Isolation and identification of leukocyte populations in intracranial blood collected during mechanical thrombectomy

Benjamin C Shaw, G Benton Maglinger, Thomas Ujas, Chintan Rupareliya, Justin F Fraser, Stephen Grupke, Melissa Kesler, Mathias Gelderblom, Keith R Pennypacker, Jadwiga Turchan-Cholewo, Ann M Stowe, Benjamin C Shaw, G Benton Maglinger, Thomas Ujas, Chintan Rupareliya, Justin F Fraser, Stephen Grupke, Melissa Kesler, Mathias Gelderblom, Keith R Pennypacker, Jadwiga Turchan-Cholewo, Ann M Stowe

Abstract

Using standard techniques during mechanical thrombectomy, the Blood and Clot Thrombectomy Registry and Collaboration (BACTRAC) protocol (NCT03153683) isolates intracranial arterial blood distal to the thrombus and proximal systemic blood in the carotid artery. We augmented the current protocol to study leukocyte subpopulations both distal and proximal to the thrombus during human stroke (n = 16 patients), and from patients with cerebrovascular disease (CVD) undergoing angiography for unrelated conditions (e.g. carotid artery stenosis; n = 12 patients). We isolated leukocytes for flow cytometry from small volume (<1 mL) intracranial blood and systemic blood (5-10 mL) to identify adaptive and innate leukocyte populations, in addition to platelets and endothelial cells (ECs). Intracranial blood exhibited significant increases in T cell representation and decreases in myeloid/macrophage representation compared to within-patient carotid artery samples. CD4+ T cells and classical dendritic cells were significantly lower than CVD controls and correlated to within-patient edema volume and last known normal. This novel protocol successfully isolates leukocytes from small volume intracranial blood samples of stroke patients at time of mechanical thrombectomy and can be used to confirm preclinical results, as well as identify novel targets for immunotherapies.

Keywords: Cerebrovascular endothelium; T cells; emergent large vessel occlusion (ELVO); flow cytometry; ischemic stroke.

Conflict of interest statement

Declaration of conflicting interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Figures

Figure 1.
Figure 1.
Cell yield and viability. (a) Viability of cells is independent of volume processed for 1 mL volumes (squares) and 9 mL volumes (circles). F1,44 = 0.67, p = 0.42 for volume; F1,44 = 0.04, p = 0.84 for injury status; n = 10 controls (white symbols); n = 14 stroke patients (red/blue symbols). (b) Significant viability loss occurred during the cryopreservation process (clear bars) compared to viability prior to banking (hatched bars). * p < 0.05; **** p < 0.0001; n = 12 controls (clear circles), n = 16 stroke (filled circles, systemic; squares, intracranial), n = 12 cerebrovascular disease patients (triangles). One intracranial sample was unreadable using the Nexcelom Cellometer after recovery from cryopreservation but did have viable cells by flow cytometry. (c) Correlation between last known normal and % viability in intracranial blood at time of collection does not show an effect of time to mechanical thrombectomy on leukocyte yield; F1,11 = 1.81, p = 0.21. r2 = 0.14.
Figure 2.
Figure 2.
Distribution of tSNE-identified leukocyte populations. (a) tSNE dimensionality reduction and clustering for the general immune panel including innate and adaptive leukocytes. (b) Cell populations identified in A are color coded. (c–f) tSNE overlays of total (grey) and healthy venous control (c), cerebrovascular disease (CVD) control (d), systemic stroke sample (e), and intracranial stroke sample (f). Note the striking differences between systemic and intracranial density localizations, and how they resemble either healthy control or CVD control, respectively.
Figure 3.
Figure 3.
Leukocyte changes in intracranial blood prior to thrombectomy. (a–e) All panels show individual healthy subject control venous populations (dashed line), stroke patient systemic arterial stroke samples (red circles; grey bars), intracranial arterial stroke samples (blue squares; grey bars), and cerebrovascular disease (CVD) control patient samples (green triangles; white bar). Subpopulations for (a) B cells, (b) T cells, (c) innate subpopulations, (d) natural killer (NK) populations, and (e) dendritic cells (DC) are shown as the percent of total live CD45+ events and respective subgating on the y axis. *p < 0.05 for within-patient paired t-test; †p < 0.05; ††p < 0.01; †††p < 0.001 from CVD. Control n = 12, stroke n = 16, CVD n = 12. Data also shown as Table 2. (F-H) Results from within-patient Spearman Correlation matrix found association between (f) intracranial CD4+ T cells increasing with edema volume, as well as (g) systemic NK T cells increasing with patient BMI, and (h) systemic classical DCs (cDCs) decreasing with time from last known normal (LKN).
Figure 4.
Figure 4.
Thrombus, platelet, and endothelial cell analyses. (a) There is a significant inverse correlation between systemic granulocyte populations and neutrophils per clot field. Representative 40X images of (b) thrombus of Patient 99 (left panel; high systemic granulocytes, but low neutrophil count in thrombus) vs. Patient 127 (right panel; low systemic granulocytes, but high neutrophil count in thrombus). (c) Platelet populations are not significantly different between systemic and intracranial samples. Control venous (black circles), systemic arterial stroke samples (red squares) and intracranial arterial stroke samples (blue triangles). (d) Endothelial cell populations are not significantly different between systemic and intracranial samples. (e) Dot plot of systemic CD31 gating (x axes) for systemic and intracranial blood of Patient 112. Note the lack of CD31+CD45- events in systemic blood (left panel). Patient 112 shows a substantial CD31+CD45- endothelial cell population in intracranial blood (right panel). (f) Patient 112 initial CT angiogram shows vessel occlusion (right panel, black circle). (g) This patient later underwent severe hemorrhagic transformation from Day 1 to Day 6 post-thrombectomy (red elipse).

