Remote ischemic post-conditioning promotes hematoma resolution via AMPK-dependent immune regulation

Kumar Vaibhav, Molly Braun, Mohammad Badruzzaman Khan, Sumbul Fatima, Nancy Saad, Adarsh Shankar, Zenab T Khan, Ruth B S Harris, Qiuhua Yang, Yuqing Huo, Ali S Arbab, Shailendra Giri, Cargill H Alleyne Jr, John R Vender, David C Hess, Babak Baban, Md Nasrul Hoda, Krishnan M Dhandapani, Kumar Vaibhav, Molly Braun, Mohammad Badruzzaman Khan, Sumbul Fatima, Nancy Saad, Adarsh Shankar, Zenab T Khan, Ruth B S Harris, Qiuhua Yang, Yuqing Huo, Ali S Arbab, Shailendra Giri, Cargill H Alleyne Jr, John R Vender, David C Hess, Babak Baban, Md Nasrul Hoda, Krishnan M Dhandapani

Abstract

Spontaneous intracerebral hemorrhage (ICH) produces the highest acute mortality and worst outcomes of all stroke subtypes. Hematoma volume is an independent determinant of ICH patient outcomes, making clot resolution a primary goal of clinical management. Herein, remote-limb ischemic post-conditioning (RIC), the repetitive inflation-deflation of a blood pressure cuff on a limb, accelerated hematoma resolution and improved neurological outcomes after ICH in mice. Parabiosis studies revealed RIC accelerated clot resolution via a humoral-mediated mechanism. Whereas RIC increased anti-inflammatory macrophage activation, myeloid cell depletion eliminated the beneficial effects of RIC after ICH. Myeloid-specific inactivation of the metabolic regulator, AMPKα1, attenuated RIC-induced anti-inflammatory macrophage polarization and delayed hematoma resolution, providing a molecular link between RIC and immune activation. Finally, chimera studies implicated myeloid CD36 expression in RIC-mediated neurological recovery after ICH. Thus, RIC, a clinically well-tolerated therapy, noninvasively modulates innate immune responses to improve ICH outcomes. Moreover, immunometabolic changes may provide pharmacodynamic blood biomarkers to clinically monitor the therapeutic efficacy of RIC.

© 2018 Vaibhav et al.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
RIC promotes delayed hematoma resolution and improves outcomes after ICH. (A) Mixed sex C57BL/6J littermates were randomized to receive once-daily mock conditioning or bilateral RIC beginning at 2 h after sham or collagenase-induced ICH. At day 5, hematoma area was quantified in serial 2-mm coronal slices. Bar, 4 mm. Data are mean ± SEM from n = 8 mice/group and were analyzed by one-way ANOVA followed by Tukey’s post-hoc test (***, P < 0.001). Data are representative of two independent experiments. (B) Mixed sex C57BL/6J littermates were randomized to receive once-daily mock conditioning or bilateral RIC beginning at 2 h after sham or collagenase-induced ICH. At day 5, peri-hematoma blood flow was assessed by laser speckle contrast imaging. Bar, 4 mm. Data are mean ± SEM from n = 8 mice/group and were analyzed by one-way ANOVA followed by Tukey’s post-hoc test (***, P < 0.001; ns, not statistically significant). Data are representative of two independent experiments. (C and D) Mixed sex C57BL/6J littermates were randomized to receive once-daily mock conditioning or bilateral RIC beginning at 2 h after sham or intrastriatal placement of 30 µl autologous blood. Littermates were randomized to receive once-daily mock conditioning or bilateral RIC beginning at 2 h after ICH. At day 3 and 6 after injury, hematoma area was quantified in serial 2-mm coronal slices. Bar, 4 mm. Data are mean ± SEM from n = 5–10 mice/group and were analyzed by one-way ANOVA followed by Tukey’s post-hoc test (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not statistically significant). Data are representative of two independent experiments.
Figure 2.
Figure 2.
RIC improves neurobehavioral outcomes after ICH. Mixed sex C57BL/6J littermates were randomized to receive once-daily mock conditioning or bilateral RIC beginning at 2 h after sham or collagenase-induced ICH. (A) Neurological outcomes were assessed at day 4–5 after injury, using a composite focal deficits score (a measure of global injury severity), the elevated body swing test (a task of asymmetric motor behavior), and by the narrow beam test (a measure of motor balance and coordination). Data are mean ± SEM from n = 10 mice/group and were analyzed by one-way ANOVA followed by Tukey’s post-hoc test (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not statistically significant). Data are representative of two independent experiments. (B) Mixed sex C57BL/6J littermates were randomized to receive once-daily mock conditioning or bilateral RIC beginning at 2 h after sham or collagenase-induced ICH. At day 4 after injury, neurological outcomes were assessed using the open field test. Representative heat maps are depicted, and frequency in the center zone, distance traveled, and movement velocity were quantified. Data are mean ± SEM from n = 10 mice/group and were analyzed by one-way ANOVA followed by Tukey’s post-hoc test (*, P < 0.05; ns, not statistically significant). Data are representative of two independent experiments. (C) Mixed sex C57BL/6J littermates were randomized to receive once-daily mock conditioning or bilateral RIC beginning at 2 h after sham or intrastriatal placement of 30 µl autologous blood. Neurobehavioral outcomes, including the narrow beam test and grip strength test, were assessed at day 6 after injury. Data are mean ± SEM from n = 5–10 mice/group and were analyzed by one-way ANOVA followed by Tukey’s post-hoc test (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not statistically significant). Data are representative of two independent experiments.
