193
Views
11
CrossRef citations to date
0
Altmetric
Original Research

Rosuvastatin Nanomicelles Target Neuroinflammation and Improve Neurological Deficit in a Mouse Model of Intracerebral Hemorrhage

, , , , , , , & show all
Pages 2933-2947 | Published online: 20 Apr 2021

References

  • van Asch CJ, Luitse MJ, Rinkel GJ, van der Tweel I, Algra A, Klijn CJ. Incidence, case fatality, and functional outcome of intracerebral haemorrhage over time, according to age, sex, and ethnic origin: a systematic review and meta-analysis. Lancet Neurol. 2010;9(2):167–176. doi:10.1016/S1474-4422(09)70340-0
  • Cordonnier C, Demchuk A, Ziai W, Anderson CS. Intracerebral haemorrhage: current approaches to acute management. Lancet. 2018;392(10154):1257–1268. doi:10.1016/S0140-6736(18)31878-6
  • Zhu H, Wang Z, Yu J, et al. Role and mechanisms of cytokines in the secondary brain injury after intracerebral hemorrhage. Prog Neurobiol. 2019;178:101610.
  • Zhou Y, Wang Y, Wang J, Anne Stetler R, Yang QW. Inflammation in intracerebral hemorrhage: from mechanisms to clinical translation. Prog Neurobiol. 2014;115:25–44.
  • Sabatine MS, Giugliano RP, Keech AC, et al. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med. 2017;376(18):1713–1722. doi:10.1056/NEJMoa1615664
  • Li GM, Zhao J, Li B, et al. The anti-inflammatory effects of statins on patients with rheumatoid arthritis: a systemic review and meta-analysis of 15 randomized controlled trials. Autoimmun Rev. 2018;17(3):215–225. doi:10.1016/j.autrev.2017.10.013
  • Satoh M, Takahashi Y, Tabuchi T, et al. Cellular and molecular mechanisms of statins: an update on pleiotropic effects. Clin Sci (Lond). 2015;129(2):93–105. doi:10.1042/CS20150027
  • Zhang Q, Dong J, Yu Z. Pleiotropic use of statins as non-lipid-lowering drugs. Int J Biol Sci. 2020;16(14):2704–2711. doi:10.7150/ijbs.42965
  • White CM. A review of the pharmacologic and pharmacokinetic aspects of rosuvastatin. J Clin Pharmacol. 2002;42(9):963–970. doi:10.1177/009127002401102876
  • Kobayashi M, Chisaki I, Narumi K, et al. Association between risk of myopathy and cholesterol-lowering effect: a comparison of all statins. Life Sci. 2008;82(17–18):969–975. doi:10.1016/j.lfs.2008.02.019
  • Chrysant SG. New onset diabetes mellitus induced by statins: current evidence. Postgrad Med. 2017;129(4):430–435. doi:10.1080/00325481.2017.1292107
  • Pandit AK, Kumar P, Kumar A, Chakravarty K, Misra S, Prasad K. High-dose statin therapy and risk of intracerebral hemorrhage: a meta-analysis. Acta Neurol Scand. 2016;134(1):22–28. doi:10.1111/ane.12540
  • Lin HP, Tu HP, Hsieh YP, Lee BS. Controlled release of lovastatin from poly(lactic-co-glycolic acid) nanoparticles for direct pulp capping in rat teeth. Int J Nanomedicine. 2017;12:5473–5485. doi:10.2147/IJN.S138410
  • Tai IC, Wang YH, Chen CH, Chuang SC, Chang JK, Ho ML. Simvastatin enhances Rho/actin/cell rigidity pathway contributing to mesenchymal stem cells’ osteogenic differentiation. Int J Nanomedicine. 2015;10:5881–5894. doi:10.2147/IJN.S84273
  • Chen J, Jiang Z, Xu W, et al. Spatiotemporally targeted nanomedicine overcomes hypoxia-induced drug resistance of tumor cells after disrupting neovasculature. Nano Lett. 2020;20(8):6191–6198. doi:10.1021/acs.nanolett.0c02515
  • Li D, Zhang R, Liu G, Kang Y, Wu J. Redox-responsive self-assembled nanoparticles for cancer therapy. Adv Healthc Mater. 2020;9(20):e2000605. doi:10.1002/adhm.202000605
  • Wang L, You X, Dai C, Tong T, Wu J. Hemostatic nanotechnologies for external and internal hemorrhage management. Biomater Sci. 2020;8(16):4396–4412. doi:10.1039/D0BM00781A
  • Ding J, Chen J, Gao L, et al. Engineered nanomedicines with enhanced tumor penetration. Nano Today. 2019;29.