References

    1. Fransen PS, Beumer D, Berkhemer OA, et al.. MR CLEAN, a multicenter randomized clinical trial of endovascular treatment for acute ischemic stroke in The Netherlands: study protocol for a randomized controlled trial. Trials 2014; 15: 343.
    1. Saver JL, Goyal M, Bonafe A, et al.. Solitaire with the intention for thrombectomy as primary endovascular treatment for acute ischemic stroke (SWIFT PRIME) trial: protocol for a randomized, controlled, multicenter study comparing the solitaire revascularization device with IV tPA with IV tPA alone in acute ischemic stroke. Int J Stroke 2015; 10: 439–448.
    1. Campbell BC, Mitchell PJ, Kleinig TJ, et al.. Endovascular therapy for ischemic stroke with perfusion-imaging selection. N Engl J Med 2015; 372: 1009–1018.
    1. Jovin TG, Chamorro A, Cobo E, et al.. Thrombectomy within 8 hours after symptom onset in ischemic stroke. N Engl J Med 2015; 372: 2296–2306.
    1. Goyal M, Demchuk AM, Menon BK, et al.. Randomized assessment of rapid endovascular treatment of ischemic stroke. N Engl J Med 2015; 372: 1019–1030.
    1. Berkhemer OA, Fransen PS, Beumer D, et al.. A randomized trial of intraarterial treatment for acute ischemic stroke. N Engl J Med 2015; 372: 11–20.
    1. Nogueira RG, Jadhav AP, Haussen DC, et al.. Thrombectomy 6 to 24 hours after stroke with a mismatch between deficit and infarct. N Engl J Med 2018; 378: 11–21.
    1. Malhotra K, Gornbein J, Saver JL. Ischemic strokes due to large-vessel occlusions contribute disproportionately to stroke-related dependence and death: a review. Front Neurol 2017; 8: 651–19.
    1. Boeckh-Behrens T, Kleine JF, Zimmer C, et al.. Thrombus histology suggests cardioembolic cause in cryptogenic stroke. Stroke 2016; 47: 1864–1871.
    1. Boeckh-Behrens T, Schubert M, Forschler A, et al.. The impact of histological clot composition in embolic stroke. Clin Neuroradiol 2016; 26: 189–197.
    1. Almekhlafi MA, Hu WY, Hill MD, et al.. Calcification and endothelialization of thrombi in acute stroke. Ann Neurol 2008; 64: 344–348.
    1. Liebeskind DS, Sanossian N, Yong WH, et al.. CT and MRI early vessel signs reflect clot composition in acute stroke. Stroke 2011; 42: 1237–1243.
    1. Marder VJ, Chute DJ, Starkman S, et al.. Analysis of thrombi retrieved from cerebral arteries of patients with acute ischemic stroke. Stroke 2006; 37: 2086–2093.
    1. Simons N, Mitchell P, Dowling R, et al.. Thrombus composition in acute ischemic stroke: a histopathological study of thrombus extracted by endovascular retrieval. J Neuroradiol 2015; 42: 86–92.
    1. Hashimoto T, Hayakawa M, Funatsu N, et al.. Histopathologic analysis of retrieved thrombi associated with successful reperfusion after acute stroke thrombectomy. Stroke 2016; 47: 3035–3037.
    1. Fraser JF, Collier LA, Gorman AA, et al. The Blood And Clot Thrombectomy Registry And Collaboration (BACTRAC) protocol: novel method for evaluating human stroke. J Neurointerv Surg 2019; 11: 265–270.
