353
Views
2
CrossRef citations to date
0
Altmetric
Review

The potential of light therapy in Parkinson's disease

, , , , , , , , & show all
Pages 1-14 | Published online: 18 Feb 2014

References

  • Bergman H, Deuschl G. Pathophysiology of Parkinson’s disease: from clinical neurology to basic neuroscience and back. Mov Disord. 2002;17(Suppl 3):S28–S40.
  • Jankovic J. Parkinson’s disease: clinical features and diagnosis. J Neurol Neurosurg Psychiatry. 2008;79(4):368–376.
  • Rinne JO. Nigral degeneration in Parkinson’s disease. Mov Disord. 1993;(8 Suppl 1):S31–S35.
  • Parent A. Carpenter’s Human Neuroanatomy. 9th ed. Baltimore, MD: Williams and Wilkins; 1996.
  • Blandini F, Nappi G, Tassorelli C, Martignoni E. Functional changes of the basal ganglia circuitry in Parkinson’s disease. Prog Neurobiol. 2000;62(1):63–88.
  • Schober A. Classic toxin-induced animal models of Parkinson’s disease: 6-OHDA and MPTP. Cell Tissue Res. 2004;318(1):215–224.
  • Blesa J, Phani S, Jackson-Lewis V, Przedborski S. Classic and new animal models of Parkinson’s disease. J Biomed Biotechnol. 2012;2012:845618.
  • Bové J, Perier C. Neurotoxin-based models of Parkinson’s disease. Neuroscience. 2012;211:51–76.
  • Bezard E, Dovero S, Imbert C, Boraud T, Gross CE. Spontaneous long-term compensatory dopaminergic sprouting in MPTP-treated mice. Synapse. 2000;38(3):363–368.
  • Farrer MJ. Genetics of Parkinson disease: paradigm shifts and future prospects. Nat Rev Genet. 2006;7(4):306–318.
  • Corti O, Brice A. Mitochondrial quality control turns out to be the principal suspect in parkin and PINK1-related autosomal recessive Parkinson’s disease. Curr Opin Neurobiol. 2013;23(1):100–108.
  • Giasson BI, Duda JE, Quinn SM, Zhang B, Trojanowski JQ, Lee VM. Neuronal alpha-synucleinopathy with severe movement disorder in mice expressing A53T human alpha-synuclein. Neuron. 2002;34(4):521–533.
  • Ittner LM, Fath T, Ke YD, et al. Parkinsonism and impaired axonal transport in a mouse model of frontotemporal dementia. Proc Natl Acad Sci U S A. 2008;105(41):15997–16002.
  • Muqit MM, Abou-Sleiman PM, Saurin AT, et al. Altered cleavage and localization of PINK1 to aggresomes in the presence of proteasomal stress. J Neurochem. 2006;98(1):156–169.
  • Exner N, Lutz AK, Haass C, Winklhofer KF. Mitochondrial dysfunction in Parkinson’s disease: molecular mechanisms and pathophysiological consequences. EMBO J. 2012;31(14):3038–3062.
  • Goedert M, Spillantini MG, Del Tredici K, Braak H. 100 years of Lewy pathology. Nat Rev Neurol. 2013;9(1):13–24.
  • Albin RL, Greenamyre JT. Alternative excitotoxic hypotheses. Neurology. 1992;42(4):733–738.
  • Beal MF, Brouillet E, Jenkins BG, et al. Neurochemical and histologic characterization of striatal excitotoxic lesions produced by the mitochondrial toxin 3-nitropropionic acid. J Neurosci. 1993;13(10):4181–4192.
  • Piallat B, Benazzouz A, Benabid AL. Subthalamic nucleus lesion in rats prevents dopaminergic nigral neuron degeneration after striatal 6-OHDA injection: behavioural and immunohistochemical studies. Eur J Neurosci. 1996;8(7):1408–1414.
  • Wallace BA, Ashkan K, Heise CE, et al. Survival of midbrain dopaminergic cells after lesion or deep brain stimulation of the subthalamic nucleus in MPTP-treated monkeys. Brain. 2007;130(Pt 8):2129–2145.
  • Whitton PS. Neuroinflammation and the prospects for anti-inflammatory treatment of Parkinson’s disease. Curr Opin Investig Drugs. 2010;11(7):788–794.
  • McGeer PL, McGeer EG. Glial reactions in Parkinson’s disease. Mov Disord. 2008;23(4):474–483.
  • Jankovic P, Poewe W. Therapies in Parkinson’s disease. Curr Opin Neurol. 2012;25(4):433–447.
  • Schapira AH. Present and future drug treatment for Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2005;76(11):1472–1478.
  • Worth PF. How to treat Parkinson’s disease in 2013. Clin Med. 2013 13(1):93–96.
  • Hart RG, Pearce LA, Ravina BM, Yaltho TC, Marler JR. Neuroprotection trials in Parkinson’s disease: systematic review. Mov Disord. 2009; 24(5):647–654.
  • Ashkan K, Wallace B, Bell BA, Benabid AL. Deep brain stimulation of the subthalamic nucleus in Parkinson’s disease 1993–2003: where are we 10 years on? Br J Neurosurg. 2004;18(1):19–34.
  • Benabid AL, Chabardes S, Mitrofanis J, Pollak P. Deep brain stimulation of the subthalamic nucleus for the treatment of Parkinson’s disease. Lancet Neurol. 2009;8(1):67–81.
  • Charles PD, Gill CE, Davis TL, Konrad PE, Benabid AL. Is deep brain stimulation neuroprotective if applied early in the course of PD? Nat Clin Pract Neurol. 2008;4(8):424–426.
  • Stocchi F, Olanow CW. Obstacles to the development of a neuroprotective therapy for Parkinson’s disease. Mov Disord. 2013;28(1):3–7.
  • Kordower JH, Bjorklund A. Trophic factor gene therapy for Parkinson’s disease. Mov Disord. 2013;28(1):96–109.
  • Maruyama W, Naoi M. “70th Birthday Professor Riederer” induction of glial cell line-derived and brain-derived neurotrophic factors by rasagiline and (-)deprenyl: a way to a disease-modifying therapy? J Neural Transm. 2013;120(1):83–89.
  • Arias-Carrión O, Yamada E, Freundlieb N, et al. Neurogenesis in substantia nigra of parkinsonian brains? J Neural Transm Suppl. 2009;(73):279–285.
  • Grazina R, Massano J. Physical exercise and Parkinson’s disease: influence on symptoms, disease course and prevention. Rev Neurosci. 2013;24(2):139–152.
  • Kones R. Parkinson’s disease: mitochondrial molecular pathology, inflammation, statins, and therapeutic neuroprotective nutrition. Nutr Clin Pract. 2010;25(4):371–389.
  • Romeo S, Viaggi C, Di Camillo D, et al. Bright light exposure reduces TH-positive dopamine neurons: implications of light pollution in Parkinson’s disease epidemiology. Sci Rep. 2013;3:1395.
  • Eells JT, Henry MM, Summerfelt P, et al. Therapeutic photobiomodulation for methanol-induced retinal toxicity. Proc Natl Acad Sci U S A. 2003;100(6):3439–3444.
  • Natoli R, Zhu Y, Valter K, Bisti S, Eells J, Stone J. Gene and noncoding RNA regulation underlying photoreceptor protection: microarray study of dietary antioxidant saffron and photobiomodulation in rat retina. Mol Vis. 2010;16:1801–1822.
  • Natoli R, Valter K, Barbosa M, et al. 670 nm photobiomodulation as a novel protection against retinopathy of prematurity: evidence from oxygen induced retinopathy models. PLoS One. 2013;8(8):e72135.
  • Albarracin R, Valter K. 670 nm red light preconditioning supports Müller cell function: evidence from the white light-induced damage model in the rat retina. Photochem Photobiol. 2012;88(6):1418–1427.
  • Begum R, Powner MB, Hudson N, Hogg C, Jeffery G. Treatment with 670 nm light up regulates cytochrome C oxidase expression and reduces inflammation in an age-related macular degeneration model. PloS One. 2013;8(2):e57828.
  • Ando T, Xuan W, Xu T, et al. Comparison of therapeutic effects between pulsed and continuous wave 810-nm wavelength laser irradiation for traumatic brain injury in mice. PLoS One. 2011;6(10):e26212.
  • Oron A, Oron U, Streeter J, et al. Near infrared transcranial laser therapy applied at various modes to mice following traumatic brain injury significantly reduces long-term neurological deficits. J Neurotrauma. 2012;29(2):401–407.
  • Quirk BJ, Torbey M, Buchmann E, Verma S, Whelan HT. Near-infrared photobiomodulation in an animal model of traumatic brain injury: improvements at the behavioral and biochemical levels. Photomed Laser Surg. 2012;30(9):523–529.
  • Lapchak PA, Wei J, Zivin JA. Transcranial infrared laser therapy improves clinical rating scores after embolic strokes in rabbits. Stroke. 2004;35(8):1985–1988.
  • De Taboada L, Ilic S, Leichliter-Martha S, Oron U, Oron A, Streeter J. Transcranial application of low-energy laser irradiation improves neurological deficits in rats following acute stroke. Lasers Surg Med. 2006;38(1):70–73.
  • Oron A, Oron U, Chen J, et al. Low-level laser therapy applied transcranially to rats after induction of stroke significantly reduces long-term neurological deficits. Stroke. 2006;37(10):2620–2624.
  • Moges H, Vasconcelos OM, Campbell WW, et al. Light therapy and supplementary Riboflavin in the SOD1 transgenic mouse model of familial amyotrophic lateral sclerosis (FALS). Lasers Surg Med. 2009; 41(1):52–59.
  • Muili KA, Gopalakrishnan S, Meyer SL, Eells JT, Lyons JA. Amelioration of experimental autoimmune encephalomyelitis in C57BL/6 mice by photobiomodulation induced by 670 nm light. PLoS One. 2012;7(1):e30655.
  • Liang HL, Whelan HT, Eells JT, Wong-Riley MT. Near-infrared light via light-emitting diode treatment is therapeutic against rotenone- and 1-methyl-4-phenylpyridinium ion-induced neurotoxicity. Neuroscience. 2008;153(4):963–974.
  • Ying R, Liang HL, Whelan HT, Eells JT, Wong-Riley MT. Pretreatment with near-infrared light via light-emitting diode provides added benefit against rotenone- and MPP+-induced neurotoxicity. Brain Res. 2008;1243:167–173.
  • Whelan H, Desmet K, Buchmann E, et al. Harnessing the cell’s own ability to repair and prevent neurodegenerative disease. SPIE Newsroom. 2008;2008:1–3.
  • Shaw VE, Spana S, Ashkan K, et al. Neuroprotection of midbrain dopaminergic cells in MPTP-treated mice after near-infrared light treatment. J Comp Neurol. 2010;518(1):25–40.
  • Peoples CL, Spana S, Ashkan K, et al. Photobiomodulation enhances nigral dopaminergic cell survival in a chronic MPTP mouse model of Parkinson’s disease. Parkinsonism Relat Disord. 2012;18(5):469–476.
  • Michalikova S, Ennaceur A, van Rensburg R, Chazot PL. Emotional responses and memory performance of middle-aged CD1 mice in a 3D maze: effects of low infrared light. Neurobiol Learn Mem. 2008;89(4):480–488.
  • De Taboada L, Yu J, El-Amouri S, et al. Transcranial laser therapy attenuates amyloid-β peptide neuropathology in amyloid-β protein precursor transgenic mice. J Alzheimers Dis. 2011;23(3):521–535.
  • Grillo SL, Duggett NA, Ennaceur A, Chazot PL. Non-invasive infra-red therapy (1072 nm) reduces β-amyloid protein levels in the brain of an Alzheimer’s disease mouse model, TASTPM. J Photochem Photobiol B. 2013;123:13–22.
  • Barrett DW, Gonzalez-Lima F. Transcranial infrared laser stimulation produces beneficial cognitive and emotional effects in humans. Neuroscience. 2013;230:13–23.
  • Lampl Y, Zivin JA, Fisher M, et al. Infrared laser therapy for ischemic stroke: a new treatment strategy: results of the NeuroThera Effectiveness and Safety Trial-1 (NEST-1). Stroke. 2007;38(6):1843–1849.
  • Lapchak PA. Taking a light approach to treating acute ischemic stroke patients: transcranial near-infrared laser therapy translational science. Ann Med. 2010;42(8):576–586.
  • Naeser MA, Saltmarche A, Krengel MH, Hamblin MR, Knight JA. Improved cognitive function after transcranial, light-emitting diode treatments in chronic, traumatic brain injury: two case reports. Photomed Laser Surg. 2011;29(5):351–358.
  • Schiffer F, Johnston AL, Ravichandran C, et al. Psychological benefits 2 and 4 weeks after a single treatment with near infrared light to the forehead: a pilot study of 10 patients with major depression and anxiety. Behav Brain Funct. 2009;5:46.
  • Merry G, Devenyi R, Dotson R, Markowitz S, Reyes S. Treatment of dry age-related macular degeneration with photobiomodulation. Presented in: Proceedings of the 9th World Association of Laser Therapy Congress; September 28–30, 2012; Gold Coast (Australia). Paper P928C0072.
  • Galluzzi L, Kepp O, Trojel-Hansen C, Kroemer G. Mitochondrial control of cellular life, stress, and death. Circ Res. 2012;111(9):1198–1207.
  • Desmet KD, Paz DA, Corry JJ, et al. Clinical and experimental applications of NIR-LED photobiomodulation. Photomed Laser Surg. 2006;24(2):121–128.
  • Hamblin MR, Demidova TN. Mechanisms of low level light therapy. Proc SPIE. 2006;6140:614001.
  • Rojas JC, Gonzalez-Lima F. Low-level light therapy of the eye and brain. Eye and Brain. 2011;3:49–67.
  • Chung H, Dai T, Sharma SK, Huang YY, Carroll JD, Hamblin MR. The nuts and bolts of low-level laser (light) therapy. Ann Biomed Eng. 2012;40(2):516–533.
  • Quirk BJ, Whelan HT. Near-infrared irradiation photobiomodulation: the need for basic science. Photomed Laser Surg. 2011;29(3):143–144.
  • Braverman B, McCarthy RJ, Ivankovich AD, Forde DE, Overfield M, Bapna MS. Effect of helium-neon and infrared laser irradiation on wound healing in rabbits. Lasers Surg Med. 1989;9(1):50–58.
  • Stone J, Johnstone DM, Mitrofanis J. The helmet experiment in Parkinson’s disease: an observation of the mechanism of neuroprotection by near infra-red light. Presented in: Proceedings of the 9th World Association of Laser Therapy Congress; September 28–30, 2012; Gold Coast (Australia). Paper P928C0072.
  • Johnstone DM, Moro C, el Massri N, Torres N, Jaeger XD, Reinhart F, Purushothuman S, Benabid AL, Stone J, Mitrofanis J. Indirect application of near infrared light induces neuroprotection in a mouse model of Parkinson’s disease – an abscopal neuroprotective effect. In preparation. 2014. In press.
  • Byrnes KR, Waynant RW, Ilev IK, et al. Light promotes regeneration and functional recovery and alters the immune response after spinal cord injury. Lasers Surg Med. 2005;36(3):171–185.
  • Tuby H, Maltz L, Oron U. Modulations of VEGF and iNOS in the rat heart by low level laser therapy are associated with cardioprotection and enhanced angiogenesis. Lasers Surg Med. 2006;38(7):682–688.
  • Tuby H, Maltz L, Oron U. Induction of autologous mesenchymal stem cells in the bone marrow by low-level laser therapy has profound beneficial effects on the infarcted rat heart. Lasers Surg Med. 2011;43(5):401–409.
  • Hou ST, Jiang SX, Smith RA. Permissive and repulsive cues and signalling pathways of axonal outgrowth and regeneration. Int Rev Cell Mol Biol. 2008;267:125–181.
  • Postow MA, Callahan MK, Barker CA, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med. 2012;366(10):925–931.
  • Moro C, el Massri N, Torres N, et al. Photobiomodulation inside the brain: a novel method of applying near-infrared light intracranially and its impact on dopaminergic cell survival in MPTP-treated mice. J Neurosurgery. Epub October 25, 2014.
  • Moro C, Torres N, el Massri N, et al. Photobiomodulation preserves behaviour and midbrain dopaminergic cells from MPTP toxicity: evidence from two mouse strains. BMC Neurosci. 2013;14:40.
  • Purushothuman S, Nandasena C, Johnstone DM, Stone J, Mitrofanis J. The impact of near-infrared light on dopaminergic cell survival in a transgenic mouse model of parkinsonism. Brain Res. 2013;1535:61–70.
  • Trimmer PA, Schwartz KM, Borland MK, De Taboada L, Streeter J, Oron U. Reduced axonal transport in Parkinson’s disease cybrid neurites is restored by light therapy. Mol Neurodegener. 2009;4:26.
  • Ashkan K, Wallace BA, Mitrofanis J, et al. SPECT imaging, immunohistochemical and behavioural correlations in the primate models of Parkinson’s disease. Parkinsonism Relat Disord. 2007;13(5):266–275.
  • Peoples C, Shaw VE, Stone J, Jeffery G, Baker GE, Mitrofanis J. Survival of dopaminergic amacrine cells after near-infrared light treatment in MPTP-treated mice. ISRN Neurol. 2012;2012:850150.
  • Lu J, Jhou TC, Saper CB. Identification of wake-active dopaminergic neurons in the ventral periaqueductal gray matter. J Neurosci. 2006;26(1):193–202.
  • Shaw VE, Peoples C, Spana S, et al. Patterns of cell activity in the subthalamic region associated with the neuroprotective action of near-infrared light treatment in MPTP-treated mice. Parkinson’s Dis. 2012;2012:296875.
  • DeSmet K, Buchmann E, Henry M, et al. Near-infrared light as a possible treatment option for Parkinson’s disease and laser eye injury. Proc SPIE. 2009;7165:7 16503.
  • Quirk BJ, Desmet KD, Henry M, et al. Therapeutic effect of near infrared (NIR) light on Parkinson’s disease models. Front Biosci (Elite Ed). 2012;4:818–823.
  • Lanzafame RJ, Stadler I, Kurtz AF, et al. Reciprocity of exposure time and irradiance on energy density during photoradiation on wound healing in a murine pressure ulcer model. Lasers Surg Med. 2007;39(6):534–542.
  • Quietmind Foundation. Quietmind Foundation clinical Trial. Available from: http://www.quietmindfdn.org/. Marvin Berman. 2013. http://www.youtube.com/watch?v=9X-hjgay7pg. Accessed January16, 2012.
  • Maloney R, Shanks S, Maloney J. The application of low-level laser therapy for the symptomatic care of late stage Parkinson’s disease: a non-controlled, non-randomized study. April 14–15, 2010. Am Soc Laser Med Surg Abs. 2010;185.
  • Li Q, Song L, Guo K, Yu Y, Ma S, Shen L. [The effect of endonasal low energy He-Ne laser treatment of Parkinson’s disease on CCK-8 content in blood]. Chin J Neurol. 1999;32:364. Chinese.
  • Xu C, Lu C, Wang L, Li Q. [The effects of endonasal low energy He-Ne laser therapy on antioxydation of Parkinson’s disease]. Prac J Med Pharm. 2003;11:816–817. Chinese.
  • Zhao G, Guo K, Dan J. [36 case analysis of Parkinson’s disease treated by endonasal low energy He-Ne laser]. Acta Academiae medicinae Qingdao Universitatis. 2003;39:398. Chinese.
  • Burchman MA. Using Photobiomodulation on a severe Parkinson’s patient to enable extractions, root canal treatment and partial denture fabrication. J Laser Dent. 2011;19:297–300.
  • Wong-Riley MT, Liang HL, Eells JT, et al. Photobiomodulation directly benefits primary neurons functionally inactivated by toxins: role of cytochrome c oxidase. J Biol Chem. 2005;280(6):4761–4771.
  • Eells JT, DeSmet K, Kirk DK, et al. Photobiomodulation for the treatment of retinal injury and retinal degenerative diseases. Proceed Light-Activated Tissue Regeneration Therapy Conference. 2008;12:39–51.
  • Zivin JA, Albers GW, Bornstein N, et al; NeuroThera Effectiveness and Safety Trial-2 Investigators. Effectiveness and safety of transcranial laser therapy for acute ischemic stroke. Stroke. 2009;40(4):1359–1364.
  • Jagdeo JR, Adams LE, Brody NI, Siegel DM. Transcranial red and near infrared light transmission in a cadaveric model. PLoS One. 2012;7(10):e47460.
  • Abdo A, Sahin M. . NIR Light Penetration Depth in the Rat Peripheral Nerve and Brain Cortex. In: Conference Proceedings of the IEEE. Eng Med Biol Soc. 2007:1723–1725. doi:10.1109/IEMBS.2007. 4352642.
  • Torres N, Chabardes S, Piallat B, Devergnas A, Benabid AL. Body fat and body weight reduction following hypothalamic deep brain stimulation in monkeys: an intraventricular approach. Int J Obes (Lond). 2012;36(12):1537–1544.
  • Samuel M, Ceballos-Baumann AO, Blin J, et al. Evidence for lateral premotor and parietal overactivity in Parkinson’s disease during sequential and bimanual movements. A PET study. Brain. 1997; 120(Pt 6):963–976.
  • Sabatini U, Boulanouar K, Fabre N, et al. Cortical motor reorganization in akinetic patients with Parkinson’s disease: a functional MRI study. Brain. 2000;123(Pt 2):394–403.
  • Haslinger B, Erhard P, Kämpfe N, et al. Event-related functional magnetic resonance imaging in Parkinson’s disease before and after levodopa. Brain. 2001;124(Pt 3):558–570.
  • LeWitt PA. Neuroprotection for Parkinson’s disease. J Neural Transm Suppl. 2006;71:113–122.
  • Ma J, Shaw VE, Mitrofanis J. Does melatonin help save dopaminergic cells in MPTP-treated mice? Parkinsonism Relat Disord. 2009;15(4):307–314.
  • Ilic S, Leichliter S, Streeter J, Oron A, DeTaboada L, Oron U. Effects of power densities, continuous and pulse frequencies, and number of sessions of low-level laser therapy on intact rat brain. Photomed Laser Surg. 2006;24(4):458–466.
  • McCarthy TJ, De Taboada L, Hildebrandt PK, Ziemer EL, Richieri SP, Streeter J. Long-term safety of single and multiple infrared transcranial laser treatments in Sprague-Dawley rats. Photomed Laser Surg. 2010;28(5):663–667.
  • Tata DB, Waynant RW. Laser therapy: a review of its mechanism of action and potential medical applications. Laser Photonics Rev. 2011;5(1):1–12.