Abstract
While the detrimental role of non-regulatory T cells in ischemic stroke is meanwhile unequivocally recognized, there are controversies about the properties of regulatory T cells (Treg). The aim of this study was to elucidate the role of Treg by applying superagonistic anti-CD28 antibody expansion of Treg. Stroke outcome, thrombus formation, and brain-infiltrating cells were determined on day 1 after transient middle cerebral artery occlusion. Antibody-mediated expansion of Treg enhanced stroke size and worsened functional outcome. Mechanistically, Treg increased thrombus formation in the cerebral microvasculature. These findings confirm that Treg promote thrombo-inflammatory lesion growth during the acute stage of ischemic stroke.
Keywords: brain ischemia, inflammation, microcirculation, T cells, thrombosis
Introduction
It has been observed for many years that immune cells transmigrate over the blood–brain barrier into infarcted brain tissue. This process follows a defined time course, with neutrophils and macrophages beginning to accumulate in the brain some hours after stroke, and lymphocytes reaching maximum infiltration by approximately day 3.1 While the detrimental role of non-regulatory T-cell subsets in the acute phase of ischemic stroke is widely accepted,2, 3, 4 there is an ongoing debate about the contribution of regulatory T cells (Treg) to this pathology.4, 5, 6, 7
We were able to show that Treg cause microvascular dysfunction and consequently thrombus formation and secondary infarct growth in the acute phase after experimental ischemic stroke by interaction with endothelial cells and platelets.4,8 This interplay between inflammatory processes and thrombus formation has recently been referred to as ‘thrombo-inflammation'.8
Other groups reported secondary deterioration of stroke volumes and functional outcome in animals 7 days after blocking of Treg.5 Therefore, the aim of this study was to clarify the impact of Treg in the acute phase of ischemic stroke by the means of a superagonistic anti-CD28 antibody (CD28 SA) that leads to an expansion of pre-existing Treg in the lymphoid organs and the dissemination of increased Treg numbers in the peripheral blood.9,10
Materials and Methods
Details of the experimental procedure are provided in the Supplementary Information. The animal experiments were conducted in accordance with the recommendations of the European Convention for the Protection of Vertebrate Animals used for Experimentation and the current ARRIVE guidelines (http://www.nc3rs.org/ARRIVE). Experiments were approved by legal state authorities (Government of Lower Franconia).
Ischemia Model
Focal cerebral ischemia was induced in 6 to 8-week-old male C57BL/6, DEREG or Rag1−/− mice by 30-minute transient middle cerebral artery occlusion (tMCAO) as described.3,4,11 Infarct volumes were calculated from brain slices stained with 2,3,5-triphenyltetrazolium chloride. The Bederson score and the grip test score were used to monitor neurologic function.12,13
Expansion of Treg by CD28 SA Treatment in vivo
CD28 SA (clone D665, Exbio, Praha, Czech Republic, 50 μg/mouse) was applied 3 days before tMCAO (prophylactic application) or immediately after tMCAO (therapeutic application) by an intraperitoneal injection. MOPC-21 antibody (BioXCell, West Lebanon, NH, USA) served as control. Regulatory T cells were quantified by flow cytometry analysis (Foxp3 staining). Depletion of Treg in DEREG mice was achieved by the administration of diphtheria toxin (DTX, Merck, Darmstadt, Germany, 1 μg/mouse per day intraperitoneally) on three consecutive days before tMCAO.
Immunohistochemistry
Cryoembedded brain slices were stained with antibodies against CD31 (Abcam, ab9498, Cambridge, UK), CD4 (100506, BioLegend, San Diego, CA, USA), Ly6B.2 (MCA771GA, Serotec, Puchheim, Germany) or AF488-conjugated anti-platelet glycoprotein IX (generated by B Nieswandt, Würzburg, Germany). For quantification of occluded vessels hematoxylin–eosin staining was performed (10 μm slices), and the percentage of occluded vessels in every tenth slice was counted under 40-fold magnification.
Results
First, we confirmed that CD28 SA treatment leads to an expansion of Treg in mice (maximum on day 3), as has previously been described.10 We found a significant increase in the relative numbers of Treg in the blood and lymph nodes of wild-type mice (Supplementary Figure 1, P<0.05). In the next step, we ruled out that the CD28 SA itself influences the cerebral blood flow, hemodynamics (heart rate and mean arterial pressure) and the blood gas analysis (Supplementary Figures 2 and 3, Supplementary Table 1). These findings exclude that CD28 SA treatment alters important physiologic parameters that might influence stroke outcome and prove that middle cerebral artery occlusion and reperfusion were sufficient in our model.