    1. Martha SR, Fraser JF, Pennypacker KR. Acid-Base and electrolyte changes drive early pathology in ischemic stroke. Neuromolecular Med. Epub ahead of print 8 July 2019. DOI: 10.1007/s12017-019-08555-5.
    1. Martha SR, Cheng Q, Fraser JF, et al. Expression of Cytokines and Chemokines as Predictors of Stroke Outcomes in Acute Ischemic Stroke. Front Neurol 2020; 10: 1391. doi: 10.3389/fneur.2019.01391
    1. Maglinger B, Frank JA, McLouth CJ, et al.. Proteomic changes in intracranial blood during human ischemic stroke. J Neurointerv Surg. Epub ahead of print 8 July 2020. DOI: 10.1136/neurintsurg-2020-016118.
    1. Maglinger B, Sands M, Frank JA, et al.. Intracranial VCAM1 at time of mechanical thrombectomy predicts ischemic stroke severity. J Neuroinflammation 2021; 18: 109.
    1. Ortega SB, Pandiyan P, Windsor J, et al.. A pilot study identifying brain-targeting adaptive immunity in pediatric extracorporeal membrane oxygenation patients with acquired brain injury. Crit Care Med 2019; 47: e206–e213.
    1. Monaco G, Chen H, Poidinger M, et al.. flowAI: automatic and interactive anomaly discerning tools for flow cytometry data. Bioinformatics 2016; 32: 2473–2480.
    1. Chamorro A, Meisel A, Planas AM, et al.. The immunology of acute stroke. Nat Rev Neurol 2012; 8: 401–410.
    1. Iadecola C, andAnrather J.. The immunology of stroke: from mechanisms to translation. Nat Med 2011; 17: 796–808.
    1. Macrez R, Ali C, Toutirais O, et al.. Stroke and the immune system: from pathophysiology to new therapeutic strategies. Lancet Neurol 2011; 10: 471–480.
    1. Gelderblom M, Leypoldt F, Steinbach K, et al.. Temporal and spatial dynamics of cerebral immune cell accumulation in stroke. Stroke 2009; 40: 1849–1857.
    1. Wang J, Xie L, Yang C, et al.. Activated regulatory T cell regulates neural stem cell proliferation in the subventricular zone of normal and ischemic mouse brain through interleukin 10. Front Cell Neurosci 2015; 9: 361.
    1. Ritzel RM, Crapser J, Patel AR, et al.. Age-associated resident memory CD8 T cells in the Central nervous system are primed to potentiate inflammation after ischemic brain injury. J Immunol 2016; 196: 3318–3330.
    1. Liesz A, Zhou W, Mracsko E, et al.. Inhibition of lymphocyte trafficking shields the brain against deleterious neuroinflammation after stroke. Brain 2011; 134: 704–720.
    1. Shichita T, Sugiyama Y, Ooboshi H, et al.. Pivotal role of cerebral interleukin-17-producing gammadeltaT cells in the delayed phase of ischemic brain injury. Nat Med 2009; 15: 946–950.
    1. Bodhankar S, Chen Y, Vandenbark AA, et al.. IL-10-producing B-cells limit CNS inflammation and infarct volume in experimental stroke. Metab Brain Dis 2013; 28: 375–386.
    1. Ortega SB, Torres VO, Latchney SE, et al.. B cells migrate into remote brain areas and support neurogenesis and functional recovery after focal stroke in mice. Proc Natl Acad Sci U S A 2020; 117: 4983–4993.
