Skip to main content
Log in

Photobiomodulation for Global Cerebral Ischemia: Targeting Mitochondrial Dynamics and Functions

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Hypothermia is currently the only approved therapy for global cerebral ischemia (GCI) after cardiac arrest; however, it unfortunately has multiple adverse effects. As a noninvasive procedure, photobiomodulation (PBM) therapy has emerged as a potential novel treatment for brain injury. PBM involves the use of low-level laser light therapy to influence cell behavior. In this study, we evaluated the therapeutic effects of PBM treatment with an 808-nm diode laser initiated 6 h after GCI. It was noted that PBM dose-dependently protected against GCI-induced neuronal death in the vulnerable hippocampal CA1 subregion. Functional assessments demonstrated that PBM markedly preserved both short-term (a week) and long-term (6 months) spatial learning and memory function following GCI. Further mechanistic studies revealed that PBM post-treatment (a) preserved healthy mitochondrial dynamics and suppressed substantial mitochondrial fragmentation of CA1 neurons, by reducing the detrimental Drp1 GTPase activity and its interactions with adaptor proteins Mff and Fis1 and by balancing mitochondrial targeting fission and fusion protein levels; (b) reduced mitochondrial oxidative damage and excessive mitophagy and restored mitochondrial overall health status and preserved mitochondrial function; and (c) suppressed mitochondria-dependent apoptosome formation/caspase-3/9 apoptosis-processing activities. Additionally, we validated, in an in vitro ischemia model, that cytochrome c oxidase served as a key PBM target for mitochondrial function preservation and neuroprotection. Our findings suggest that PBM serves as a promising therapeutic strategy for the functional recovery after GCI, with mechanisms involving PBM’s preservation on mitochondrial dynamics and functions and the inhibition of delayed apoptotic neuronal death in GCI.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  1. Roger VL, Go AS, Lloyd-Jones DM, Adams RJ, Berry JD, Brown TM, Carnethon MR, Dai S et al (2011) Heart disease and stroke statistics—2011 update: a report from the American Heart Association. Circulation 123(4):e18–e209. https://doi.org/10.1161/CIR.0b013e3182009701

    Article  PubMed  Google Scholar 

  2. Moulaert VR, Verbunt JA, van Heugten CM, Wade DT (2009) Cognitive impairments in survivors of out-of-hospital cardiac arrest: a systematic review. Resuscitation 80(3):297–305. https://doi.org/10.1016/j.resuscitation.2008.10.034

    Article  PubMed  Google Scholar 

  3. Sauve MJ, Doolittle N, Walker JA, Paul SM, Scheinman MM (1996) Factors associated with cognitive recovery after cardiopulmonary resuscitation. Am J Crit Care 5(2):127–139

    CAS  PubMed  Google Scholar 

  4. Roine RO, Kajaste S, Kaste M (1993) Neuropsychological sequelae of cardiac arrest. JAMA 269(2):237–242

    Article  CAS  PubMed  Google Scholar 

  5. Wolman RL, Nussmeier NA, Aggarwal A, Kanchuger MS, Roach GW, Newman MF, Mangano CM, Marschall KE et al (1999) Cerebral injury after cardiac surgery: identification of a group at extraordinary risk. Multicenter Study of Perioperative Ischemia Research Group (McSPI) and the Ischemia Research Education Foundation (IREF) Investigators. Stroke 30(3):514–522

    Article  CAS  PubMed  Google Scholar 

  6. Brillman J (1993) Central nervous system complications in coronary artery bypass graft surgery. Neurol Clin 11(2):475–495

    Article  CAS  PubMed  Google Scholar 

  7. Swain JA, Anderson RV, Siegman MG (1993) Low-flow cardiopulmonary bypass and cerebral protection: a summary of investigations. Ann Thorac Surg 56(6):1490–1492

    Article  CAS  PubMed  Google Scholar 

  8. Pulsinelli WA, Brierley JB, Plum F (1982) Temporal profile of neuronal damage in a model of transient forebrain ischemia. Ann Neurol 11(5):491–498. https://doi.org/10.1002/ana.410110509

    Article  CAS  PubMed  Google Scholar 

  9. Kirino T (1982) Delayed neuronal death in the gerbil hippocampus following ischemia. Brain Res 239(1):57–69

    Article  CAS  PubMed  Google Scholar 

  10. Kirino T, Sano K (1984) Selective vulnerability in the gerbil hippocampus following transient ischemia. Acta Neuropathol 62(3):201–208

