Changes in cerebral autoregulation and blood biomarkers after remote ischemic preconditioning

Zhen-Ni Guo, Wei-Tong Guo, Jia Liu, Junlei Chang, Hongyin Ma, Peng Zhang, Fu-Liang Zhang, Ke Han, Han-Hwa Hu, Hang Jin, Xin Sun, David Martin Simpson, Yi Yang, Zhen-Ni Guo, Wei-Tong Guo, Jia Liu, Junlei Chang, Hongyin Ma, Peng Zhang, Fu-Liang Zhang, Ke Han, Han-Hwa Hu, Hang Jin, Xin Sun, David Martin Simpson, Yi Yang

Abstract

Objective: To determine the effect of remote ischemic preconditioning (RIPC) on dynamic cerebral autoregulation (dCA) and various blood biomarkers in healthy adults.

Methods: A self-controlled interventional study was conducted. Serial measurements of dCA were performed at 7 time points (7, 9, and 11 am; 2, 5, and 8 pm, and 8 am on the next day) without or with RIPC, carried out at 7:20 to 8 am. Venous blood samples were collected at baseline (7 am) and 1 hour after RIPC, and blood biomarkers, including 5 neuroprotective factors and 25 inflammation-related biomarkers, were measured with a quantitative protein chip.

Results: Fifty participants were enrolled (age 34.54 ± 12.01 years, 22 men). Compared with the results on the day without RIPC, dCA was significantly increased at 6 hours after RIPC, and the increase was sustained for at least 24 hours. After RIPC, 2 neuroprotective factors (glial cell-derived neurotrophic factor and vascular endothelial growth factor-A) and 4 inflammation-related biomarkers (transforming growth factor-β1, leukemia inhibitory factor, matrix metallopeptidase-9, and tissue inhibitor of metalloproteinase-1) were significantly elevated compared with their baseline levels. Conversely, monocyte chemoattractant protein-1 was significantly lower compared with its baseline level.

Conclusions: RIPC induces a sustained increase of dCA from 6 to at least 24 hours after treatment in healthy adults. In addition, several neuroprotective and inflammation-related blood biomarkers were differentially regulated shortly after RIPC. The increased dCA and altered blood biomarkers may collectively contribute to the beneficial effects of RIPC on cerebrovascular function.

Clinicaltrialsgov identifier: NCT02965547.

Copyright © 2019 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology.

Figures

Figure 1. Flowcharts and protocols of the…
Figure 1. Flowcharts and protocols of the study
(A) Flowcharts of the study. (B) Study protocols. ABP = arterial blood pressure; DCA = dynamic cerebral autoregulation; HR = heart rate; RIPC = remote ischemic preconditioning.
Figure 2. Autoregulatory parameter and statistical analysis…
Figure 2. Autoregulatory parameter and statistical analysis of dCA, MAP, and HR
(A) Autoregulatory parameter derived from the transfer function analysis. (B) Statistical analysis of dynamic cerebral autoregulation (dCA), mean arterial pressure (MAP), and heart rate (HR) by serial measurements. *p < 0.05 for comparison between the control day and remote ischemic preconditioning (RIPC) day.
Figure 3. Heat map of quantitative protein…
Figure 3. Heat map of quantitative protein chip of 30 biomarkers
Adip = adiponectin; BDNF = brain-derived neurotrophic factor; β-NGF = β-nerve growth factor; CNTF = ciliary neurotrophic factor; CRP = C-reactive protein; EOT = eotaxin; Fas = tumor necrosis factor receptor superfamily member 6; GDNF = glial cell line–derived neurotrophic factor; GM-CSF = granulocyte-macrophage colony-stimulating factor; IFN-γ= interferon-γ; IL = interleukin; LIF = leukemia inhibitory factor; MCP-1 = monocyte chemotactic protein-1; MIP-1β = macrophage inflammatory protein-1β; MMP, matrix metalloproteinase; NX = blood sample number; RIPC = remote ischemic preconditioning; TARC = thymus and activation-regulated chemokine; TGF-β1= transforming growth factor-β1; TIMP-1 = tissue inhibitor of metalloproteinases-1; TNF-α= tumor necrosis factor-α; VEGF-A = vascular endothelial growth factor A.
Figure 4. Statistical distributions of 7 biomarkers…
Figure 4. Statistical distributions of 7 biomarkers with significant differences
(A) Statistical distributions of the neuroprotective factors vascular endothelial growth factor A (VEGF-A) and glial cell line–derived neurotrophic factor (GDNF) at baseline and 1 hour after remote ischemic preconditioning (RIPC) in each group. (B) Statistical distributions of inflammation-related biomarkers transforming growth factor-β1 (TGF-β1), leukemia inhibitory factor (LIF), matrix metalloproteinase-9 (MMP-9), tissue inhibitor of metalloproteinases-1 (TIMP-1), and monocyte chemotactic protein-1 (MCP-1) at baseline and 1 hour after RIPC in each group. Whiskers represent highest and lowest values; middle square represents interquartile values; and middle line indicates median. *Adjusted p < 0.05 for comparison with the baseline level.

