated with inflammatory neurodegenerative processes but recently also been shown to be neuroprotective (Szalay et al., 2016) by tightly monitoring neuronal status through somatic junctions (Cserép et al., 2020). Microglia interact with the extra-neuronal space by not only regulating the elimination of existing synapses but also by modifying the extracellular matrix to enable efficient synaptic remodeling (Zaki and Cai, 2020). Accordingly, we found T cell-dependent regulation of several microglial genes that can mediate such extracellular matrix modifications involved in phagocytosis and proteases (
Whereas at this acute time point, the transcriptomic changes in microglia are mainly attributed to their reactivity to the tissue injury itself, we have been able to demonstrate that brain-invading T cells can specifically ‘fine-tune’ the transition of the stroke-associated microglia to a distinct cell morphology and transcriptomic profile. Our data suggested that the anti-inflammatory TREG cells induce a shift of microglial genes associated with a homeostatic state and immune cell recruitment. However, the specific functional change of microglia induced by T cell subsets and biological significance for stroke remain to be further investigated. We postulate that the development of engineered T cells could have important translational implication by targeting a specific effector function of microglia with a relevant impact on the chronic progression of stroke pathobiology.
Materials and methods
Animal experiments
All animal procedures were performed in accordance with the guidelines for the use of experimental animals and were approved by the respective governmental committees (Licenses: 02-21-46 and 02-21-95; Regierungspraesidium Oberbayern, the Rhineland Palatinate Landesuntersuchungsamt Koblenz). Male WT C57BL6/J mice were purchased from Charles River,
Permanent distal middle cerebral artery occlusion model
Permanent coagulation of the middle cerebral artery (MCA) was performed as previously described (Llovera et al., 2014). Briefly, animals (male; age = 8–12 weeks) were anesthetized with volatile anesthesia (isoflurane in 30%O2/70%N2O) and placed in lateral position. After a skin incision between eye and ear, the temporal muscle was removed, and the MCA was identified. Then, a burr hole was drilled over the MCA, and the dura mater was removed. The MCA was permanently occluded using bipolar electrocoagulation forceps. Permanent occlusion of the MCA was visually verified before suturing the wound. During the surgery, body temperature was maintained using a feedback-controlled heating pad. Mice that developed a subarachnoid hemorrhage during surgery were excluded from the analysis.
Cylinder test
To evaluate forepaw use and asymmetry, the cylinder test was performed 2 days prior to stroke (baseline) and day 2 and day 4 post stroke. Mice were placed in a transparent acrylic glass cylinder (diameter 8 cm; height: 25 cm) in front of two mirrors and videotaped. To assess independent forelimb use, contact with one forelimb (left and right forelimbs) during full rearing and landing performance of mice was scored by frame-to-frame analysis of recorded videos. Mice with forepaw preference at baseline (absolute value difference between right and left forepaws >10) were excluded from the analysis. All rearing movements during the trial were counted and used as indication of the animal’s overall activity.
Intra-CM injection
Mice were anesthetized with isoflurane in 30%O2/70%N2O and fixed in a stereotaxic frame by the zygomatic arch, with the head slightly tilted to form an angle of 120° in relation to the body. A small incision was made at the nape of the neck between the ears to expose the neck muscles, which were bluntly dissected to expose the CM. Cannulas composed of a glass capillary (ID, inner diameter 0.67 mm; OD, outside diameter, 1.20 mm) attached to a polyethylene tubing (ID 0.86 mm and OD 1.52 mm; Fisher Scientific UK Ltd.) were used to perform the CM injections. Glass capillaries were sharpened using a flaming micropipette puller (P-1000, Sutter Instrument GmbH), filled with 10 µL of the cell suspension diluted in artificial CSF (aCSF: 126 mM NaCl, 2.5 mM KCl, 1.25 mM NaH2PO4, 2 mM Mg2SO4, 2 mM CaCl2, 10 mM glucose, and 26 mM NaHCO3; pH 7.4 when gassed with 95% O2 and 5% CO2), and fixed to the micromanipulator arm of the stereotaxic. Cell suspension was injected into the CM at a rate of 2 μL/min. At the end of the injection, mice are sutured and allowed to recover in a preheated awake cage for 1 hr, after which they are returned to the animal husbandry.