  • Feng X, Xu W, Li Z, Song W, Ding J, Chen X. Immunomodulatory Nanosystems. Adv Sci (Weinh). 2019;6(17):1900101. doi:10.1002/advs.201900101
  • Han S, Huang K, Gu Z, Wu J. Tumor immune microenvironment modulation-based drug delivery strategies for cancer immunotherapy. Nanoscale. 2020;12(2):413–436. doi:10.1039/C9NR08086D
  • Wu C, Xu Q, Chen X, Liu J. Delivery luteolin with folacin-modified nanoparticle for glioma therapy. Int J Nanomedicine. 2019;14:7515–7531. doi:10.2147/IJN.S214585
  • Wang Y, Xie J, Ai Z, Su J. Nobiletin-loaded micelles reduce ovariectomy-induced bone loss by suppressing osteoclastogenesis. Int J Nanomedicine. 2019;14:7839–7849. doi:10.2147/IJN.S213724
  • Yang L, Zhang Z, Hou J, et al. Targeted delivery of ginsenoside compound K using TPGS/PEG-PCL mixed micelles for effective treatment of lung cancer. Int J Nanomedicine. 2017;12:7653–7667. doi:10.2147/IJN.S144305
  • Lin H, Wang Q, Zhong R, et al. Biomimetic phosphorylcholine strategy to improve the hemocompatibility of pH-responsive micelles containing tertiary amino groups. Colloids Surf B Biointerfaces. 2019;184:110545. doi:10.1016/j.colsurfb.2019.110545
  • Zhou W, Zi L, Cen Y, You C, Tian M. Copper sulfide nanoparticles-incorporated hyaluronic acid injectable hydrogel with enhanced angiogenesis to promote wound healing. Front Bioeng Biotechnol. 2020;8:417. doi:10.3389/fbioe.2020.00417
  • Karki K, Knight RA, Han Y, et al. Simvastatin and atorvastatin improve neurological outcome after experimental intracerebral hemorrhage. Stroke. 2009;40(10):3384–3389. doi:10.1161/STROKEAHA.108.544395
  • Xu J, Duan Z, Qi X, et al. Injectable gelatin hydrogel suppresses inflammation and enhances functional recovery in a mouse model of intracerebral hemorrhage. Front Bioeng Biotechnol. 2020;8:785. doi:10.3389/fbioe.2020.00785
  • Fang H, Chen J, Lin S, et al. CD36-mediated hematoma absorption following intracerebral hemorrhage: negative regulation by TLR4 signaling. J Immunol. 2014;192(12):5984–5992. doi:10.4049/jimmunol.1400054
  • Krafft PR, Altay O, Rolland WB, et al. α7 nicotinic acetylcholine receptor agonism confers neuroprotection through GSK-3β inhibition in a mouse model of intracerebral hemorrhage. Stroke. 2012;43(3):844–850. doi:10.1161/STROKEAHA.111.639989
  • Duan Z, Li H, Qi X, et al. Crocin attenuation of neurological deficits in a mouse model of intracerebral hemorrhage. Brain Res Bull. 2019;150:186–195. doi:10.1016/j.brainresbull.2019.05.023
  • Kapur NK, Musunuru K. Clinical efficacy and safety of statins in managing cardiovascular risk. Vasc Health Risk Manag. 2008;4(2):341–353. doi:10.2147/VHRM.S1653
  • Chen Q, Shi X, Tan Q, et al. Simvastatin promotes hematoma absorption and reduces hydrocephalus following intraventricular hemorrhage in part by upregulating CD36. Transl Stroke Res. 2017;8(4):362–373. doi:10.1007/s12975-017-0521-y
  • Wang Y, Chen Q, Tan Q, et al. Simvastatin accelerates hematoma resolution after intracerebral hemorrhage in a PPARγ-dependent manner. Neuropharmacology. 2018;128:244–254. doi:10.1016/j.neuropharm.2017.10.021
  • Bouët V, Freret T, Toutain J, Divoux D, Boulouard M, Schumann-Bard P. Sensorimotor and cognitive deficits after transient middle cerebral artery occlusion in the mouse. Exp Neurol. 2007;203(2):555–567. doi:10.1016/j.expneurol.2006.09.006
  • Wang YC, Wang PF, Fang H, Chen J, Xiong XY, Yang QW. Toll-like receptor 4 antagonist attenuates intracerebral hemorrhage-induced brain injury. Stroke. 2013;44(9):2545–2552. doi:10.1161/STROKEAHA.113.001038