References

    1. Heusch G, Bøtker HE, Przyklenk K, Redington A, Yellon D. Remote ischemic conditioning. J Am Coll Cardiol 2015;65:177–195.
    1. White SK, Frohlich GM, Sado DM, et al. . Remote ischemic conditioning reduces myocardial infarct size and edema in patients with ST-segment elevation myocardial infarction. JACC Cardiovasc Interv 2015;8:178–188.
    1. Albrecht M, Zitta K, Bein B, et al. . Remote ischemic preconditioning regulates HIF-1alpha levels, apoptosis and inflammation in heart tissue of cardiosurgical patients: a pilot experimental study. Basic Res Cardiol 2013;108:314.
    1. Verouhis D, Sörensson P, Gourine A, et al. . Effect of remote ischemic conditioning on infarct size in patients with anterior ST-elevation myocardial infarction. Am Heart J 2016;181:66–73.
    1. Wang Y, Meng R, Song H, et al. . Remote ischemic conditioning may improve outcomes of patients with cerebral small-vessel disease. Stroke 2017;48:3064–3072.
    1. Jensen HA, Loukogeorgakis S, Yannopoulos F, et al. . Remote ischemic preconditioning protects the brain against injury after hypothermic circulatory arrest. Circulation 2011;123:714–721.
    1. Hu S, Dong H, Zhang H, et al. . Noninvasive limb remote ischemic preconditioning contributes neuroprotective effects via activation of adenosine A1 receptor and redox status after transient focal cerebral ischemia in rats. Brain Res 2012;1459:81–90.
    1. Blauenfeldt RA, Hougaard KD, Mouridsen K, Andersen G. High prestroke physical activity is associated with reduced infarct growth in acute ischemic stroke patients treated with intravenous tPA and randomized to remote ischemic perconditioning. Cerebrovasc Dis (Basel, Switzerland) 2017;44:88–95.
    1. Pan J, Li X, Peng Y. Remote ischemic conditioning for acute ischemic stroke: dawn in the darkness. Rev Neurosci 2016;27:501–510.
    1. Zhao W, Meng R, Ma C, et al. . Safety and efficacy of remote ischemic preconditioning in patients with severe carotid artery stenosis before carotid artery stenting: a proof-of-concept, randomized controlled trial. Circulation 2017;135:1325–1335.
    1. Xiong L, Liu X, Shang T, et al. . Impaired cerebral autoregulation: measurement and application to stroke. J Neurol Neurosurg Psychiatry 2017;88:520–531.
    1. Reinhard M, Rutsch S, Lambeck J, et al. . Dynamic cerebral autoregulation associates with infarct size and outcome after ischemic stroke. Acta Neurol Scand 2012;125:156–162.
    1. Randhawa PK, Jaggi AS. Unraveling the role of adenosine in remote ischemic preconditioning-induced cardioprotection. Life Sci 2016;155:140–146.
    1. Rassaf T, Totzeck M, Hendgen-Cotta UB, Shiva S, Heusch G, Kelm M. Circulating nitrite contributes to cardioprotection by remote ischemic preconditioning. Circ Res 2014;114:1601–1610.
    1. Takada J, Ibayashi S, Nagao T, Ooboshi H, Kitazono T, Fujishima M. Bradykinin mediates the acute effect of an angiotensin-converting enzyme inhibitor on cerebral autoregulation in rats. Stroke 2001;32:1216–1219.
    1. Guo ZN, Shao A, Tong LS, Sun W, Liu J, Yang Y. The role of nitric oxide and sympathetic control in cerebral autoregulation in the setting of subarachnoid hemorrhage and traumatic brain injury. Mol Neurobiol 2016;53:3606–3615.
    1. Yang J, Liu C, Du X, et al. . Hypoxia inducible factor 1alpha plays a key role in remote ischemic preconditioning against stroke by modulating inflammatory responses in rats. J Am Heart Assoc 2018;7;e007589.
    1. Durukan A, Tatlisumak T. Preconditioning-induced ischemic tolerance: a window into endogenous gearing for cerebroprotection. Exp Transl Stroke Med 2010;2:2.
    1. Claassen JA, Meel-van den Abeelen AS, Simpson DM, Panerai RB. Transfer function analysis of dynamic cerebral autoregulation: a white paper from the International Cerebral Autoregulation Research Network. J Cereb Blood Flow Metab 2016;36:665–680.
    1. Ma H, Guo ZN, Liu J, Xing Y, Zhao R, Yang Y. Temporal course of dynamic cerebral autoregulation in patients with intracerebral hemorrhage. Stroke 2016;47:674–681.
    1. Meller R. The role of the ubiquitin proteasome system in ischemia and ischemic tolerance. Neuroscientist 2009;15:243–260.
    1. Ren C, Gao X, Steinberg GK, Zhao H. Limb remote-preconditioning protects against focal ischemia in rats and contradicts the dogma of therapeutic time windows for preconditioning. Neuroscience 2008;151:1099–1103.
    1. Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor signalling: in control of vascular function. Nat Rev Mol Cell Biol 2006;7:359–371.
    1. Sorond FA, Tan CO, LaRose S, et al. . Deferoxamine, cerebrovascular hemodynamics, and vascular aging: potential role for hypoxia-inducible transcription factor-1-regulated pathways. Stroke 2015;46:2576–2583.
    1. Huang SM, Chen TS, Chiu CM, et al. . GDNF increases cell motility in human colon cancer through VEGF-VEGFR1 interaction. Endocr Relat Cancer 2014;21:73–84.
    1. Yang JP, Liu HJ, Liu XF. VEGF promotes angiogenesis and functional recovery in stroke rats. J Invest Surg 2010;23:149–155.
    1. England TJ, Hedstrom A, O'Sullivan S, et al. . RECAST (Remote Ischemic Conditioning After Stroke Trial): a pilot randomized placebo controlled phase II trial in acute ischemic stroke. Stroke 2017;48:1412–1415.
    1. Duarte EP, Curcio M, Canzoniero LM, Duarte CB. Neuroprotection by GDNF in the ischemic brain. Growth Factors 2012;30:242–257.
    1. Morcuende S, Muñoz-Hernández R, Benítez-Temiño B, Pastor AM, de la Cruz RR. Neuroprotective effects of NGF, BDNF, NT-3 and GDNF on axotomized extraocular motoneurons in neonatal rats. Neuroscience 2013;250:31–48.
    1. McDonough A, Weinstein JR. Neuroimmune response in ischemic preconditioning. Neurotherapeutics 2016;13:748–761.
    1. Ney J, Hoffmann K, Meybohm P, et al. . Remote ischemic preconditioning does not affect the release of humoral factors in propofol-anesthetized cardiac surgery patients: a secondary analysis of the RIPHeart study. Int J Mol Sci 2018;19:E1094.
    1. Shimizu M, Saxena P, Konstantinov IE, et al. . Remote ischemic preconditioning decreases adhesion and selectively modifies functional responses of human neutrophils. J Surg Res 2010;158:155–161.
    1. Iadecola C, Anrather J. The immunology of stroke: from mechanisms to translation. Nat Med 2011;17:796–808.
    1. Maddahi A, Chen Q, Edvinsson L. Enhanced cerebrovascular expression of matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 via the MEK/ERK pathway during cerebral ischemia in the rat. BMC Neurosci 2009;10:56.
    1. Li DD, Song JN, Huang H, et al. . The roles of MMP-9/TIMP-1 in cerebral edema following experimental acute cerebral infarction in rats. Neurosci Lett 2013;550:168–172.
    1. Douma LG, Gumz ML. Circadian clock-mediated regulation of blood pressure. Free Radic Biol Med 2018;119:108–114.
    1. Rana OR, Schauerte P, Hommes D, et al. . Mechanical stretch induces nerve sprouting in rat sympathetic neurocytes. Auton Neurosci 2010;155:25–32.
    1. Ho GJ, Drego R, Hakimian E, Masliah E. Mechanisms of cell signaling and inflammation in Alzheimer's disease. Curr Drug Targets Inflamm Allergy 2005;4:247–256.
    1. Yilmaz G, Arumugam TV, Stokes KY, Granger DN. Role of T lymphocytes and interferon-gamma in ischemic stroke. Circulation 2006;113:2105–2112.
    1. Arakelyan A, Petrkova J, Hermanova Z, Boyajyan A, Lukl J, Petrek M. Serum levels of the MCP-1 chemokine in patients with ischemic stroke and myocardial infarction. Mediators Inflamm 2005;2005:175–179.
    1. Zhu M, Allard JS, Zhang Y, et al. . Age-related brain expression and regulation of the chemokine CCL4/MIP-1beta in APP/PS1 double-transgenic mice. J Neuropathol Exp Neurol 2014;73:362–374.
    1. Schulz S, Lüdike H, Lierath M, et al. . C-reactive protein levels and genetic variants of CRP as prognostic markers for combined cardiovascular endpoint (cardiovascular death, death from stroke, myocardial infarction, and stroke/TIA). Cytokine 2016;88:71–76.
    1. Spath S, Komuczki J, Hermann M, et al. . Dysregulation of the cytokine GM-CSF induces spontaneous phagocyte invasion and immunopathology in the central nervous system. Immunity 2017;46:245–260.
    1. Suzuki Y, Nagai N, Umemura K, Collen D, Lijnen HR. Stromelysin-1 (MMP-3) is critical for intracranial bleeding after t-PA treatment of stroke in mice. J Thromb Haemost 2007;5:1732–1739.
    1. Owczarek W, Paplińska M, Targowski T, et al. . Analysis of eotaxin 1/CCL11, eotaxin 2/CCL24 and eotaxin 3/CCL26 expression in lesional and non-lesional skin of patients with atopic dermatitis. Cytokine 2010;50:181–185.
    1. Gairolla J, Kler R, Modi M, Khurana D. Leptin and adiponectin: pathophysiological role and possible therapeutic target of inflammation in ischemic stroke. Rev Neurosci 2017;28:295–306.
    1. Chollangi S, Wang J, Martin A, Quinn J, Ash JD. Preconditioning-induced protection from oxidative injury is mediated by leukemia inhibitory factor receptor (LIFR) and its ligands in the retina. Neurobiol Dis 2009;34:535–544.
    1. Garlisi CG, Xiao H, Tian F, et al. . The assignment of chemokine-chemokine receptor pairs: TARC and MIP-1 beta are not ligands for human CC-chemokine receptor 8. Eur J Immunol 1999;29:3210–3215.

Source: PubMed

3
Abonnieren