    1. Heindl S, Ricci A, Carofiglio O, et al.. Chronic T cell proliferation in brains after stroke could interfere with the efficacy of immunotherapies. J Exp Med 2021; 218: 218: e20202411. doi: .
    1. Li S, Huang Y, Liu Y, et al.. Change and predictive ability of circulating immunoregulatory lymphocytes in long-term outcomes of acute ischemic stroke. J Cereb Blood Flow Metab. Epub ahead of print 2 March 2021. DOI: 10.1177/0271678X21995694.
    1. Mansilla MJ, Navarro-Barriuso J, Presas-Rodriguez S, et al.. Optimal response to dimethyl fumarate is mediated by a reduction of Th1-like Th17 cells after 3 months of treatment. CNS Neurosci Ther 2019; 25: 995–1005.
    1. Neal EG, Acosta SA, Kaneko Y, et al.. Regulatory T-cells within bone marrow-derived stem cells actively confer immunomodulatory and neuroprotective effects against stroke. J Cereb Blood Flow Metab 2019; 39: 1750–1758.
    1. Stubbe T, Ebner F, Richter D, et al.. Regulatory T cells accumulate and proliferate in the ischemic hemisphere for up to 30 days after MCAO. J Cereb Blood Flow Metab 2013; 33: 37–47.
    1. Xie L, Li W, Hersh J, et al.. Experimental ischemic stroke induces long-term T cell activation in the brain. J Cereb Blood Flow Metab 2019; 39: 2268–2276.
    1. Martha SR, Collier LA, Davis SM, et al.. Evaluation of sex differences in acid/base and electrolyte concentrations in acute large vessel stroke. Exp Neurol 2020; 323: 113078.
    1. Martha SR, Cheng Q, Fraser JF, et al.. Expression of cytokines and chemokines as predictors of stroke outcomes in acute ischemic stroke. Front Neurol 2019; 10: 1391.
    1. Deniset JF, Surewaard BG, Lee WY, et al.. Splenic Ly6G(high) mature and Ly6G(int) immature neutrophils contribute to eradication of S. pneumoniae. J Exp Med 2017; 214: 1333–1350.
    1. Kollikowski AM, Schuhmann MK, Nieswandt B, et al.. Local leukocyte invasion during hyperacute human ischemic stroke. Ann Neurol 2020; 87: 466–479.
    1. Zuo Y, Estes SK, Ali RA, et al.. Prothrombotic autoantibodies in serum from patients hospitalized with COVID-19. Sci Transl Med 2020; 12: eabd3876.
    1. Semerano A, Laredo C, Zhao Y, et al.. Leukocytes, collateral circulation, and reperfusion in ischemic stroke patients treated with mechanical thrombectomy. Stroke 2019; 50: 3456–3464.
    1. Jaff N, Grankvist R, Muhl L, et al.. Transcriptomic analysis of the harvested endothelial cells in a swine model of mechanical thrombectomy. Neuroradiology 2018; 60: 759–768.
    1. Woywodt A, Streiber F, de Groot K, et al.. Circulating endothelial cells as markers for ANCA-associated small-vessel vasculitis. Lancet 2003; 361: 206–210.
    1. Boulouis G, de Boysson H, Zuber M, et al.. Primary angiitis of the central nervous system: magnetic resonance imaging spectrum of parenchymal, meningeal, and vascular lesions at baseline. Stroke 2017; 48: 1248–1255.

Source: PubMed

3
구독하다