In vivo depletion of microglia
For microglia depletion, WT and
Infarct volume quantification
Mice were deeply anesthetized 5 days after stroke induction and transcardially perfused with 20 mL saline. Brains were removed, frozen immediately on powdered dry ice, and stored at −20°C until use. For infarct volumetry, brains were serially sectioned (400 μm intervals, 20 μm thick) and stained for cresyl violet (CV) as previously described (Llovera et al., 2014). CV-stained sections were scanned at 600 dpi on a fatbed scanner (Canon). Direct infarct measurement was used after validating the absence of edema at the investigated time point. The total infarct volume was measured with ImageJ and determined by integrating measured areas and distances between sections.
Immunohistochemistry and confocal microscopy
Microglia morphology analysis was performed on brain coronal sections as previously described (Heindl et al., 2018). Briefly, mice were perfused with 4% paraformaldehyde (PFA), and brains were post-fixed overnight and placed in sucrose for dehydration. Then, free floating 100 µm coronal sections were stained for microglia with 1:200 anti-Iba1 (rabbit, Wako, #019–19741). Nuclei were stained using 4’,6-Diamidin-2-phenylindol (DAPI, Invitrogen, #D1306), and images were acquired at a distance of 900 μm from the border of the lesion in layer 4 (ipsilateral) and the homotypic contralateral region using a Zeiss confocal microscope with 40× magnification (objective: EC Plan-Neofluar 40×/1.30 Oil DIC M27) with an image size of 1024×1024 pixel, a pixel scaling of 0.2×0.2 μm, and a depth of 8 bit. Confocal images were collected in Z-stacks with a slice-distance of 0.4 μm. Morphological features of microglia were acquired using a fully automated analysis as previously described (Heindl et al., 2018).
Fluorescent in situ hybridization
Single-molecule fluorescence in situ hybridization (smFISH) was performed using the RNAscope Multiplex Fluorescent Reagent Kit v2 (Advanced Cell Diagnostics) by the manufacturer’s protocols. Briefly, free floating 100 µm coronal brain sections (Figure 3e) or 20 µm cryo-sections (Figure 4g) were first dried, washed, and then incubated in RNAscope hydrogen peroxide. Antigen retrieval and protease treatment were performed as per protocol. Sections were then incubated with the probe mix (C2-
Whole skull immunofluorescence
Rag-1−/− mice were anesthetized with isoflurane and perfused transcardially with ice-cold PBS followed by 4% PFA. After removing the mandibles, skin and muscles were carefully detached from the skull (http://www.nature.com/protocolexchange/protocols/3389). The skull decalcification was performed as previously described (Benakis et al., 2016). Coronal skull sections (20 μm) were stained with GFP-booster Atto647N (1:500, ChromoTek GmbH) to visualize eGFP-labeled T cells. Sections were counterstained with DAPI (Invitrogen) to visualize cell nuclei and observed by confocal laser microscopy (Leica SP5).