Figure 3.
Figure 3.
Humoral factors mediate RIC-induced hematoma resolution. (A) Parabiotic pairs (n = 6 pairs/group) were generated using age-matched male C57BL/6J mice and randomized to treatment arms, as illustrated. In all pairs, the left pair mate was treated with either once-daily mock conditioning or RIC for 5 d without injury. The right pair mate was untreated and received either a sham injury or ICH. (B) Photograph of a representative parabiotic pair (left). Mock conditioning or RIC was performed in the left pair mate and sham/ICH was performed on the right pair mate, as indicated. Representative MRI brain scans at day 5 after ICH (right). The brain on the left side is uninjured, and the perimeter of the hematoma is indicated by a red dotted line in the brain on the right side. Bar, 2 mm. (C) Quantification of hematoma volume, as assessed by MRI. Data are mean ± SEM from n = 6 pairs/treatment group. Data are representative of two independent experiments and were analyzed by Student’s t test (**, P < 0.01).
Figure 4.
Figure 4.
Macrophage activation is required for RIC-induced hematoma resolution after ICH. (A) Control liposomes (encapsome) or clodronate liposomes (clodrosome) were administered intraperitoneally to mixed sex C57BL/6J littermates for three consecutive days to deplete myeloid cells. Monocyte/granulocyte populations were identified using FSC/side scatter (SSC) and myeloid cell depletion was quantified in CD11b+, F4/80+ myeloid cells. Inset numbers indicate the percentage of total blood cells. Data are representative of n = 8 mice/group from two independent experiments. (B) Mixed sex C57BL/6J littermates were administered either encapsome or clodrosome for three consecutive days, as indicated in A. Mice were then randomized to receive once-daily mock conditioning or bilateral RIC beginning at 2 h after collagenase-induced ICH. Hematoma volume and edema volume were quantified by MRI at day 5 after ICH. Bar, 4 mm. Data are mean ± SEM from n = 8/group and were analyzed using a Student’s t test (**, P < 0.01; ns, not statistically significant). Data are representative of two independent experiments.
Figure 5.
Figure 5.
RIC promotes anti-inflammatory macrophage polarization. (A and B) CFSE-labeled donor macrophages were intravenously administered (5 × 105 cells/recipient) to male C57BL/6J littermates at 1 h after sham or collagenase-induced ICH. Mice were randomized to receive once-daily mock or RIC beginning at 2 h after injury, and both blood and brain tissue were collected at 72 h after sham/ICH. Live cells were gated according to the FSC/SSC profile and further gating was performed to detect CD11b+, CFSE+ cells populations. The effect of RIC on the phenotype of blood and brain macrophages were further defined as anti-inflammatory (CFSE+, CD11b+, F4/80+, CD206+, IL-10+) or pro-inflammatory (CFSE+, CD11b+, CD206−, F4/80+, TNF-α+). Data are mean ± SEM from n = 6 mice/group and were analyzed by one-way ANOVA followed by Tukey’s post-hoc test (**, P < 0.01; ****, P < 0.0001; ns, not statistically significant). Data are representative of two independent experiments. (C) Graphic representation of the ratio of pro-inflammatory/anti-inflammatory macrophages, as determined in B. (D) Representative histograms and graphic representation of the ratio of pro-inflammatory/anti-inflammatory macrophages in blood collected from parabiotic pairs, as defined in A. Data are mean ± SEM from n = 6 pairs/group and are representative of two independent experiments.
Figure 6.
Figure 6.
RIC induces anti-inflammatory macrophage polarization after ICH. (A and B) Mixed sex C57BL/6J littermates were randomized to receive once-daily mock conditioning or bilateral RIC beginning at 2 h after sham or collagenase-induced ICH. Phenotypic analysis of macrophages was performed in peri-hematoma brain tissue (0.2 g) or blood (B; 200 µl) at 72 h after sham, ICH with mock conditioning (ICH), or ICH+RIC. Live cells were gated using FSC/SSC and CD11b+F4/80+ cells were further selected. CD11b+CD45hi-infiltrated myeloid cells and CD11b+CD45low residential myeloid cells were selected from brain tissue, or CD11b+CD68+ macrophages were selected from blood. Selected populations were further defined using F4/80, CD36, MerTK, MHC-II, CD206, Ly-6C, Ly-6G, or IL-10. Representative histograms are provided for each marker. Gray shaded areas indicate isotype controls. Quantified data, which are expressed as the mean ± SEM from n = 6 mice/group, were analyzed using a one-way ANOVA followed by Tukey’s post-hoc test (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, not statistically significant) and are representative of two independent experiments.
Figure 7.
Figure 7.