Next, we assessed if the increase in Treg numbers before tMCAO influences stroke development in wild-type mice. Stroke volumes on day 1 were significantly larger (82.0±35.2 mm3) compared with wild-type animals that had received isotype control antibodies (46.5±36.4 mm3, P<0.05) (Figure 1A). To exclude that the CD28 SA influences outcome measures independent of its boosting effect on the Treg population, we additionally analyzed Rag1−/− mice that lack lymphocytes. As the CD28 SA had no impact on stroke volumes (control group: 30.4±2.8 mm3; CD28 SA group: 37.3±12.5 mm3, P>0.05) (Figure 1A) a direct effect of the CD28 SA on clinical outcome in the absence of T and B cells could be excluded. Importantly, increased stroke size in the wild-type mice also translated into worse functional outcome as assessed by the grip test (values are the median with 25th and 75th percentiles, respectively, in brackets (control: 3.0 (2.0, 4.5); CD28 SA: 2.0 (0.0, 3.0), P<0.05), but not in the Bederson score (control: 2.0 (2.0, 3.0); CD28 SA: 3.0 (2.0, 4.0), P>0.05) (Figure 1B). Again, there was no difference regarding behavioral testing between the treatment groups in Rag1−/− animals (not shown). Moreover, survival rates on days 1 and 3 were lower in CD28 SA pretreated animals (Figure 1C). Importantly, treatment with CD28 SA after tMCAO (therapeutic approach) had no impact on stroke volumes (Supplementary Figure 4) and functional outcome (not shown) on day 3.
Figure 1.
Increase in superagonistic anti-CD28 antibody (CD28 SA)-induced Treg worsens stroke outcome. (A) Left panel: representative 2,3,5-triphenyltetrazolium chloride stains of coronal brain sections of control and CD28 SA-treated wild-type mice on day 1 after transient middle cerebral artery occlusion (tMCAO). Right panel: infarct volume was increased in the CD28 SA-treated group (n=11–13 per group) in wild-type animals, but not in Rag1−/− mice (n=7 or 8 per group), unpaired, two-tailed Student's t-test. (B) CD28 SA-treated mice only had a slightly worse Bederson score (left panel) but performed significantly worse in the gript test (right panel) (n=11–13 per group), Mann–Whitney test. (C) Analysis of survival using a Kaplan–Meier curve (plotted from four independent experiments with each experiment including five mice per group), Log-rank test. (D) CD28 SA induces larger infarctions in DEREG mice still containing Treg, i.e., without diphtheria toxin (DTX) pretreatment, compared with mice receiving control antibody. In contrast, CD28 SA was unable to enhance lesion size in DEREG mice devoid of Treg, i.e., after DTX pretreatment (n=5–6 per group), one-way analysis of variance with post hoc Bonferroni adjustment for P values. NS=not significant; *P<0.05.
Rag1−/− mice do not exclusively lack Treg. To rule out the fact that the detrimental effect of the CD28 SA was at a relevant portion mediated by other lymphocyte subpopulations apart from Treg, we introduced the DEREG mouse model. In these mice Treg can be selectively depleted by the repetitive application of DTX.11 Superagonistic anti-CD28 antibody again significantly increased the numbers of circulating Treg in naïve DEREG mice but not in Treg-depleted DEREG mice (Supplementary Figure 5). Accordingly, CD28 SA pretreatment induced significantly larger infarctions in DEREG mice containing Treg, while this was no longer the case in the group of DEREG mice without Treg (Figure 1D).
In the next step, we analyzed if the increase in peripheral Treg alters the composition of the cellular infiltrate within the ischemic brain. Indeed, we found more ipsilesional CD4+ cells (Figure 2A, P<0.01) and neutrophils (Figure 2B, P<0.01) after pre-tMCAO CD28 SA treatment compared with control animals. A negative immunostaining control is provided in Supplementary Figure 6. The fact that CD28 SA treatment not only increased the amount but also the density of immune cells (Supplementary Figure 7) argues against an unspecific effect, related for instance to larger infarct volumes. Nevertheless, data must be interpreted with caution since brains have not been flushed before sampling in these experiments. Hence, the total cell numbers obtained here potentially include a considerable amount of cells derived from within the brain vasculature.
Figure 2.