In vitro T cell polarization
Single-cell suspensions were generated from spleen, inguinal, axial, brachial, and mandibular lymph nodes of C57BL/6 or b-actin-EGFP mice by passing the tissue through a 70 μm cell strainer. Naive CD4+ T cells were obtained by pre-enrichment using an ‘untouched’ CD4+ T Cell Isolation Kit (Miltenyi Biotec) with subsequent flow cytometric analysis (CD4+ [clone RM4-5, 0.5 ng/μL], CD44low [clone IM7, 2 ng/μL], and CD62Lhigh [MEL-14, 0.8 ng/μL]). Cells were seeded at a density of 300,000 or 400,000 cells/well in a flat-bottom 96-well plate and stimulated with plate-bound anti-CD3 and anti-CD28 Abs 0.5 μg/mL or 2 μg/mL anti-CD3 (clone 145–2 C11) for TREG and TH1, respectively and 2 μg/mL anti-CD28 (clone 37.51). Different mixtures of cytokines and mAbs were added to RPMI (supplemented with 10% fetal calf serum (FCS), 50 μM β-mercaptoethanol, 50 U/mL penicillin, 50 μg/mL streptomycin, 1% GlutaMAX, and 1% N-2-hydroxyethylpiperazine-N-2-ethane sulfonic acid [Gibco HEPES]) and used as follow: TH1 conditions with anti-IL-4 (10 μg/mL, BioXCell, #BE0045) and IL-12 (10 ng/mL, BioLegend, #577002); TREG conditions: anti–IL-4 (10 μg/mL, BioXCell, #BE0045), anti-IFN-γ (10 μg/mL, BioXCell, #BE0055), and TGFβ (3 ng/mL, BioLegend, #580702). After 2 days in culture, cells were split into two new 96-well plates and incubated with freshly prepared supplemented RPMI media with IL-2 (10 ng/mL, BioLegend, #575402). Cells were cultured for a total of 5 days before injection. Quality control was performed on day 4 to assess the percentage of T cell expressing Tbet (clone 4B10, 2 ng/μL; TH1) or FoxP3 (clone FJK-16s, 2 ng/μL; TREG; Figure 3—figure supplement 1A). One million differentiated T cells were resuspended in sterile aCSF and injected into the CM in
IL-10 overexpression in naïve T cells
Engineered T cells overexpressing IL-10 (eTc-IL10) were generated by transfection of naïve T cells with an IL-10 plasmid (pRP[Exp]-TagBFP2-CMV>mIl10[NM_010548.2]) designed and prepared by VectorBuilder (Figure 4—figure supplement 1B). First, splenocytes were isolated from C57BL/6 mice (male, 6–12 weeks old) and enriched using a CD4+ T Cell Isolation Kit (Miltenyi Biotec, No:130-104-453). Quality control was performed by flow cytometry (CD4+ [clone RM4-5, 1:25], CD44low [clone IM7, 1:25], and CD62Lhigh [clone MEL-14, 1:25]). Cells were resuspended in RPMI (supplemented with 10% fetal bovine serum (FBS), 50 μM β-mercaptoethanol, 50 U/mL penicillin, 50 μg/mL streptomycin, 1% GlutaMAX, and 1% HEPES and 10 ng/mL IL-2). To induce CD4+ cells to enter the cell cycle for efficient DNA uptake, 4×105 cells/well were seeded in a flat-bottom 96-well plate containing bound anti-CD3 (2 μg/mL, clone 145–2 C11) and anti-CD28 (2 μg/mL, clone 37.51) for 48 hr. After 48 hr stimulation, 1.5×106 cells multiplied by the number of mice to be injected were transfected with the pIL-10 vector (1×106 cells/1.5 μg pIL-10 DNA per cuvette) using the Mouse T Cell Nucleofector Kit (Lonza, No: VPA-1006) with Nucleofector II Device (program X-100). Once electroporated, cells were diluted with conditioned RPMI from the 48 hr stimulation and fresh supplemented RPMI (1:1) and seeded in a 12-well plate (1 cuvette of cells/ well). 24 hr post transfection, cells and supernatant were collected. Supernatant was used to confirm IL-10 secretion by ELISA (Figure 4—figure supplement 1C), and cells were collected for intra-CM injection (1×106/mouse).
ELISA
Secreted IL-10 was determined by ELISA as per the manufacturer’s protocol (Mouse IL-10, Invitrogen, No: 88-7105-88). The color reaction was measured as OD450 units on a Bio-Rad iMark microplate reader. The concentration of supernatant IL-10 was determined using the manufacturer’s standard curve over the range of 32–4000 pg/mL.