Myeloid AMPKα1 mediates hematoma resolution after RIC. (A) Western blot analysis of AMPKα1 and AMPKα2 subunits in microglia, macrophages, CD4+ T cells, and liver (positive control). β-actin was used as a loading control. (B) Phosphorylated AMPKα1/2 (p-AMPKα1/2) and total AMPKα were assessed in brain tissue of male C57BL/6J mice at day 5 after sham/ICH by Western blotting. Densitometry analysis, which are expressed as the mean ± SEM from n = 6 mice/group, were analyzed using a one-way ANOVA followed by Tukey’s post-hoc test (***, P < 0.001; ns, not statistically significant) and are representative of two independent experiments. (C) Phosphorylated AMPKα1 (p-AMPKα1) was assessed in brain tissue of male C57BL/6J mice at day 5 after sham/ICH by flow cytometry. Data are expressed as the mean ± SEM from n = 6 mice/group and were analyzed using a one-way ANOVA followed by Tukey’s post-hoc test (**, P < 0.01; ns, not statistically significant). Data are representative of two independent experiments. (D) Mixed sex WT (AMPKα1f/f) or myeloid-specific AMPKα1 knockout (LysMCreAMPKα1f/f) littermates were randomized to sham/collagenase-induced ICH groups and received either mock conditioning or RIC beginning at 2 h after injury. At 72 h after injury, macrophages were phenotypically defined in peri-hematoma brain tissue (0.2 g) after sham, sham + RIC, ICH with mock conditioning, or ICH+RIC. Live cells were gated using FSC/SSC and CD11b+F4/80+ cells were selected. CD11b+CD45hi-infiltrated macrophages were further defined using F4/80, CD36, MerTK, MHC-II, CD206, Ly-6G, or IL-10. Representative histograms are provided for each marker and isotype controls are indicated by gray shaded areas. Data from two independent experiments are expressed as the mean ± SEM from n = 6 mice/group and were analyzed using a one-way ANOVA followed by Tukey’s post-hoc test (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, not statistically significant). (E) Mixed sex AMPKα1f/f or LysMCreAMPKα1f/f littermates were randomized to sham/collagenase-induced ICH groups and received either mock conditioning or RIC beginning at 2 h after injury. At day 5 after sham/ICH, hematoma volume and edema were assessed by MRI. Representative images are shown and quantified data are mean ± SEM from n = 8 mice/group. Bar, 4 mm. Data were analyzed using a one-way ANOVA followed by Tukey’s post-hoc test (**, P < 0.01; ***, P < 0.001; ns, not statistically significant) and are representative of three independent experiments.
Figure 8.
Figure 8.
Myeloid CD36 mediates hematoma resolution by RIC. (A) Schematic depicting the generation of myeloid-specific CD36 knockout mice using an irradiation bone marrow chimera approach. Male WT (C57BL/6J) or CD36−/− mice were irradiated and reconstituted using bone marrow from either genotype, as indicated. Flow cytometric analysis of blood (200 µl) collected from mice at 4 wk after bone marrow transplant, showing successful generation of chimeric mice following transplantation of CD36−/− bone marrow into irradiated WT mice (CD36−/− >> WT) or transplantation of WT bone marrow into irradiated CD36−/− mice (WT >> CD36−/−). Inset numbers represent the percentage of CD36+, CD11b+ myeloid cells. (B) WT >> CD36−/− mice and CD36−/− >> WT mice were randomized to sham/ICH groups and received either mock conditioning or RIC. At day 5 after sham/ICH, MRI was used to assess hematoma volume and cerebral edema. Bar, 4 mm. Representative images are depicted and data are expressed as mean ± SEM from n = 8 mice/group. Data were analyzed using a one-way ANOVA followed by Tukey’s post-hoc test (*, P < 0.05; **, P < 0.01; ns = not statistically significant) and are representative of two independent experiments.
Figure 9.
Figure 9.
Myeloid-specific deletion of CD36 increases long-term white matter injury after ICH. (A) Mixed sex C57BL/6J littermates were randomized to receive once-daily mock conditioning or bilateral RIC beginning at 2 h after sham or collagenase-induced ICH. White matter was assessed in coronal brain sections by LFB staining at 8 wk after injury. Bar, 1 mm. (B) Sequential micrographs (20× magnification) demonstrate a sparing effect of RIC on white matter after ICH. Bar, 100 µm. Roman numerals (i.vii.) denote anatomical locations, as indicated in C. Bar, 1 mm. Data are representative of n = 5–7 mice/group from two independent experiments. (D) White matter loss was quantified by mean integrated density and LFB grading score in regions adjacent to the clot location, including the lateral corpus callosum, fimbria, and internal capsule. Representative tissue sections are shown in B (*, P < 0.05; **, P < 0.01; ***, P < 0.001). (E) Male WT >> CD36−/− and CD36−/− >> WT chimera mice were generated, as in Figure 8, randomized to sham/ICH groups, and received either mock conditioning or RIC. At 8 wk after injury, white matter loss was quantified by mean integrated density in selected brain regions. Data are representative of n = 6 mice/group from two independent experiments and were analyzed using a one-way ANOVA followed by Tukey’s post-hoc test (*, P < 0.05; ***, P < 0.001; ns, not statistically significant). (F) Ventricular enlargement, a common manifestation of white matter loss, was quantified by fluid attenuation inversion recovery at 1 mo after ICH in WT >> CD36−/− and CD36−/− >> WT chimera mice. Data are mean ± SEM from n = 5–7/mice group and were analyzed using a one-way ANOVA followed by Tukey’s post-hoc test. Data are representative of two independent experiments.

References

    1. Andreka G., Vertesaljai M., Szantho G., Font G., Piroth Z., Fontos G., Juhasz E.D., Szekely L., Szelid Z., Turner M.S., et al. . 2007. Remote ischaemic postconditioning protects the heart during acute myocardial infarction in pigs. Heart. 93:749–752. 10.1136/hrt.2006.114504
    1. Barrett R.J., Hussain R., Coplin W.M., Berry S., Keyl P.M., Hanley D.F., Johnson R.R., and Carhuapoma J.R.. 2005. Frameless stereotactic aspiration and thrombolysis of spontaneous intracerebral hemorrhage. Neurocrit. Care. 3:237–245. 10.1385/NCC:3:3:237
    1. Bhasin R.R., Xi G., Hua Y., Keep R.F., and Hoff J.T.. 2002. Experimental intracerebral hemorrhage: effect of lysed erythrocytes on brain edema and blood-brain barrier permeability. Acta Neurochir. Suppl. (Wien). 81:249–251.
    1. Biswas S.K., and Mantovani A.. 2012. Orchestration of metabolism by macrophages. Cell Metab. 15:432–437. 10.1016/j.cmet.2011.11.013
    1. Bøtker H.E., Kharbanda R., Schmidt M.R., Bøttcher M., Kaltoft A.K., Terkelsen C.J., Munk K., Andersen N.H., Hansen T.M., Trautner S., et al. . 2010. Remote ischaemic conditioning before hospital admission, as a complement to angioplasty, and effect on myocardial salvage in patients with acute myocardial infarction: a randomised trial. Lancet. 375:727–734. 10.1016/S0140-6736(09)62001-8
    1. Braun M., Vaibhav K., Saad N., Fatima S., Brann D.W., Vender J.R., Wang L.P., Hoda M.N., Baban B., and Dhandapani K.M.. 2017. Activation of Myeloid TLR4 Mediates T Lymphocyte Polarization after Traumatic Brain Injury. J. Immunol. 198:3615–3626. 10.4049/jimmunol.1601948
    1. Broderick J., Connolly S., Feldmann E., Hanley D., Kase C., Krieger D., Mayberg M., Morgenstern L., Ogilvy C.S., Vespa P., et al. Quality of Care and Outcomes in Research Interdisciplinary Working Group . 2007. Guidelines for the management of spontaneous intracerebral hemorrhage in adults: 2007 update: a guideline from the American Heart Association/American Stroke Association Stroke Council, High Blood Pressure Research Council, and the Quality of Care and Outcomes in Research Interdisciplinary Working Group. Stroke. 38:2001–2023. 10.1161/STROKEAHA.107.183689
    1. Carroll K.C., Viollet B., and Suttles J.. 2013. AMPKα1 deficiency amplifies proinflammatory myeloid APC activity and CD40 signaling. J. Leukoc. Biol. 94:1113–1121. 10.1189/jlb.0313157
    1. Chang C.F., Wan J., Li Q., Renfroe S.C., Heller N.M., and Wang J.. 2017. Alternative activation-skewed microglia/macrophages promote hematoma resolution in experimental intracerebral hemorrhage. Neurobiol. Dis. 103:54–69. 10.1016/j.nbd.2017.03.016
    1. Chao de la Barca J.M., Bakhta O., Kalakech H., Simard G., Tamareille S., Catros V., Callebert J., Gadras C., Tessier L., Reynier P., et al. . 2016. Metabolic Signature of Remote Ischemic Preconditioning Involving a Cocktail of Amino Acids and Biogenic Amines. J. Am. Heart Assoc. 5:e003891 10.1161/JAHA.116.003891
    1. Chiu D.T., van den Berg J., Kuypers F.A., Hung I.J., Wei J.S., and Liu T.Z.. 1996. Correlation of membrane lipid peroxidation with oxidation of hemoglobin variants: possibly related to the rates of hemin release. Free Radic. Biol. Med. 21:89–95. 10.1016/0891-5849(96)00035-4
    1. Christoforidis G.A., Slivka A., Mohammad Y., Karakasis C., Avutu B., and Yang M.. 2007. Size matters: hemorrhage volume as an objective measure to define significant intracranial hemorrhage associated with thrombolysis. Stroke. 38:1799–1804. 10.1161/STROKEAHA.106.472282
    1. Crisostomo P.R., Wairiuko G.M., Wang M., Tsai B.M., Morrell E.D., and Meldrum D.R.. 2006. Preconditioning versus postconditioning: mechanisms and therapeutic potentials. J. Am. Coll. Surg. 202:797–812. 10.1016/j.jamcollsurg.2005.12.002
    1. Darrow V.C., Alvord E.C. Jr., Mack L.A., and Hodson W.A.. 1988. Histologic evolution of the reactions to hemorrhage in the premature human infant’s brain. A combined ultrasound and autopsy study and a comparison with the reaction in adults. Am. J. Pathol. 130:44–58.