  • Witsch J, Al-Mufti F, Connolly ES, et al. Statins and perihemorrhagic edema in patients with spontaneous intracerebral hemorrhage. Neurology. 2019;92(18):e2145–e2149. doi:10.1212/WNL.0000000000006931
  • Selim M, Norton C. Perihematomal edema: implications for intracerebral hemorrhage research and therapeutic advances. J Neurosci Res. 2020;98(1):212–218. doi:10.1002/jnr.24372
  • Matsushita K, Meng W, Wang X, et al. Evidence for apoptosis after intercerebral hemorrhage in rat striatum. J Cereb Blood Flow Metab. 2000;20(2):396–404. doi:10.1097/00004647-200002000-00022
  • Ehara A, Ueda S. Application of Fluoro-Jade C in acute and chronic neurodegeneration models: utilities and staining differences. Acta Histochem Cytochem. 2009;42(6):171–179. doi:10.1267/ahc.09018
  • Lan X, Han X, Li Q, Yang QW, Wang J. Modulators of microglial activation and polarization after intracerebral haemorrhage. Nat Rev Neurol. 2017;13(7):420–433.
  • Mracsko E, Veltkamp R. Neuroinflammation after intracerebral hemorrhage. Front Cell Neurosci. 2014;8:388. doi:10.3389/fncel.2014.00388
  • Jung KH, Chu K, Jeong SW, et al. HMG-CoA reductase inhibitor, atorvastatin, promotes sensorimotor recovery, suppressing acute inflammatory reaction after experimental intracerebral hemorrhage. Stroke. 2004;35(7):1744–1749. doi:10.1161/01.STR.0000131270.45822.85
  • Ewen T, Qiuting L, Chaogang T, et al. Neuroprotective effect of atorvastatin involves suppression of TNF-α and upregulation of IL-10 in a rat model of intracerebral hemorrhage. Cell Biochem Biophys. 2013;66(2):337–346. doi:10.1007/s12013-012-9453-z
  • Lan X, Han X, Li Q, et al. Pinocembrin protects hemorrhagic brain primarily by inhibiting toll-like receptor 4 and reducing M1 phenotype microglia. Brain Behav Immun. 2017;61:326–339. doi:10.1016/j.bbi.2016.12.012
  • Youssef S, Stüve O, Patarroyo JC, et al. The HMG-CoA reductase inhibitor, atorvastatin, promotes a Th2 bias and reverses paralysis in central nervous system autoimmune disease. Nature. 2002;420(6911):78–84. doi:10.1038/nature01158
  • Cheng SM, Lai JH, Yang SP, et al. Modulation of human T cells signaling transduction by lovastatin. Int J Cardiol. 2010;140(1):24–33. doi:10.1016/j.ijcard.2008.10.044
  • Bagheri H, Ghasemi F, Barreto GE, Sathyapalan T, Jamialahmadi T, Sahebkar A. The effects of statins on microglial cells to protect against neurodegenerative disorders: a mechanistic review. Biofactors. 2020;46(3):309–325. doi:10.1002/biof.1597
  • Baldanta S, Fernández-Escobar M, Acín-Perez R, et al. ISG15 governs mitochondrial function in macrophages following vaccinia virus infection. PLoS Pathog. 2017;13(10):e1006651. doi:10.1371/journal.ppat.1006651
  • Zhang X, Jin J, Peng X, Ramgolam VS, Markovic-Plese S. Simvastatin inhibits IL-17 secretion by targeting multiple IL-17-regulatory cytokines and by inhibiting the expression of IL-17 transcription factor RORC in CD4+ lymphocytes. J Immunol. 2008;180(10):6988–6996. doi:10.4049/jimmunol.180.10.6988
  • Kagami S, Owada T, Kanari H, et al. Protein geranylgeranylation regulates the balance between Th17 cells and Foxp3+ regulatory T cells. Int Immunol. 2009;21(6):679–689. doi:10.1093/intimm/dxp037
  • Dehnavi S, Sohrabi N, Sadeghi M, et al. Statins and autoimmunity: state-of-the-art. Pharmacol Ther. 2020;214:107614. doi:10.1016/j.pharmthera.2020.107614