CD28 SA treatment increases intracerebral immune cell accumulation and thrombosis after transient middle cerebral artery occlusion (tMCAO). (A) Representative immunocytologic stainings of brain-infiltrating CD4+ T lymphocytes (indicated by red arrows) on day 1 after tMCAO. Quantification revealed significantly higher numbers of CD4+ T lymphocytes in the ipsilesional hemispheres of superagonistic anti-CD28 antibody (CD28 SA)-treated mice when compared with MOPC-21 (control) treated mice (n=5 per group). **P<0.01, unpaired, two-tailed Student's t-test. (B) Representative immunocytologic stainings of brain-infiltrating Ly6B.2+ neutrophils on day 1 after tMCAO. Quantification also revealed significantly higher numbers of Ly6B.2+ neutrophils in the ipsilesional hemispheres of CD28 SA-treated mice when compared with MOPC-21 (control) treated mice (n=3 per group), **P<0.01, unpaired, two-tailed Student's t-test. (C) Representative hematoxylin–eosin (H&E)-stained brain sections on day 1 after tMCAO. Quantification of occluded ipsilesional vessels (black dots) revealed a significant increase in the CD28 SA-treated group (n=4 per group), *P<0.05, unpaired, two-tailed Student's t-test. The asterisk indicates a patent microvessel. (D) Representative immunocytologic stainings of platelet aggregates within the vasculature on day 1 after tMCAO. Hoechst depicts cell nuclei, CD31 stains endothelial cells. Quantification revealed significantly higher numbers of ipsilesional glycoprotein IX (GPIX)-positive aggregates in CD28 SA-treated mice when compared with control mice. (n=4 per group), *P<0.05, unpaired, two-tailed Student's t-test. (E) Immunohistochemical staining of brain sections from DEREG mice on day 1 after 30 minutes of tMCAO showing green fluorescent protein-positive Foxp3+ Treg predominantly in the cerebral vasculature (co-stained with CD31).
To confirm that Treg contribute to ischemic brain injury by boosting ‘thrombo-inflammation',8 we demonstrated a higher number of occluded brain vessels (Figure 2C, P<0.05) and glycoprotein IX-positive platelets (Figure 2D, P<0.05) in CD28 SA-pretreated mice compared with control mice. Again, the CD28 SA also increased the density of occluded vessels (Supplementary Figure 8). To further analyze the location of Treg in the ischemic brain during the early phase of stroke, we performed immunohistochemistry of brain specimen taking advantage of genetically modified mice, in which Foxp3-expressing cells are visible by a transgenic construct linking green fluoresecent protein and the DTX receptor (DEREG mice).4,11 We could show that Treg were predominantly located within the vessel lumina (Figure 2E) and did not transmigrate into the brain parenchyma until day 1 after stroke, which is in line with previous studies.1,4
Discussion
In the present study, with the use of a pharmacologic expansion of Treg, we independently confirm that Treg are strong potentiators of acute ischemic stroke.4 Superagonistic anti-CD28 antibody-induced expansion of Treg positively correlated with increased stroke size 24 hours after ischemia. In analogy to our previous study, Treg interact with endothelial cells and platelets to induce microvascular dysfunction and thrombosis.4 In contrast to our ancestor study4 and the results of this study—both strongly arguing for a detrimental role of Treg during the acute phase of brain ischemia/reperfusion injury—it has been reported that Treg are key modulators of cerebroprotection in brain ischemia in mice in the late phase.5 Therefore, it would be worthwhile to study whether CD28 SA application at later time points influences stroke outcome although Treg expansion with CD28 SA after tMCAO had no impact on the neurological status on day 3 in our hands (Supplementary Figure 4).
Treg promote stroke progression within a few hours after cessation of cerebral blood flow. At this early stage, they are mainly found within cerebral blood vessels and interact with endothelial cells and platelets. This interplay between thrombotic and inflammatory processes has recently been described as ‘thrombo-inflammation'.8 Nevertheless, we also found an increased number of CD4+ and Ly6B.2+ cells in the brain parenchyma as early as day 1 after tMCAO. While it is well-known that neutrophils start to transmigrate into the brain within hours after stroke, the pathophysiologic role of these cells in acute ischemic stroke is still under debate.1,4,14 Moreover, the mechanisms through which peripheral Treg expansion by the CD28 SA enhances neutrophil recruitment to the ischemic brain need to be further established. One potential explanation could include the formation of a pro-inflammatory milieu triggered by activated T cells present in high numbers within the brain vasculature and subsequent upregulation of cell adhesion molecules. However, based on the observation that Treg (and other T cells) trigger ischemic brain damage already after a few hours after vessel occlusion, it can be assumed that mechanisms operating mainly within the brain-perfusing vessels rather than within the brain parenchyma contribute to the detrimental T-cell effect to a large extent.2, 3, 4
In summary, the present study independently confirms that Treg promote lesion growth during the acute stage of ischemic stroke. Short-term inhibition of Treg might become a promising therapeutic approach to combat this devastating neurologic condition.