    1. de Lima Portella R., Lynn Bickta J., and Shiva S.. 2015. Nitrite Confers Preconditioning and Cytoprotection After Ischemia/Reperfusion Injury Through the Modulation of Mitochondrial Function. Antioxid. Redox Signal. 23:307–327. 10.1089/ars.2015.6260
    1. Dennis M.S. 2003. Outcome after brain haemorrhage. Cerebrovasc. Dis. 16(Suppl 1):9–13. 10.1159/000069935
    1. Dennis M.S., Burn J.P., Sandercock P.A., Bamford J.M., Wade D.T., and Warlow C.P.. 1993. Long-term survival after first-ever stroke: the Oxfordshire Community Stroke Project. Stroke. 24:796–800. 10.1161/01.STR.24.6.796
    1. England T.J., Hedstrom A., O’Sullivan S., Donnelly R., Barrett D.A., Sarmad S., Sprigg N., and Bath P.M.. 2017. RECAST (Remote Ischemic Conditioning After Stroke Trial): A Pilot Randomized Placebo Controlled Phase II Trial in Acute Ischemic Stroke. Stroke. 48:1412–1415. 10.1161/STROKEAHA.116.016429
    1. Fang H., Chen J., Lin S., Wang P., Wang Y., Xiong X., and Yang Q.. 2014. CD36-mediated hematoma absorption following intracerebral hemorrhage: negative regulation by TLR4 signaling. J. Immunol. 192:5984–5992. 10.4049/jimmunol.1400054
    1. Flores J.J., Klebe D., Rolland W.B., Lekic T., Krafft P.R., and Zhang J.H.. 2016. PPARγ-induced upregulation of CD36 enhances hematoma resolution and attenuates long-term neurological deficits after germinal matrix hemorrhage in neonatal rats. Neurobiol. Dis. 87:124–133. 10.1016/j.nbd.2015.12.015
    1. Foerch C., Arai K., Jin G., Park K.P., Pallast S., van Leyen K., and Lo E.H.. 2008. Experimental model of warfarin-associated intracerebral hemorrhage. Stroke. 39:3397–3404. 10.1161/STROKEAHA.108.517482
    1. Gebel J.M. Jr., Jauch E.C., Brott T.G., Khoury J., Sauerbeck L., Salisbury S., Spilker J., Tomsick T.A., Duldner J., and Broderick J.P.. 2002. Relative edema volume is a predictor of outcome in patients with hyperacute spontaneous intracerebral hemorrhage. Stroke. 33:2636–2641. 10.1161/01.STR.0000035283.34109.EA
    1. Geng X., Ren C., Wang T., Fu P., Luo Y., Liu X., Yan F., Ling F., Jia J., Du H., et al. . 2012. Effect of remote ischemic postconditioning on an intracerebral hemorrhage stroke model in rats. Neurol. Res. 34:143–148. 10.1179/1743132811Y.0000000073
    1. Gonzalez N.R., Hamilton R., Bilgin-Freiert A., Dusick J., Vespa P., Hu X., and Asgari S.. 2013. Cerebral hemodynamic and metabolic effects of remote ischemic preconditioning in patients with subarachnoid hemorrhage. Acta Neurochir. Suppl. (Wien). 115:193–198.
    1. Gonzalez N.R., Connolly M., Dusick J.R., Bhakta H., and Vespa P.. 2014. Phase I clinical trial for the feasibility and safety of remote ischemic conditioning for aneurysmal subarachnoid hemorrhage. Neurosurgery. 75:590–598, discussion :598. 10.1227/NEU.0000000000000514
    1. Gordon S., and Martinez F.O.. 2010. Alternative activation of macrophages: mechanism and functions. Immunity. 32:593–604. 10.1016/j.immuni.2010.05.007
    1. Gregson B.A., Broderick J.P., Auer L.M., Batjer H., Chen X.C., Juvela S., Morgenstern L.B., Pantazis G.C., Teernstra O.P., Wang W.Z., et al. . 2012. Individual patient data subgroup meta-analysis of surgery for spontaneous supratentorial intracerebral hemorrhage. Stroke. 43:1496–1504. 10.1161/STROKEAHA.111.640284
    1. Hammond M.D., Ai Y., and Sansing L.H.. 2012. Gr1+ Macrophages and Dendritic Cells Dominate the Inflammatory Infiltrate 12 Hours After Experimental Intracerebral Hemorrhage. Transl. Stroke Res. 3(1, S1):s125–s131. 10.1007/s12975-012-0174-9
    1. Han N., Ding S.J., Wu T., and Zhu Y.L.. 2008. Correlation of free radical level and apoptosis after intracerebral hemorrhage in rats. Neurosci. Bull. 24:351–358. 10.1007/s12264-008-0711-4
    1. Hemphill J.C. III, Greenberg S.M., Anderson C.S., Becker K., Bendok B.R., Cushman M., Fung G.L., Goldstein J.N., Macdonald R.L., Mitchell P.H., et al. Council on Clinical Cardiology . 2015. Guidelines for the Management of Spontaneous Intracerebral Hemorrhage: A Guideline for Healthcare Professionals From the American Heart Association/American Stroke Association. Stroke. 46:2032–2060. 10.1161/STR.0000000000000069
    1. Hess D.C., Blauenfeldt R.A., Andersen G., Hougaard K.D., Hoda M.N., Ding Y., and Ji X.. 2015. Remote ischaemic conditioning-a new paradigm of self-protection in the brain. Nat. Rev. Neurol. 11:698–710. 10.1038/nrneurol.2015.223
    1. Hess D.C., Hoda M.N., and Khan M.B.. 2016. Humoral Mediators of Remote Ischemic Conditioning: Important Role of eNOS/NO/Nitrite. Acta Neurochir. Suppl. (Wien). 121:45–48. 10.1007/978-3-319-18497-5_8
    1. Hoda M.N., Siddiqui S., Herberg S., Periyasamy-Thandavan S., Bhatia K., Hafez S.S., Johnson M.H., Hill W.D., Ergul A., Fagan S.C., and Hess D.C.. 2012. Remote ischemic perconditioning is effective alone and in combination with intravenous tissue-type plasminogen activator in murine model of embolic stroke. Stroke. 43:2794–2799. 10.1161/STROKEAHA.112.660373
    1. Hougaard K.D., Hjort N., Zeidler D., Sørensen L., Nørgaard A., Hansen T.M., von Weitzel-Mudersbach P., Simonsen C.Z., Damgaard D., Gottrup H., et al. . 2014. Remote ischemic perconditioning as an adjunct therapy to thrombolysis in patients with acute ischemic stroke: a randomized trial. Stroke. 45:159–167. 10.1161/STROKEAHA.113.001346
    1. Huang F.P., Xi G., Keep R.F., Hua Y., Nemoianu A., and Hoff J.T.. 2002. Brain edema after experimental intracerebral hemorrhage: role of hemoglobin degradation products. J. Neurosurg. 96:287–293. 10.3171/jns.2002.96.2.0287
    1. Huffman L.J., Miles P.R., Shi X., and Bowman L.. 2000. Hemoglobin potentiates the production of reactive oxygen species by alveolar macrophages. Exp. Lung Res. 26:203–217. 10.1080/019021400269871
    1. Joseph M.J., Caliaperumal J., and Schlichter L.C.. 2016. After Intracerebral Hemorrhage, Oligodendrocyte Precursors Proliferate and Differentiate Inside White-Matter Tracts in the Rat Striatum. Transl. Stroke Res. 7:192–208. 10.1007/s12975-015-0445-3
    1. Khan M.B., Hoda M.N., Vaibhav K., Giri S., Wang P., Waller J.L., Ergul A., Dhandapani K.M., Fagan S.C., and Hess D.C.. 2015. Remote ischemic postconditioning: harnessing endogenous protection in a murine model of vascular cognitive impairment. Transl. Stroke Res. 6:69–77. 10.1007/s12975-014-0374-6
    1. Khan M.B., Hafez S., Hoda M.N., Baban B., Wagner J., Awad M.E., Sangabathula H., Haigh S., Elsalanty M., Waller J.L., and Hess D.C.. 2018. Chronic Remote Ischemic Conditioning Is Cerebroprotective and Induces Vascular Remodeling in a VCID Model. Transl. Stroke Res. 9:51–63. 10.1007/s12975-017-0555-1
    1. Kim C.K., Lee S.H., Kim B.J., Ryu W.S., Choi S.H., Oh B.H., and Yoon B.W.. 2011. Elevated leukocyte count in asymptomatic subjects is associated with a higher risk for cerebral white matter lesions. Clin. Neurol. Neurosurg. 113:177–180. 10.1016/j.clineuro.2010.10.013
    1. King M.D., McCracken D.J., Wade F.M., Meiler S.E., Alleyne C.H. Jr., and Dhandapani K.M.. 2011. Attenuation of hematoma size and neurological injury with curcumin following intracerebral hemorrhage in mice. J. Neurosurg. 115:116–123. 10.3171/2011.2.JNS10784
    1. Koch S., Katsnelson M., Dong C., and Perez-Pinzon M.. 2011. Remote ischemic limb preconditioning after subarachnoid hemorrhage: a phase Ib study of safety and feasibility. Stroke. 42:1387–1391. 10.1161/STROKEAHA.110.605840
    1. Kröller-Schön S., Jansen T., Hauptmann F., Schüler A., Heeren T., Hausding M., Oelze M., Viollet B., Keaney J.F. Jr., Wenzel P., et al. . 2012. α1AMP-activated protein kinase mediates vascular protective effects of exercise. Arterioscler. Thromb. Vasc. Biol. 32:1632–1641. 10.1161/ATVBAHA.111.243980
    1. Lai Y.C., Tabima D.M., Dube J.J., Hughan K.S., Vanderpool R.R., Goncharov D.A., St Croix C.M., Garcia-Ocaña A., Goncharova E.A., Tofovic S.P., et al. . 2016. SIRT3-AMP-Activated Protein Kinase Activation by Nitrite and Metformin Improves Hyperglycemia and Normalizes Pulmonary Hypertension Associated With Heart Failure With Preserved Ejection Fraction. Circulation. 133:717–731.
    1. Lee S.H., Kim B.J., Ryu W.S., Kim C.K., Kim N., Park B.J., and Yoon B.W.. 2010. White matter lesions and poor outcome after intracerebral hemorrhage: a nationwide cohort study. Neurology. 74:1502–1510. 10.1212/WNL.0b013e3181dd425a
    1. Letarte P.B., Lieberman K., Nagatani K., Haworth R.A., Odell G.B., and Duff T.A.. 1993. Hemin: levels in experimental subarachnoid hematoma and effects on dissociated vascular smooth-muscle cells. J. Neurosurg. 79:252–255. 10.3171/jns.1993.79.2.0252
    1. Lou M., Al-Hazzani A., Goddeau R.P. Jr., Novak V., and Selim M.. 2010. Relationship between white-matter hyperintensities and hematoma volume and growth in patients with intracerebral hemorrhage. Stroke. 41:34–40. 10.1161/STROKEAHA.109.564955
    1. Lyden P.D., Shuaib A., Lees K.R., Davalos A., Davis S.M., Diener H.C., Grotta J.C., Ashwood T.J., Hardemark H.G., Svensson H.H., et al. CHANT Trial Investigators . 2007. Safety and tolerability of NXY-059 for acute intracerebral hemorrhage: the CHANT Trial. Stroke. 38:2262–2269. 10.1161/STROKEAHA.106.472746
    1. Mangalam A.K., Rattan R., Suhail H., Singh J., Hoda M.N., Deshpande M., Fulzele S., Denic A., Shridhar V., Kumar A., et al. . 2016. AMP-Activated Protein Kinase Suppresses Autoimmune Central Nervous System Disease by Regulating M1-Type Macrophage-Th17 Axis. J. Immunol. 197:747–760. 10.4049/jimmunol.1501549
    1. Mantovani A., Sica A., and Locati M.. 2005. Macrophage polarization comes of age. Immunity. 23:344–346. 10.1016/j.immuni.2005.10.001
    1. Marquardt G., Wolff R., Janzen R.W., and Seifert V.. 2005. Basal ganglia haematomas in non-comatose patients: subacute stereotactic aspiration improves long-term outcome in comparison to purely medical treatment. Neurosurg. Rev. 28:64–69.
    1. Mayor F., Bilgin-Freiert A., Connolly M., Katsnelson M., Dusick J.R., Vespa P., Koch S., and Gonzalez N.R.. 2013. Effects of remote ischemic preconditioning on the coagulation profile of patients with aneurysmal subarachnoid hemorrhage: a case-control study. Neurosurgery. 73:808–815, discussion :815. 10.1227/NEU.0000000000000098
    1. McFadden J.W., and Corl B.A.. 2009. Activation of AMP-activated protein kinase (AMPK) inhibits fatty acid synthesis in bovine mammary epithelial cells. Biochem. Biophys. Res. Commun. 390:388–393. 10.1016/j.bbrc.2009.09.017
    1. Mendelow A.D., Gregson B.A., Fernandes H.M., Murray G.D., Teasdale G.M., Hope D.T., Karimi A., Shaw M.D., and Barer D.H.. STICH investigators . 2005. Early surgery versus initial conservative treatment in patients with spontaneous supratentorial intracerebral haematomas in the International Surgical Trial in Intracerebral Haemorrhage (STICH): a randomised trial. Lancet. 365:387–397. 10.1016/S0140-6736(05)70233-6
    1. Meng R., Asmaro K., Meng L., Liu Y., Ma C., Xi C., Li G., Ren C., Luo Y., Ling F., et al. . 2012. Upper limb ischemic preconditioning prevents recurrent stroke in intracranial arterial stenosis. Neurology. 79:1853–1861. 10.1212/WNL.0b013e318271f76a
    1. Mikita J., Dubourdieu-Cassagno N., Deloire M.S., Vekris A., Biran M., Raffard G., Brochet B., Canron M.H., Franconi J.M., Boiziau C., and Petry K.G.. 2011. Altered M1/M2 activation patterns of monocytes in severe relapsing experimental rat model of multiple sclerosis. Amelioration of clinical status by M2 activated monocyte administration. Mult. Scler. 17:2–15. 10.1177/1352458510379243
    1. Miron V.E., Boyd A., Zhao J.W., Yuen T.J., Ruckh J.M., Shadrach J.L., van Wijngaarden P., Wagers A.J., Williams A., Franklin R.J.M., and Ffrench-Constant C.. 2013. M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat. Neurosci. 16:1211–1218. 10.1038/nn.3469
    1. Murry C.E., Jennings R.B., and Reimer K.A.. 1986. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation. 74:1124–1136. 10.1161/01.CIR.74.5.1124
    1. Nakamura T., Keep R.F., Hua Y., Hoff J.T., and Xi G.. 2005. Oxidative DNA injury after experimental intracerebral hemorrhage. Brain Res. 1039:30–36. 10.1016/j.brainres.2005.01.036
    1. Nakamura T., Keep R.F., Hua Y., Nagao S., Hoff J.T., and Xi G.. 2006. Iron-induced oxidative brain injury after experimental intracerebral hemorrhage. Acta Neurochir. Suppl. (Wien). 96:194–198. 10.1007/3-211-30714-1_42
    1. Nguyen J.P., Decq P., Brugieres P., Yepes C., Melon E., Gaston A., and Keravel Y.. 1992. A technique for stereotactic aspiration of deep intracerebral hematomas under computed tomographic control using a new device. Neurosurgery. 31:330–334, discussion :334–335. 10.1227/00006123-199208000-00019
    1. Nishino Y., Miura T., Miki T., Sakamoto J., Nakamura Y., Ikeda Y., Kobayashi H., and Shimamoto K.. 2004. Ischemic preconditioning activates AMPK in a PKC-dependent manner and induces GLUT4 up-regulation in the late phase of cardioprotection. Cardiovasc. Res. 61:610–619. 10.1016/j.cardiores.2003.10.022
    1. O’Neill L.A., and Hardie D.G.. 2013. Metabolism of inflammation limited by AMPK and pseudo-starvation. Nature. 493:346–355. 10.1038/nature11862
    1. Pearce E.L., and Pearce E.J.. 2013. Metabolic pathways in immune cell activation and quiescence. Immunity. 38:633–643. 10.1016/j.immuni.2013.04.005
    1. Qing W.G., Dong Y.Q., Ping T.Q., Lai L.G., Fang L.D., Min H.W., Xia L., and Heng P.Y.. 2009. Brain edema after intracerebral hemorrhage in rats: the role of iron overload and aquaporin 4. J. Neurosurg. 110:462–468. 10.3171/2008.4.JNS17512
    1. Qureshi A.I., Tuhrim S., Broderick J.P., Batjer H.H., Hondo H., and Hanley D.F.. 2001. Spontaneous intracerebral hemorrhage. N. Engl. J. Med. 344:1450–1460. 10.1056/NEJM200105103441907
    1. Rincon F., and Mayer S.A.. 2004. Novel therapies for intracerebral hemorrhage. Curr. Opin. Crit. Care. 10:94–100. 10.1097/00075198-200404000-00003
    1. Sadrzadeh S.M., Anderson D.K., Panter S.S., Hallaway P.E., and Eaton J.W.. 1987. Hemoglobin potentiates central nervous system damage. J. Clin. Invest. 79:662–664. 10.1172/JCI112865
    1. Sag D., Carling D., Stout R.D., and Suttles J.. 2008. Adenosine 5′-monophosphate-activated protein kinase promotes macrophage polarization to an anti-inflammatory functional phenotype. J. Immunol. 181:8633–8641. 10.4049/jimmunol.181.12.8633
    1. Samovski D., Su X., Xu Y., Abumrad N.A., and Stahl P.D.. 2012. Insulin and AMPK regulate FA translocase/CD36 plasma membrane recruitment in cardiomyocytes via Rab GAP AS160 and Rab8a Rab GTPase. J. Lipid Res. 53:709–717. 10.1194/jlr.M023424
    1. Sehgal V., Delproposto Z., Haddar D., Haacke E.M., Sloan A.E., Zamorano L.J., Barger G., Hu J., Xu Y., Prabhakaran K.P., et al. . 2006. Susceptibility-weighted imaging to visualize blood products and improve tumor contrast in the study of brain masses. J. Magn. Reson. Imaging. 24:41–51. 10.1002/jmri.20598
    1. Sharma M.D., Hou D.Y., Baban B., Koni P.A., He Y., Chandler P.R., Blazar B.R., Mellor A.L., and Munn D.H.. 2010. Reprogrammed foxp3(+) regulatory T cells provide essential help to support cross-presentation and CD8(+) T cell priming in naive mice. Immunity. 33:942–954. 10.1016/j.immuni.2010.11.022
    1. Shen H.Y., Coelho J.E., Ohtsuka N., Canas P.M., Day Y.J., Huang Q.Y., Rebola N., Yu L., Boison D., Cunha R.A., et al. . 2008. A critical role of the adenosine A2A receptor in extrastriatal neurons in modulating psychomotor activity as revealed by opposite phenotypes of striatum and forebrain A2A receptor knock-outs. J. Neurosci. 28:2970–2975. 10.1523/JNEUROSCI.5255-07.2008
    1. Sukumari-Ramesh S., Alleyne C.H. Jr., and Dhandapani K.M.. 2012. Astrocyte-specific expression of survivin after intracerebral hemorrhage in mice: a possible role in reactive gliosis? J. Neurotrauma. 29:2798–2804. 10.1089/neu.2011.2243
    1. Teernstra O.P., Evers S.M., Lodder J., Leffers P., Franke C.L., and Blaauw G.. Multicenter randomized controlled trial (SICHPA) . 2003. Stereotactic treatment of intracerebral hematoma by means of a plasminogen activator: a multicenter randomized controlled trial (SICHPA). Stroke. 34:968–974. 10.1161/01.STR.0000063367.52044.40
    1. Thiex R., Rohde V., Rohde I., Mayfrank L., Zeki Z., Thron A., Gilsbach J.M., and Uhl E.. 2004. Frame-based and frameless stereotactic hematoma puncture and subsequent fibrinolytic therapy for the treatment of spontaneous intracerebral hemorrhage. J. Neurol. 251:1443–1450. 10.1007/s00415-004-0554-5
    1. Vaibhav K., Shrivastava P., Tabassum R., Khan A., Javed H., Ahmed M.E., Islam F., Safhi M.M., and Islam F.. 2013. Delayed administration of zingerone mitigates the behavioral and histological alteration via repression of oxidative stress and intrinsic programmed cell death in focal transient ischemic rats. Pharmacol. Biochem. Behav. 113:53–62. 10.1016/j.pbb.2013.10.008
    1. Wakita H., Tomimoto H., Akiguchi I., and Kimura J.. 1995. Protective effect of cyclosporin A on white matter changes in the rat brain after chronic cerebral hypoperfusion. Stroke. 26:1415–1422. 10.1161/01.STR.26.8.1415
    1. Weimar C., Weber C., Wagner M., Busse O., Haberl R.L., Lauterbach K.W., and Diener H.C.. German Stroke Data Bank Collaborators . 2003. Management patterns and health care use after intracerebral hemorrhage. a cost-of-illness study from a societal perspective in Germany. Cerebrovasc. Dis. 15:29–36. 10.1159/000067119
    1. Xi G., Wagner K.R., Keep R.F., Hua Y., de Courten-Myers G.M., Broderick J.P., Brott T.G., and Hoff J.T.. 1998. Role of blood clot formation on early edema development after experimental intracerebral hemorrhage. Stroke. 29:2580–2586. 10.1161/01.STR.29.12.2580
    1. Yellon D.M., and Hausenloy D.J.. 2005. Realizing the clinical potential of ischemic preconditioning and postconditioning. Nat. Clin. Pract. Cardiovasc. Med. 2:568–575. 10.1038/ncpcardio0346
    1. Yip S., and Sastry B.R.. 2000. Effects of hemoglobin and its breakdown products on synaptic transmission in rat hippocampal CA1 neurons. Brain Res. 864:1–12. 10.1016/S0006-8993(00)02067-9
    1. Zhao X., Sun G., Zhang J., Strong R., Song W., Gonzales N., Grotta J.C., and Aronowski J.. 2007. Hematoma resolution as a target for intracerebral hemorrhage treatment: role for peroxisome proliferator-activated receptor gamma in microglia/macrophages. Ann. Neurol. 61:352–362. 10.1002/ana.21097
    1. Zhao X., Sun G., Ting S.M., Song S., Zhang J., Edwards N.J., and Aronowski J.. 2015. Cleaning up after ICH: the role of Nrf2 in modulating microglia function and hematoma clearance. J. Neurochem. 133:144–152. 10.1111/jnc.12974
    1. Zhu Y.P., Brown J.R., Sag D., Zhang L., and Suttles J.. 2015. Adenosine 5′-monophosphate-activated protein kinase regulates IL-10-mediated anti-inflammatory signaling pathways in macrophages. J. Immunol. 194:584–594. 10.4049/jimmunol.1401024

Source: PubMed

3
订阅