    Article  CAS  PubMed  Google Scholar 

  11. Chen J, Zhu RL, Nakayama M, Kawaguchi K, Jin K, Stetler RA, Simon RP, Graham SH (1996) Expression of the apoptosis-effector gene, Bax, is up-regulated in vulnerable hippocampal CA1 neurons following global ischemia. J Neurochem 67(1):64–71

    Article  CAS  PubMed  Google Scholar 

  12. Harukuni I, Bhardwaj A (2006) Mechanisms of brain injury after global cerebral ischemia. Neurol Clin 24(1):1–21. https://doi.org/10.1016/j.ncl.2005.10.004

    Article  PubMed  Google Scholar 

  13. Kim YM, Yim HW, Jeong SH, Klem ML, Callaway CW (2012) Does therapeutic hypothermia benefit adult cardiac arrest patients presenting with non-shockable initial rhythms?: a systematic review and meta-analysis of randomized and non-randomized studies. Resuscitation 83(2):188–196. https://doi.org/10.1016/j.resuscitation.2011.07.031

    Article  PubMed  Google Scholar 

  14. Nielsen N, Friberg H, Gluud C, Herlitz J, Wetterslev J (2011) Hypothermia after cardiac arrest should be further evaluated—a systematic review of randomised trials with meta-analysis and trial sequential analysis. Int J Cardiol 151(3):333–341. https://doi.org/10.1016/j.ijcard.2010.06.008

    Article  PubMed  Google Scholar 

  15. Tucker D, Lu Y, Zhang Q (2017) From mitochondrial function to neuroprotection—an emerging role for methylene blue. Mol Neurobiol 55:5137–5153. https://doi.org/10.1007/s12035-017-0712-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kumar R, Bukowski MJ, Wider JM, Reynolds CA, Calo L, Lepore B, Tousignant R, Jones M et al (2016) Mitochondrial dynamics following global cerebral ischemia. Mol Cell Neurosci 76:68–75. https://doi.org/10.1016/j.mcn.2016.08.010

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Chan PH (2004) Mitochondria and neuronal death/survival signaling pathways in cerebral ischemia. Neurochem Res 29(11):1943–1949

    Article  CAS  PubMed  Google Scholar 

  18. Herst PM, Rowe MR, Carson GM, Berridge MV (2017) Functional mitochondria in health and disease. Front Endocrinol 8:296. https://doi.org/10.3389/fendo.2017.00296

    Article  Google Scholar 

  19. Bakthavachalam P, Shanmugam PST (2017) Mitochondrial dysfunction—silent killer in cerebral ischemia. J Neurol Sci 375:417–423. https://doi.org/10.1016/j.jns.2017.02.043

    Article  CAS  PubMed  Google Scholar 

  20. Niizuma K, Yoshioka H, Chen H, Kim GS, Jung JE, Katsu M, Okami N, Chan PH (2010) Mitochondrial and apoptotic neuronal death signaling pathways in cerebral ischemia. Biochim Biophys Acta 1802(1):92–99. https://doi.org/10.1016/j.bbadis.2009.09.002

    Article  CAS  PubMed  Google Scholar 

  21. Frezza C, Cipolat S, Martins de Brito O, Micaroni M, Beznoussenko GV, Rudka T, Bartoli D, Polishuck RS et al (2006) OPA1 controls apoptotic cristae remodeling independently from mitochondrial fusion. Cell 126(1):177–189. https://doi.org/10.1016/j.cell.2006.06.025

    Article  CAS  PubMed  Google Scholar 

  22. Olichon A, Baricault L, Gas N, Guillou E, Valette A, Belenguer P, Lenaers G (2003) Loss of OPA1 perturbates the mitochondrial inner membrane structure and integrity, leading to cytochrome c release and apoptosis. J Biol Chem 278(10):7743–7746. https://doi.org/10.1074/jbc.C200677200

    Article  CAS  PubMed  Google Scholar 

  23. Cipolat S, Martins de Brito O, Dal Zilio B, Scorrano L (2004) OPA1 requires mitofusin 1 to promote mitochondrial fusion. Proc Natl Acad Sci U S A 101(45):15927–15932. https://doi.org/10.1073/pnas.0407043101

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Arnoult D, Grodet A, Lee YJ, Estaquier J, Blackstone C (2005) Release of OPA1 during apoptosis participates in the rapid and complete release of cytochrome c and subsequent mitochondrial fragmentation. J Biol Chem 280(42):35742–35750. https://doi.org/10.1074/jbc.M505970200

    Article  CAS  PubMed  Google Scholar 

  25. Sharp WW (2015) Dynamin-related protein 1 as a therapeutic target in cardiac arrest. J Mol Med 93(3):243–252. https://doi.org/10.1007/s00109-015-1257-3

    Article  CAS  PubMed  Google Scholar 

  26. Smirnova E, Griparic L, Shurland DL, van der Bliek AM (2001) Dynamin-related protein Drp1 is required for mitochondrial division in mammalian cells. Mol Biol Cell 12(8):2245–2256

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  27. Bleazard W, McCaffery JM, King EJ, Bale S, Mozdy A, Tieu Q, Nunnari J, Shaw JM (1999) The dynamin-related GTPase Dnm1 regulates mitochondrial fission in yeast. Nat Cell Biol 1(5):298–304. https://doi.org/10.1038/13014

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Park JH, Ko J, Hwang J, Koh HC (2015) Dynamin-related protein 1 mediates mitochondria-dependent apoptosis in chlorpyrifos-treated SH-SY5Y cells. Neurotoxicology 51:145–157. https://doi.org/10.1016/j.neuro.2015.10.008

    Article  CAS  PubMed  Google Scholar 

  29. Zhang QG, Wang RM, Scott E, Han D, Dong Y, Tu JY, Yang F, Reddy Sareddy G et al (2013) Hypersensitivity of the hippocampal CA3 region to stress-induced neurodegeneration and amyloidogenesis in a rat model of surgical menopause. Brain 136(Pt 5):1432–1445. https://doi.org/10.1093/brain/awt046

    Article  PubMed  PubMed Central  Google Scholar 

  30. Zhang QG, Han D, Wang RM, Dong Y, Yang F, Vadlamudi RK, Brann DW (2011) C terminus of Hsc70-interacting protein (CHIP)-mediated degradation of hippocampal estrogen receptor-alpha and the critical period hypothesis of estrogen neuroprotection. Proc Natl Acad Sci U S A 108(35):E617–E624. https://doi.org/10.1073/pnas.1104391108

    Article  PubMed  PubMed Central  Google Scholar 

  31. Lu Y, Wang R, Dong Y, Tucker D, Zhao N, Ahmed ME, Zhu L, Liu TC et al (2017) Low-level laser therapy for beta amyloid toxicity in rat hippocampus. Neurobiol Aging 49:165–182. https://doi.org/10.1016/j.neurobiolaging.2016.10.003

    Article  CAS  PubMed  Google Scholar 

  32. Tu J, Zhang X, Zhu Y, Dai Y, Li N, Yang F, Zhang Q, Brann DW et al (2015) Cell-permeable peptide targeting the Nrf2-Keap1 interaction: a potential novel therapy for global cerebral ischemia. J Neurosci 35(44):14727–14739. https://doi.org/10.1523/JNEUROSCI.1304-15.2015

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhang QG, Raz L, Wang R, Han D, De Sevilla L, Yang F, Vadlamudi RK, Brann DW (2009) Estrogen attenuates ischemic oxidative damage via an estrogen receptor alpha-mediated inhibition of NADPH oxidase activation. J Neurosci 29(44):13823–13836. https://doi.org/10.1523/JNEUROSCI.3574-09.2009

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lu Y, Dong Y, Tucker D, Wang R, Ahmed ME, Brann D, Zhang Q (2017) Treadmill exercise exerts neuroprotection and regulates microglial polarization and oxidative stress in a streptozotocin-induced rat model of sporadic Alzheimer’s disease. J Alzheimers Dis 56(4):1469–1484. https://doi.org/10.3233/JAD-160869

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lu Q, Tucker D, Dong Y, Zhao N, Zhang Q (2016) Neuroprotective and functional improvement effects of methylene blue in global cerebral ischemia. Mol Neurobiol 53(8):5344–5355. https://doi.org/10.1007/s12035-015-9455-0

    Article  CAS  PubMed  Google Scholar 

  36. Bondarenko AL, Serova LD, Shabalin VN (1991) The role of the major histocompatibility complex antigens in the development of allergic diseases in the Korean population. Sovetskaia meditsina (4):26–28

  37. Sareddy GR, Zhang Q, Wang R, Scott E, Zou Y, O’Connor JC, Chen Y, Dong Y et al (2015) Proline-, glutamic acid-, and leucine-rich protein 1 mediates estrogen rapid signaling and neuroprotection in the brain. Proc Natl Acad Sci U S A 112(48):E6673–E6682. https://doi.org/10.1073/pnas.1516729112

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhu Y, Zhang Q, Zhang W, Li N, Dai Y, Tu J, Yang F, Brann DW et al (2017) Protective effect of 17beta-estradiol upon hippocampal spine density and cognitive function in an animal model of vascular dementia. Sci Rep 7:42660. https://doi.org/10.1038/srep42660

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Xu Z, Guo X, Yang Y, Tucker D, Lu Y, Xin N, Zhang G, Yang L et al (2017) Low-level laser irradiation improves depression-like behaviors in mice. Mol Neurobiol 54(6):4551–4559. https://doi.org/10.1007/s12035-016-9983-2

    Article  CAS  PubMed  Google Scholar 

  40. Liu B, Li L, Zhang Q, Chang N, Wang D, Shan Y, Li L, Wang H et al (2010) Preservation of GABAA receptor function by PTEN inhibition protects against neuronal death in ischemic stroke. Stroke 41(5):1018–1026. https://doi.org/10.1161/STROKEAHA.110.579011

    Article  CAS  PubMed  Google Scholar 

  41. Li L, Prabhakaran K, Mills EM, Borowitz JL, Isom GE (2005) Enhancement of cyanide-induced mitochondrial dysfunction and cortical cell necrosis by uncoupling protein-2. Toxicol Sci 86(1):116–124. https://doi.org/10.1093/toxsci/kfi164

    Article  CAS  PubMed  Google Scholar 

  42. Zhang QG, Wang R, Tang H, Dong Y, Chan A, Sareddy GR, Vadlamudi RK, Brann DW (2014) Brain-derived estrogen exerts anti-inflammatory and neuroprotective actions in the rat hippocampus. Mol Cell Endocrinol 389(1–2):84–91. https://doi.org/10.1016/j.mce.2013.12.019

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Cereghetti GM, Stangherlin A, Martins de Brito O, Chang CR, Blackstone C, Bernardi P, Scorrano L (2008) Dephosphorylation by calcineurin regulates translocation of Drp1 to mitochondria. Proc Natl Acad Sci U S A 105(41):15803–15808. https://doi.org/10.1073/pnas.0808249105

    Article  PubMed  PubMed Central  Google Scholar 

  44. Cribbs JT, Strack S (2007) Reversible phosphorylation of Drp1 by cyclic AMP-dependent protein kinase and calcineurin regulates mitochondrial fission and cell death. EMBO Rep 8(10):939–944. https://doi.org/10.1038/sj.embor.7401062

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kiryk A, Pluta R, Figiel I, Mikosz M, Ulamek M, Niewiadomska G, Jablonski M, Kaczmarek L (2011) Transient brain ischemia due to cardiac arrest causes irreversible long-lasting cognitive injury. Behav Brain Res 219(1):1–7. https://doi.org/10.1016/j.bbr.2010.12.004

    Article  PubMed  Google Scholar 

  46. Ulamek-Koziol M, Pluta R, Bogucka-Kocka A, Januszewski S, Kocki J, Czuczwar SJ (2016) Brain ischemia with Alzheimer phenotype dysregulates Alzheimer’s disease-related proteins. Pharmacol Rep 68(3):582–591. https://doi.org/10.1016/j.pharep.2016.01.006

    Article  CAS  PubMed  Google Scholar 

  47. Lipton P (1999) Ischemic cell death in brain neurons. Physiol Rev 79(4):1431–1568

    Article  CAS  PubMed  Google Scholar 

  48. Abe K, Aoki M, Kawagoe J, Yoshida T, Hattori A, Kogure K, Itoyama Y (1995) Ischemic delayed neuronal death. A mitochondrial hypothesis. Stroke 26(8):1478–1489

    Article  CAS  PubMed  Google Scholar 

  49. Mishra P, Chan DC (2016) Metabolic regulation of mitochondrial dynamics. J Cell Biol 212(4):379–387. https://doi.org/10.1083/jcb.201511036

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Stein LR, Imai S (2012) The dynamic regulation of NAD metabolism in mitochondria. Trends Endocrinol Metab 23(9):420–428. https://doi.org/10.1016/j.tem.2012.06.005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. de la Torre JC (2017) Treating cognitive impairment with transcranial low level laser therapy. J Photochem Photobiol B 168:149–155. https://doi.org/10.1016/j.jphotobiol.2017.02.008

    Article  CAS  PubMed  Google Scholar 

  52. Agrawal T, Gupta GK, Rai V, Carroll JD, Hamblin MR (2014) Pre-conditioning with low-level laser (light) therapy: light before the storm. Dose Response 12(4):619–649. https://doi.org/10.2203/dose-response.14-032.Agrawal

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Gonzalez-Lima F, Barksdale BR, Rojas JC (2014) Mitochondrial respiration as a target for neuroprotection and cognitive enhancement. Biochem Pharmacol 88(4):584–593. https://doi.org/10.1016/j.bcp.2013.11.010

    Article  CAS  PubMed  Google Scholar 

  54. Hashmi JT, Huang YY, Osmani BZ, Sharma SK, Naeser MA, Hamblin MR (2010) Role of low-level laser therapy in neurorehabilitation. PM R 2(12 Suppl 2):S292–S305. https://doi.org/10.1016/j.pmrj.2010.10.013

    Article  PubMed  PubMed Central  Google Scholar 

  55. Lee JC, Won MH (2014) Neuroprotection of antioxidant enzymes against transient global cerebral ischemia in gerbils. Anat Cell Biol 47(3):149–156. https://doi.org/10.5115/acb.2014.47.3.149

    Article  PubMed  PubMed Central  Google Scholar 

  56. Friberg H, Wieloch T, Castilho RF (2002) Mitochondrial oxidative stress after global brain ischemia in rats. Neurosci Lett 334(2):111–114

    Article  CAS  PubMed  Google Scholar 

  57. Yu Z, Liu N, Zhao J, Li Y, McCarthy TJ, Tedford CE, Lo EH, Wang X (2015) Near infrared radiation rescues mitochondrial dysfunction in cortical neurons after oxygen-glucose deprivation. Metab Brain Dis 30(2):491–496. https://doi.org/10.1007/s11011-014-9515-6

    Article  CAS  PubMed  Google Scholar 

  58. Huang YY, Nagata K, Tedford CE, Hamblin MR (2014) Low-level laser therapy (810 nm) protects primary cortical neurons against excitotoxicity in vitro. J Biophotonics 7(8):656–664. https://doi.org/10.1002/jbio.201300125

    Article  CAS  PubMed  Google Scholar 

  59. Shi RY, Zhu SH, Li V, Gibson SB, Xu XS, Kong JM (2014) BNIP3 interacting with LC3 triggers excessive mitophagy in delayed neuronal death in stroke. CNS Neurosci Ther 20(12):1045–1055. https://doi.org/10.1111/cns.12325

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Meyer JN, Leuthner TC, Luz AL (2017) Mitochondrial fusion, fission, and mitochondrial toxicity. Toxicology 391:42–53. https://doi.org/10.1016/j.tox.2017.07.019

    Article  CAS  PubMed  Google Scholar 

  61. Wang X, Su B, Zheng L, Perry G, Smith MA, Zhu X (2009) The role of abnormal mitochondrial dynamics in the pathogenesis of Alzheimer’s disease. J Neurochem 109(Suppl 1):153–159. https://doi.org/10.1111/j.1471-4159.2009.05867.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Cartron PF, Bellot G, Oliver L, Grandier-Vazeille X, Manon S, Vallette FM (2008) Bax inserts into the mitochondrial outer membrane by different mechanisms. FEBS Lett 582(20):3045–3051. https://doi.org/10.1016/j.febslet.2008.07.047

    Article  CAS  PubMed  Google Scholar 

  63. Arnoult D, Parone P, Martinou JC, Antonsson B, Estaquier J, Ameisen JC (2002) Mitochondrial release of apoptosis-inducing factor occurs downstream of cytochrome c release in response to several proapoptotic stimuli. J Cell Biol 159(6):923–929. https://doi.org/10.1083/jcb.200207071

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Wang J, Wang P, Li S, Wang S, Li Y, Liang N, Wang M (2014) Mdivi-1 prevents apoptosis induced by ischemia-reperfusion injury in primary hippocampal cells via inhibition of reactive oxygen species-activated mitochondrial pathway. J Stroke Cerebrovasc Dis 23(6):1491–1499. https://doi.org/10.1016/j.jstrokecerebrovasdis.2013.12.021

    Article  PubMed  Google Scholar 

  65. Ma X, Xie Y, Chen Y, Han B, Li J, Qi S (2016) Post-ischemia mdivi-1 treatment protects against ischemia/reperfusion-induced brain injury in a rat model. Neurosci Lett 632:23–32. https://doi.org/10.1016/j.neulet.2016.08.026

    Article  CAS  PubMed  Google Scholar 

  66. Chung H, Dai T, Sharma SK, Huang YY, Carroll JD, Hamblin MR (2012) The nuts and bolts of low-level laser (light) therapy. Ann Biomed Eng 40(2):516–533. https://doi.org/10.1007/s10439-011-0454-7

    Article  PubMed  Google Scholar 

  67. Lapchak PA, Han MK, Salgado KF, Streeter J, Zivin JA (2008) Safety profile of transcranial near-infrared laser therapy administered in combination with thrombolytic therapy to embolized rabbits. Stroke 39(11):3073–3078. https://doi.org/10.1161/STROKEAHA.108.516393

    Article  CAS  PubMed  Google Scholar 

  68. Lapchak PA, Salgado KF, Chao CH, Zivin JA (2007) Transcranial near-infrared light therapy improves motor function following embolic strokes in rabbits: an extended therapeutic window study using continuous and pulse frequency delivery modes. Neuroscience 148(4):907–914. https://doi.org/10.1016/j.neuroscience.2007.07.002

    Article  CAS  PubMed  Google Scholar 

  69. Lapchak PA, Wei J, Zivin JA (2004) Transcranial infrared laser therapy improves clinical rating scores after embolic strokes in rabbits. Stroke 35(8):1985–1988. https://doi.org/10.1161/01.STR.0000131808.69640.b7

    Article  PubMed  Google Scholar 

  70. Yoshioka H, Niizuma K, Katsu M, Okami N, Sakata H, Kim GS, Narasimhan P, Chan PH (2011) NADPH oxidase mediates striatal neuronal injury after transient global cerebral ischemia. J Cereb Blood Flow Metab 31(3):868–880. https://doi.org/10.1038/jcbfm.2010.166

    Article  CAS  PubMed  Google Scholar 

  71. Taraszewska A, Zelman IB, Ogonowska W, Chrzanowska H (2002) The pattern of irreversible brain changes after cardiac arrest in humans. Folia Neuropathol 40(3):133–141

    PubMed  Google Scholar 

  72. Hacke W, Schellinger PD, Albers GW, Bornstein NM, Dahlof BL, Fulton R, Kasner SE, Shuaib A et al (2014) Transcranial laser therapy in acute stroke treatment: results of neurothera effectiveness and safety trial 3, a phase III clinical end point device trial. Stroke 45(11):3187–3193. https://doi.org/10.1161/STROKEAHA.114.005795

    Article  PubMed  Google Scholar 

  73. Levine SR, Hill MD (2014) NeuroThera Effectiveness and Safety Trial 3: how do we align corporate and scientific integrity to complete and report pharma-sponsored trials properly? Stroke 45(11):3175–3177. https://doi.org/10.1161/STROKEAHA.114.006750

    Article  PubMed  Google Scholar 

Download references

Funding

This study was supported by Research Grants NS086929 (to QZ) and NS088058 (to DW) from the National Institute of Neurological Disorders and Stroke, National Institutes of Health USA and by National Natural Science Foundation Grants of China: 30970664 and 31171354 (to RMW).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Ruimin Wang, Darrell W. Brann or Quanguang Zhang.

Ethics declarations

All animal surgery protocols were approved by the Institutional Animal Care and Use Committee (IACUC) of the local university and were carried out in compliance with National Institutes of Health guidelines.

Competing Interests

The authors declare that they have no competing interests.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, R., Dong, Y., Lu, Y. et al. Photobiomodulation for Global Cerebral Ischemia: Targeting Mitochondrial Dynamics and Functions. Mol Neurobiol 56, 1852–1869 (2019). https://doi.org/10.1007/s12035-018-1191-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-018-1191-9

Keywords

Navigation