Acknowledgments
The authors thank Daniela Urlaub, Andrea Sauer, and Melanie Glaser for excellent technical assistance.
The authors declare no conflict of interest.
Footnotes
Supplementary Information accompanies the paper on the Journal of Cerebral Blood Flow & Metabolism website (http://www.nature.com/jcbfm)
This work was supported by the Deutsche Forschungsgemeinschaft (KL2323/6-1, WI1722/8-1, and SFB 688).
Supplementary Material
References
- Gelderblom M, Leypoldt F, Steinbach K, Behrens D, Choe CU, Siler DA, et al. Temporal and spatial dynamics of cerebral immune cell accumulation in stroke. Stroke. 2009;40:1849–1857. doi: 10.1161/STROKEAHA.108.534503. [DOI] [PubMed] [Google Scholar]
- Yilmaz G, Arumugam TV, Stokes KY, Granger DN. Role of T lymphocytes and interferon-gamma in ischemic stroke. Circulation. 2006;113:2105–2112. doi: 10.1161/CIRCULATIONAHA.105.593046. [DOI] [PubMed] [Google Scholar]
- Kleinschnitz C, Schwab N, Kraft P, Hagedorn I, Dreykluft A, Schwarz T, et al. Early detrimental T-cell effects in experimental cerebral ischemia are neither related to adaptive immunity nor thrombus formation. Blood. 2010;115:3835–3842. doi: 10.1182/blood-2009-10-249078. [DOI] [PubMed] [Google Scholar]
- Kleinschnitz C, Kraft P, Dreykluft A, Hagedorn I, Gobel K, Schuhmann MK, et al. Regulatory T cells are strong promoters of acute ischemic stroke in mice by inducing dysfunction of the cerebral microvasculature. Blood. 2013;121:679–691. doi: 10.1182/blood-2012-04-426734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liesz A, Suri-Payer E, Veltkamp C, Doerr H, Sommer C, Rivest S, et al. Regulatory T cells are key cerebroprotective immunomodulators in acute experimental stroke. Nat Med. 2009;15:192–199. doi: 10.1038/nm.1927. [DOI] [PubMed] [Google Scholar]
- Liesz A, Zhou W, Na SY, Hammerling GJ, Garbi N, Karcher S, et al. Boosting regulatory T cells limits neuroinflammation in permanent cortical stroke. J Neurosci. 2013;33:17350–17362. doi: 10.1523/JNEUROSCI.4901-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ren X, Akiyoshi K, Vandenbark AA, Hurn PD, Offner H. CD4+Foxp3+ regulatory T-cells in cerebral ischemic stroke. Metab Brain Dis. 2011;26:87–90. doi: 10.1007/s11011-010-9226-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nieswandt B, Kleinschnitz C, Stoll G. Ischaemic stroke: a thrombo-inflammatory disease. J Physiol. 2011;589:4115–4123. doi: 10.1113/jphysiol.2011.212886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin CH, Hünig T. Efficient expansion of regulatory T cells in vitro and in vivo with a CD28 superagonist. Eur J Immunol. 2003;33:626–638. doi: 10.1002/eji.200323570. [DOI] [PubMed] [Google Scholar]
- Gogishvili T, Langenhorst D, Luhder F, Elias F, Elflein K, Dennehy KM, et al. Rapid regulatory T-cell response prevents cytokine storm in CD28 superagonist treated mice. PloS one. 2009;4:e4643. doi: 10.1371/journal.pone.0004643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lahl K, Loddenkemper C, Drouin C, Freyer J, Arnason J, Eberl G, et al. Selective depletion of Foxp3+ regulatory T cells induces a scurfy-like disease. J Exp Med. 2007;204:57–63. doi: 10.1084/jem.20061852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bederson JB, Pitts LH, Tsuji M, Nishimura MC, Davis RL, Bartkowski H. Rat middle cerebral artery occlusion: evaluation of the model and development of a neurologic examination. Stroke. 1986;17:472–476. doi: 10.1161/01.str.17.3.472. [DOI] [PubMed] [Google Scholar]
- Moran PM, Higgins LS, Cordell B, Moser PC. Age-related learning deficits in transgenic mice expressing the 751-amino acid isoform of human beta-amyloid precursor protein. Proc Natl Acad Sci USA. 1995;92:5341–5345. doi: 10.1073/pnas.92.12.5341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Connolly ES, Jr, Winfree CJ, Springer TA, Naka Y, Liao H, Yan SD, et al. Cerebral protection in homozygous null icam-1 mice after middle cerebral artery occlusion. Role of neutrophil adhesion in the pathogenesis of stroke. J Clin Invest. 1996;97:209–216. doi: 10.1172/JCI118392. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.