Flow cytometry
For differentiation of live and dead cells, we stained cells with the Zombie Violet Fixable Viability Kit according to the manufacturer’s instructions (BioLegend). For surface marker analysis, cell suspensions were adjusted to a density of 0.5×106 cells in 50 μL FACS buffer (2% FBS, 0.05% NaN3 in PBS). Nonspecific binding was blocked by incubation for 10 min at 4°C with anti-CD16/CD32 antibody (Biolegend, clone 93, 5 ng/ μL) antibody and stained with the appropriate antibodies for 15 min at 4°C. The following antibodies were used for extracellular staining: CD45 (clone 30 F-11, 0.5 ng/μL), CD4 (clone RM4-5, 0.5 ng/μL), CD11b (clone M1/70, 0.6 ng/μL), CD19 (eBio1D3, 0.6 ng/μL), B220 (clone RA3-6B2, 0.32 ng/μL), CD3ε (clone 145–2 C11, 2 ng/μL), CD8a (clone 53–6.7, 2 ng/μL), and CD62L (clone MEL-14, 0.8 ng/μL) from Thermofisher. For intracellular cytokine staining, cells were restimulated for 4 hr with PMA (50 ng/mL, Sigma), ionomycin (1 µM, Sigma), and brefeldin A (1 µL for ~106 cells/mL). Cells were then stained for surface markers as detailed below, fixed, and permeabilized using Fixation and Permeabilization Buffers from eBiosciences following the manufacturer’s instructions. Briefly, cells were fixed for 30 min at 4°C (or RT for FoxP3), washed with permeabilization buffer, and incubated for 30 min with the appropriate antibodies in permeabilization buffer at 4°C (or RT for FoxP3). The cells were stained with the transcription factors FoxP3 (clone FJK-16s, 2 ng/μL) and T-bet (clone 4B10, 2 ng/μL) or IFN-γ (clone 4 S.B3, 2 ng/μL). Cells were washed with FACS buffer, resuspended in 200 μL of FACS buffer and acquired using a BD FACSverse flow cytometer (BD Biosciences, Germany), and analyzed using FlowJo software (Treestar, USA). Isotype controls were used to establish compensation and gating parameters.
Nanostring analysis
The ipsilateral hemispheres were lysed in Qiazol Lysis Reagent, and total RNA was extracted using the MaXtract High Density kit with further purification using the RNeasy Mini Kit (all Qiagen). 70 ng of total RNA per sample was then hybridized with reporter and capture probes for nCounter Gene Expression code sets (Mouse Neuroinflammation codeset) according to the manufacturer’s instructions (NanoString Technologies). Samples (6/condition) were injected into NanoString cartridge, and measurement run was performed according to nCounter SPRINT protocol. Background (negative control) was quantified by code set intrinsic molecular color-coded barcodes lacking the RNA linkage. As a positive control code set, intrinsic control RNAs were used at increasing concentrations. Genes below the maximal values of the negative controls were excluded from the analysis. All gene counts were normalized (by median) and scaled (mean-centered and divided by SD of each variable). Heatmaps were performed using the MetaboAnalystR package on normalized expression values. The regulated genes in microglia treated with TH1 or TREG in comparison to vehicle treated microglia (CT) are represented in the volcano plots; genes with a p<0.05 were color-coded. The significantly up-regulated genes in microglia (FC>1.5 and p<0.05) were further used for pathway analysis using Cytoscape ClueGO (Bindea et al., 2009): TH1/CT, 19 up-regulated genes (
Microglia cell isolation for RNA sequencing
Mice were perfused transcardially with ice-cold saline containing Heparin (2 U/mL). Brains were placed in HBSS (w/ divalent cations Ca2+ and Mg2+) supplemented with actinomycin D (1:1000, 1 mg/mL, Sigma, #A1410), and microglia was isolated with the Papain-based Neural Tissue Dissociation Kit (P) (# 130-092-628, Miltenyi Biotec B.V. & Co. KG) according to the manufacturer’s instructions. Cell suspension was enriched using 30% isotonic Percoll gradient. 1×103–1.5×103 live microglia cells from 3 mice per condition were sorted according to their surface marker CD45+CD11b+7-AAD negative (SH800S Cell Sorter, Sony Biotechnology) and proceed for 10× Genomics according to the manufacturer’s instructions (ChromiumTM Single Cell 3’ Reagent kits v2).
Single-cell data analysis
The CellRanger software (v2.0.0, 10× Genomics) was used for demultiplexing of binary base call files, read alignment, and filtering and counting of barcodes and unique molecular identifiers (UMIs). Reads were mapped to the mouse genome assembly reference from Ensembl (mm10/GRCm38). Downstream data analyses were performed using the Scanpy API (scanpy v ≥ 1.4 with python3 v ≥ 3.5; Wolf et al., 2018). Details on analyses, selected thresholds, and package versions are provided in available source scripts (See Code and Data availability). Outlier and low-quality cells were filtered if the fraction of mitochondria-encoded counts was greater than 10%, or the total number of counts was greater than 48,000. Thresholds were selected upon visual inspection of distributions as recommended (Luecken and Theis, 2019). Genes expressed in less than 10 cells were excluded. Furthermore, doublet cells as identified by the Scrublet algorithm (v0.2.1; Wolock et al., 2019) were excluded. Doublet scores and thresholds were determined for each sample separately. Raw counts of a cell were normalized by total counts neglecting highly expressed genes which constitute more than 5% of total counts in that cell. Then, counts were log-transformed (log[count+1]). These processed and normalized count matrices were used as input for all further analyses.
For the full data set and the microglia subset, first a single-cell nearest-neighbor graph was computed on the first 50 independent principal components. Principle components were calculated using the 3000 most variable genes of the full data set as input. The UMAP algorithm (Becht et al., 2019) as used to obtain a two-dimensional embedding for visualization. Iterative clustering was performed with the Louvain algorithm (Blondel et al., 2008) as implemented in louvain-igraph (v0.6.1, Traag et al., https://github.com/vtraag/louvain-igraph) with a varying resolution parameter. Clusters were annotated using previously described marker genes and merged if expressing the same set of marker genes. For Figure 2d and e, data were converted into a Seurat object and further analyzed in R to identify differentially expressed genes between WT and
Trajectories from homeostatic to reactive microglia were inferred with PAGA (Wolf et al., 2018) and diffusion pseudotime (DPT; Haghverdi et al., 2016) algorithms. First, clusters were grouped into two paths connecting the root and end cell cluster based on the computed cluster connectivities (PAGA), then cells were ordered along these paths based on the random-walk-based cell-to-cell distance (DPT). To capture processes specific to the path 2 trajectory in stroke-associated microglia, data was first subset to cells of path 2 and end cells clusters of stroke samples and gene expressed in less than 20 cells of the subset excluded. Then, gene sets were computed by clustering the 500 most varying genes using their pairwise-Pearson correlation values as input and Ward’s hierarchical clustering method with Euclidean distance (scipy python package v.1.5.4; Virtanen et al., 2020). One gene set with average correlation <0.05 was excluded. Finally, to obtain an activation score per cell for a given gene set, cell scores were computed as described by Satija et al., 2015 and implemented in Scanpy in the
Code availability
Jupyter notebooks with custom python scripts for scRNA-seq analysis is available in a github repository (https://github.com/Lieszlab/Benakis-et-al.-2022-eLife.git), copy archived at swh:1:rev:04f5dead312f071a4c760607d68f94047444bbaa (Benakis et al., 2022).
Statistical analysis
Data are expressed as mean ± SD or median with interquartile range and were analyzed by unpaired Student’s t-test (two-tailed) or one- or two-way ANOVA and post-hoc tests as indicated in the figure legends. Exclusion criteria are described in the individual method sections. The data for microglia morphology are shown as median ± interquartile range, and statistical significance was tested using the Wilcoxon rank sum test with continuity correction and Bonferroni post-hoc correction for multiple testing in R (version 4.0.3).
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2022, Benakis et al. This work is published under https://creativecommons.org/licenses/by/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Neuroinflammation after stroke is characterized by the activation of resident microglia and the invasion of circulating leukocytes into the brain. Although lymphocytes infiltrate the brain in small number, they have been consistently demonstrated to be the most potent leukocyte subpopulation contributing to secondary inflammatory brain injury. However, the exact mechanism of how this minimal number of lymphocytes can profoundly affect stroke outcome is still largely elusive. Here, using a mouse model for ischemic stroke, we demonstrated that early activation of microglia in response to stroke is differentially regulated by distinct T cell subpopulations – with TH1 cells inducing a type I INF signaling in microglia and regulatory T cells (TREG) cells promoting microglial genes associated with chemotaxis. Acute treatment with engineered T cells overexpressing IL-10 administered into the cisterna magna after stroke induces a switch of microglial gene expression to a profile associated with pro-regenerative functions. Whereas microglia polarization by T cell subsets did not affect the acute development of the infarct volume, these findings substantiate the role of T cells in stroke by polarizing the microglial phenotype. Targeting T cell-microglia interactions can have direct translational relevance for further development of immune-targeted therapies for stroke and other neuroinflammatory conditions.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer