Shining light on the head: Photobiomodulation for brain disorders

Michael R Hamblin, Michael R Hamblin

Abstract

Photobiomodulation (PBM) describes the use of red or near-infrared light to stimulate, heal, regenerate, and protect tissue that has either been injured, is degenerating, or else is at risk of dying. One of the organ systems of the human body that is most necessary to life, and whose optimum functioning is most worried about by humankind in general, is the brain. The brain suffers from many different disorders that can be classified into three broad groupings: traumatic events (stroke, traumatic brain injury, and global ischemia), degenerative diseases (dementia, Alzheimer's and Parkinson's), and psychiatric disorders (depression, anxiety, post traumatic stress disorder). There is some evidence that all these seemingly diverse conditions can be beneficially affected by applying light to the head. There is even the possibility that PBM could be used for cognitive enhancement in normal healthy people. In this transcranial PBM (tPBM) application, near-infrared (NIR) light is often applied to the forehead because of the better penetration (no hair, longer wavelength). Some workers have used lasers, but recently the introduction of inexpensive light emitting diode (LED) arrays has allowed the development of light emitting helmets or "brain caps". This review will cover the mechanisms of action of photobiomodulation to the brain, and summarize some of the key pre-clinical studies and clinical trials that have been undertaken for diverse brain disorders.

Keywords: Alzheimer's disease; Cognitive enhancement; Ischemic stroke; Low level laser (light) therapy; Major depression; Parkinson's disease; Photobiomodulation; Traumatic brain injury.

Figures

Graphical abstract
Graphical abstract
Fig. 1
Fig. 1
Molecular and intracellular mechanisms of transcranial low level laser (light) or photobiomodulation. AP1, activator protein 1; ATP, adenosine triphosphate; Ca2 +, calcium ions; cAMP, cyclic adenosine monophosphate; NF-kB, nuclear factor kappa B; NO, nitric oxide; ROS, reactive oxygen species; TRPV, transient receptor potential vanilloid.
Fig. 2
Fig. 2
Tissue specific processes that occur after PBM and benefit a range of brain disorders. BDNF, brain-derived neurotrophic factor; LLLT, low level light therapy; NGF, nerve growth factor; NT-3, neurotrophin 3; PBM, photobiomodulation; SOD, superoxide dismutase.
Fig. 3
Fig. 3
tPBM for TBI in a mouse model. Mice received a closed head injury and 4 hours later a single exposure of the head to one of four different lasers (36 J/cm2 delivered at 150 mW/cm2 over 4 min with spot size 1-cm diameter) . A, 665 nm; B, 730 nm; C, 810 nm; D, 980 nm.
Fig. 4
Fig. 4
tPBM for controlled cortical impact TBI in a mouse model. (A) Mice received a single exposure (810 nm laser, 36 J/cm2 delivered at 50 mW/cm2 over 12 min) . (B) Mice received 3 daily exposures starting 4 h post-TBI and were sacrificed after 7 or 28 days. BDNF and neurogenesis (BrdU) were increased at 7 days , while synaptogenesis was increased at 28 days .
Fig. 5
Fig. 5
tPBM for Alzheimer's disease. (A) Nineteen patients were randomized to receive real or sham tPBM (810 nm LED, 24.6 J/cm2 at 41 mW/cm2). (B) Significant decline in ADAS-cog (improved cognitive performance) in real but not sham (unpublished data).
Fig. 6
Fig. 6
tPBM for major depression and anxiety in humans. (A) Ten patients received a single exposure to the forehead (810 LED, 60 J/cm2 delivered at 250 mW/cm2). (B) Mean Hamilton score for depression at baseline and at two weeks post-treatment .

References

    1. McGuff P.E., Deterling R.A., Jr., Gottlieb L.S. Tumoricidal effect of laser energy on experimental and human malignant tumors. N. Engl. J. Med. 1965;273:490–492.
    1. Maiman T.H. Stimulated optical radiation in ruby. Nature. 1960;187:493–494.
    1. Mester E., Ludány G., Sellyei M., Szende B., Total G.J. The simulating effect of low power laser rays on biological systems. Laser Rev. 1968;1:3.
    1. Mester E., Szende B., Gartner P. The effect of laser beams on the growth of hair in mice. Radiobiol. Radiother. (Berl) 1968;9:621–626.
    1. Mester E., Mester A.F., Mester A. The biomedical effects of laser application. Lasers Surg. Med. 1985;5:31–39.
    1. Mester E., Nagylucskay S., Doklen A., Tisza S. Laser stimulation of wound healing. Acta Chir. Acad. Sci. Hung. 1976;17:49–55.
    1. Mester E., Spiry T., Szende B. Effect of laser rays on wound healing. Bull. Soc. Int. Chir. 1973;32:169–173.
    1. Chung H., Dai T., Sharma S.K., Huang Y.Y., Carroll J.D., Hamblin M.R. The nuts and bolts of low-level laser (light) therapy. Ann. Biomed. Eng. 2012;40:516–533.
    1. Anders J.J., Lanzafame R.J., Arany P.R. Low-level light/laser therapy versus photobiomodulation therapy. Photomed. Laser Surg. 2015;33:183–184.
    1. De Freitas L.F., Hamblin M.R. Proposed mechanisms of photobiomodulation or low-level light therapy. IEEE J. Sel. Top. Quantum Electron. 2016;22:7000417.
    1. Lane N. Cell biology: power games. Nature. 2006;443:901–903.
    1. Waypa G.B., Smith K.A., Schumacker P.T. O2 sensing, mitochondria and ROS signaling: the fog is lifting. Mol. Asp. Med. 2016;47-48:76–89.
    1. Zhao Y., Vanhoutte P.M., Leung S.W. Vascular nitric oxide: beyond eNOS. J. Pharmacol. Sci. 2015;129:83–94.
    1. Ferrante M., Petrini M., Trentini P., Perfetti G., Spoto G. Effect of low-level laser therapy after extraction of impacted lower third molars. Lasers Med. Sci. 2013;28:845–849.
    1. Skopin M.D., Molitor S.C. Effects of near-infrared laser exposure in a cellular model of wound healing. Photodermatol. Photoimmunol. Photomed. 2009;25:75–80.
    1. Barrett D.W., Gonzalez-Lima F. Transcranial infrared laser stimulation produces beneficial cognitive and emotional effects in humans. Neuroscience. 2013;230:13–23.
    1. Dougal G., Lee S.Y. Evaluation of the efficacy of low-level light therapy using 1072 nm infrared light for the treatment of herpes simplex labialis. Clin. Exp. Dermatol. 2013;38:713–718.
    1. Vatansever F., Hamblin M.R. Far infrared radiation (FIR): its biological effects and medical applications. Photonics Lasers Med. 2012;4:255–266.
    1. Palazzo E., Rossi F., de Novellis V., Maione S. Endogenous modulators of TRP channels. Curr. Top. Med. Chem. 2013;13:398–407.
    1. Planells-Cases R., Valente P., Ferrer-Montiel A., Qin F., Szallasi A. Complex regulation of TRPV1 and related thermo-TRPs: implications for therapeutic intervention. Adv. Exp. Med. Biol. 2011;704:491–515.
    1. Caterina M.J. Transient receptor potential ion channels as participants in thermosensation and thermoregulation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007;292:R64–R76.
    1. Cassano P., Petrie S.R., Hamblin M.R., Henderson T.A., Iosifescu D.V. Review of transcranial photobiomodulation for major depressive disorder: targeting brain metabolism, inflammation, oxidative stress, and neurogenesis. Neurophotonics. 2016;3:031404.
    1. Morries L.D., Cassano P., Henderson T.A. Treatments for traumatic brain injury with emphasis on transcranial near-infrared laser phototherapy. Neuropsychiatr. Dis. Treat. 2015;11:2159–2175.
    1. Tian F., Hase S.N., Gonzalez-Lima F., Liu H. Transcranial laser stimulation improves human cerebral oxygenation. Lasers Surg. Med. 2016;48:343–349.
    1. Ando T., Xuan W., Xu T., Dai T., Sharma S.K., Kharkwal G.B., Huang Y.Y., Wu Q., Whalen M.J., Sato S., Obara M., Hamblin M.R. Comparison of therapeutic effects between pulsed and continuous wave 810-nm wavelength laser irradiation for traumatic brain injury in mice. PLoS One. 2011;6
    1. Calabrese E.J. Hormesis and medicine. Br. J. Clin. Pharmacol. 2008;66:594–617.
    1. Luckey T.D. Nurture with ionizing radiation: a provocative hypothesis. Nutr. Cancer. 1999;34:1–11.
    1. Huang Y.Y., Chen A.C., Carroll J.D., Hamblin M.R. Biphasic dose response in low level light therapy. Dose-Response. 2009;7:358–383.
    1. Huang Y.Y., Sharma S.K., Carroll J.D., Hamblin M.R. Biphasic dose response in low level light therapy - an update. Dose-Response. 2011;9:602–618.
    1. Wu S., Zhou F., Wei Y., Chen W.R., Chen Q., Xing D. Cancer phototherapy via selective photoinactivation of respiratory chain oxidase to trigger a fatal superoxide anion burst. Antioxid. Redox Signal. 2014;20:733–746.
    1. Hode L. The importance of the coherency. Photomed. Laser Surg. 2005;23:431–434.
    1. Cui X., Bray S., Bryant D.M., Glover G.H., Reiss A.L. A quantitative comparison of NIRS and fMRI across multiple cognitive tasks. NeuroImage. 2011;54:2808–2821.
    1. Haeussinger F.B., Heinzel S., Hahn T., Schecklmann M., Ehlis A.C., Fallgatter A.J. Simulation of near-infrared light absorption considering individual head and prefrontal cortex anatomy: implications for optical neuroimaging. PLoS One. 2011;6
    1. Strangman G.E., Zhang Q., Li Z. Scalp and skull influence on near infrared photon propagation in the Colin27 brain template. NeuroImage. 2014;85(Pt 1):136–149.
    1. Okada E., Delpy D.T. Near-infrared light propagation in an adult head model. II. Effect of superficial tissue thickness on the sensitivity of the near-infrared spectroscopy signal. Appl. Opt. 2003;42:2915–2922.
    1. Jagdeo J.R., Adams L.E., Brody N.I., Siegel D.M. Transcranial red and near infrared light transmission in a cadaveric model. PLoS One. 2012;7
    1. Tedford C.E., DeLapp S., Jacques S., Anders J. Quantitative analysis of transcranial and intraparenchymal light penetration in human cadaver brain tissue. Lasers Surg. Med. 2015;47:312–322.
    1. Lapchak P.A., Boitano P.D., Butte P.V., Fisher D.J., Holscher T., Ley E.J., Nuno M., Voie A.H., Rajput P.S. Transcranial near-infrared laser transmission (NILT) profiles (800 nm): systematic comparison in four common research species. PLoS One. 2015;10
    1. Pitzschke A., Lovisa B., Seydoux O., Zellweger M., Pfleiderer M., Tardy Y., Wagnieres G. Red and NIR light dosimetry in the human deep brain. Phys. Med. Biol. 2015;60:2921–2937.
    1. Pitzschke A., Lovisa B., Seydoux O., Haenggi M., Oertel M.F., Zellweger M., Tardy Y., Wagnieres G. Optical properties of rabbit brain in the red and near-infrared: changes observed under in vivo, postmortem, frozen, and formalin-fixated conditions. J. Biomed. Opt. 2015;20:25006.
    1. Yaroslavsky A.N., Schulze P.C., Yaroslavsky I.V., Schober R., Ulrich F., Schwarzmaier H.J. Optical properties of selected native and coagulated human brain tissues in vitro in the visible and near infrared spectral range. Phys. Med. Biol. 2002;47:2059–2073.
    1. Henderson T.A., Morries L.D. Near-infrared photonic energy penetration: can infrared phototherapy effectively reach the human brain? Neuropsychiatr. Dis. Treat. 2015;11:2191–2208.
    1. Johnstone D.M., el Massri N., Moro C., Spana S., Wang X.S., Torres N., Chabrol C., De Jaeger X., Reinhart F., Purushothuman S., Benabid A.L., Stone J., Mitrofanis J. Indirect application of near infrared light induces neuroprotection in a mouse model of parkinsonism - an abscopal neuroprotective effect. Neuroscience. 2014;274:93–101.
    1. Johnstone D.M., Mitrofanis J., Stone J. Targeting the body to protect the brain: inducing neuroprotection with remotely-applied near infrared light. Neural Regen. Res. 2015;10:349–351.
    1. Farfara D., Tuby H., Trudler D., Doron-Mandel E., Maltz L., Vassar R.J., Frenkel D., Oron U. Low-level laser therapy ameliorates disease progression in a mouse model of Alzheimer's disease. J. Mol. Neurosci. 2015;55:430–436.
    1. Oron A., Oron U. Low-level laser therapy to the bone marrow ameliorates neurodegenerative disease progression in a mouse model of Alzheimer's disease: a minireview. Photomed. Laser Surg. 2016
    1. Iwashita T., Tada T., Zhan H., Tanaka Y., Hongo K. Harvesting blood stem cells from cranial bone at craniotomy–a preliminary study. J. Neuro-Oncol. 2003;64:265–270.
    1. Quah-Smith I., Williams M.A., Lundeberg T., Suo C., Sachdev P. Differential brain effects of laser and needle acupuncture at LR8 using functional MRI. Acupunct. Med. 2013;31:282–289.
    1. Quah-Smith I., Sachdev P.S., Wen W., Chen X., Williams M.A. The brain effects of laser acupuncture in healthy individuals: an FMRI investigation. PLoS One. 2010;5
    1. Sutalangka C., Wattanathorn J., Muchimapura S., Thukham-Mee W., Wannanon P., Tong-un T. Laser acupuncture improves memory impairment in an animal model of Alzheimer's disease. J. Acupunct. Meridian Stud. 2013;6:247–251.
    1. Khongrum J., Wattanathorn J. Laser acupuncture improves behavioral disorders and brain oxidative stress status in the valproic acid rat model of autism. J. Acupunct. Meridian Stud. 2015;8:183–191.
    1. Quah-Smith I., Suo C., Williams M.A., Sachdev P.S. The antidepressant effect of laser acupuncture: a comparison of the resting brain's default mode network in healthy and depressed subjects during functional magnetic resonance imaging. Med. Acupunct. 2013;25:124–133.
    1. Henderson T.A. Multi-watt near-infrared light therapy as a neuroregenerative treatment for traumatic brain injury. Neural Regen. Res. 2016;11:563–565.
    1. Hacke W., Schellinger P.D., Albers G.W., Bornstein N.M., Dahlof B.L., Fulton R., Kasner S.E., Shuaib A., Richieri S.P., Dilly S.G., Zivin J., Lees K.R., Committees N. Investigators, transcranial laser therapy in acute stroke treatment: results of neurothera effectiveness and safety trial 3, a phase III clinical end point device trial. Stroke. 2014;45:3187–3193.
    1. Albin R.L., Miller R.A. Mini-review: retarding aging in murine genetic models of neurodegeneration. Neurobiol. Dis. 2016;85:73–80.
    1. Bey A.L., Jiang Y.H. Overview of mouse models of autism spectrum disorders. Curr. Protoc. Pharmacol. 2014;66 (5 66 61–26)
    1. Leung M.C., Lo S.C., Siu F.K., So K.F. Treatment of experimentally induced transient cerebral ischemia with low energy laser inhibits nitric oxide synthase activity and up-regulates the expression of transforming growth factor-beta 1. Lasers Surg. Med. 2002;31:283–288.
    1. Peplow P.V. Neuroimmunomodulatory effects of transcranial laser therapy combined with intravenous tPA administration for acute cerebral ischemic injury. Neural Regen. Res. 2015;10:1186–1190.
    1. Oron A., Oron U., Chen J., Eilam A., Zhang C., Sadeh M., Lampl Y., Streeter J., DeTaboada L., Chopp M. Low-level laser therapy applied transcranially to rats after induction of stroke significantly reduces long-term neurological deficits. Stroke. 2006;37:2620–2624.
    1. Zhang L., Chen J., Li Y., Zhang Z.G., Chopp M. Quantitative measurement of motor and somatosensory impairments after mild (30 min) and severe (2 h) transient middle cerebral artery occlusion in rats. J. Neurol. Sci. 2000;174:141–146.
    1. Meyer D.M., Chen Y., Zivin J.A. Dose-finding study of phototherapy on stroke outcome in a rabbit model of ischemic stroke. Neurosci. Lett. 2016;630:254–258.
    1. Lapchak P.A., Salgado K.F., Chao C.H., Zivin J.A. 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. 2007;148:907–914.
    1. Detaboada 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;38:70–73.
    1. Lapchak P.A., Wei J., Zivin J.A. Transcranial infrared laser therapy improves clinical rating scores after embolic strokes in rabbits. Stroke. 2004;35:1985–1988.
    1. Lampl Y., Zivin J.A., Fisher M., Lew R., Welin L., Dahlof B., Borenstein P., Andersson B., Perez J., Caparo C., Ilic S., Oron U. Infrared laser therapy for ischemic stroke: a new treatment strategy: results of the NeuroThera Effectiveness and Safety Trial-1 (NEST-1) Stroke. 2007;38:1843–1849.
    1. Huisa B.N., Stemer A.B., Walker M.G., Rapp K., Meyer B.C., Zivin J.A. Nest, investigators, transcranial laser therapy for acute ischemic stroke: a pooled analysis of NEST-1 and NEST-2. Int. J. Stroke. 2013;8:315–320.
    1. Zivin J.A., Sehra R., Shoshoo A., Albers G.W., Bornstein N.M., Dahlof B., Kasner S.E., Howard G., Shuaib A., Streeter J., Richieri S.P., Hacke W., N.-. investigators NeuroThera(R) Efficacy and Safety Trial-3 (NEST-3): a double-blind, randomized, sham-controlled, parallel group, multicenter, pivotal study to assess the safety and efficacy of transcranial laser therapy with the NeuroThera(R) laser system for the treatment of acute ischemic stroke within 24 h of stroke onset. Int. J. Stroke. 2014;9:950–955.
    1. Zivin J.A., Albers G.W., Bornstein N., Chippendale T., Dahlof B., Devlin T., Fisher M., Hacke W., Holt W., Ilic S., Kasner S., Lew R., Nash M., Perez J., Rymer M., Schellinger P., Schneider D., Schwab S., Veltkamp R., Walker M., Streeter J., NeuroThera E., Safety Trial I. Effectiveness and safety of transcranial laser therapy for acute ischemic stroke. Stroke. 2009;40:1359–1364.
    1. Lapchak P.A., Boitano P.D. Transcranial near-infrared laser therapy for stroke: how to recover from futility in the NEST-3 clinical trial. Acta Neurochir. Suppl. 2016;121:7–12.
    1. Lapchak P.A. Fast neuroprotection (fast-NPRX) for acute ischemic stroke victims: the time for treatment is now. Transl. Stroke. Res. 2013;4:704–709.
    1. Lapchak P.A. Recommendations and practices to optimize stroke therapy: developing effective translational research programs. Stroke. 2013;44:841–843.
    1. Lapchak P.A., Zhang J.H., Noble-Haeusslein L.J. RIGOR guidelines: escalating STAIR and STEPS for effective translational research. Transl. Stroke. Res. 2013;4:279–285.
    1. Naeser M., Ho M., Martin P.E., Treglia E.M., Krengel M., Hamblin M.R., Baker E.H. Improved language after scalp application of red/near-infrared light-emitting diodes: pilot study supporting a new, noninvasive treatment for chronic aphasia. Procedia. Soc. Behav. Sci. 2012;61:138–139.
    1. Boonswang N.A., Chicchi M., Lukachek A., Curtiss D. A new treatment protocol using photobiomodulation and muscle/bone/joint recovery techniques having a dramatic effect on a stroke patient's recovery: a new weapon for clinicians. BMJ Case Rep. 2012;2012
    1. Doidge N. Viking Press; New York, NY: 2015. The Brain's Way of Healing: Remarkable Discoveries and Recoveries from the Frontiers of Neuroplasticity.
    1. Oron A., Oron U., Streeter J., de Taboada L., Alexandrovich A., Trembovler V., Shohami E. Low-level laser therapy applied transcranially to mice following traumatic brain injury significantly reduces long-term neurological deficits. J. Neurotrauma. 2007;24:651–656.
    1. Wu Q., Xuan W., Ando T., Xu T., Huang L., Huang Y.Y., Dai T., Dhital S., Sharma S.K., Whalen M.J., Hamblin M.R. Low-level laser therapy for closed-head traumatic brain injury in mice: effect of different wavelengths. Lasers Surg. Med. 2012;44:218–226.
    1. Karu T.I., Pyatibrat L.V., Afanasyeva N.I. Cellular effects of low power laser therapy can be mediated by nitric oxide. Lasers Surg. Med. 2005;36:307–314.
    1. Xuan W., Vatansever F., Huang L., Wu Q., Xuan Y., Dai T., Ando T., Xu T., Huang Y.Y., Hamblin M.R. Transcranial low-level laser therapy improves neurological performance in traumatic brain injury in mice: effect of treatment repetition regimen. PLoS One. 2013;8
    1. Xuan W., Agrawal T., Huang L., Gupta G.K., Hamblin M.R. Low-level laser therapy for traumatic brain injury in mice increases brain derived neurotrophic factor (BDNF) and synaptogenesis. J. Biophotonics. 2015;8:502–511.
    1. Xuan W., Vatansever F., Huang L., Hamblin M.R. Transcranial low-level laser therapy enhances learning, memory, and neuroprogenitor cells after traumatic brain injury in mice. J. Biomed. Opt. 2014;19:108003.
    1. Khuman J., Zhang J., Park J., Carroll J.D., Donahue C., Whalen M.J. Low-level laser light therapy improves cognitive deficits and inhibits microglial activation after controlled cortical impact in mice. J. Neurotrauma. 2012;29:408–417.
    1. Quirk B.J., Torbey M., Buchmann E., Verma S., Whelan H.T. Near-infrared photobiomodulation in an animal model of traumatic brain injury: improvements at the behavioral and biochemical levels. Photomed. Laser Surg. 2012;30:523–529.
    1. Zhang Q., Zhou C., Hamblin M.R., Wu M.X. Low-level laser therapy effectively prevents secondary brain injury induced by immediate early responsive gene X-1 deficiency. J. Cereb. Blood Flow Metab. 2014
    1. Dong T., Zhang Q., Hamblin M.R., Wu M.X. Low-level light in combination with metabolic modulators for effective therapy of injured brain. J. Cereb. Blood Flow Metab. 2015
    1. Naeser M.A., Hamblin M.R. Traumatic brain injury: a major medical problem that could be treated using transcranial, red/near-infrared LED photobiomodulation. Photomed. Laser Surg. 2015
    1. McClure J. The role of causal attributions in public misconceptions about brain injury. Rehabil. Psychol. 2011;56:85–93.
    1. Naeser M.A., Saltmarche A., Krengel M.H., Hamblin M.R., Knight J.A. Improved cognitive function after transcranial, light-emitting diode treatments in chronic, traumatic brain injury: two case reports. Photomed. Laser Surg. 2011;29:351–358.
    1. Naeser M.A., Martin P.I., Lundgren K., Klein R., Kaplan J., Treglia E., Ho M., Nicholas M., Alonso M., Pascual-Leone A. Improved language in a chronic nonfluent aphasia patient after treatment with CPAP and TMS. Cogn. Behav. Neurol. 2010;23:29–38.
    1. Naeser M.A., Zafonte R., Krengel M.H., Martin P.I., Frazier J., Hamblin M.R., Knight J.A., Meehan W.P., III, Baker E.H. Significant improvements in cognitive performance post-transcranial, red/near-infrared light-emitting diode treatments in chronic, mild traumatic brain injury: open-protocol study. J. Neurotrauma. 2014;31:1008–1017.
    1. Henderson T.A., Morries L.D. SPECT perfusion imaging demonstrates improvement of traumatic brain injury with transcranial near-infrared laser phototherapy. Adv. Mind Body Med. 2015;29:27–33.
    1. De Taboada L., Yu J., El-Amouri S., Gattoni-Celli S., Richieri S., McCarthy T., Streeter J., Kindy M.S. Transcranial laser therapy attenuates amyloid-beta peptide neuropathology in amyloid-beta protein precursor transgenic mice. J. Alzheimers Dis. 2011;23:521–535.
    1. (in)
    1. Saltmarche A.E., Naeser M.A., Ho K.F., Hamblin M.R., Lim L. Alzheimer's Association International Conference, Toronto, Canada. 2016. Significant Improvement in Cognition after Transcranial and Intranasal Photobiomodulation: A Controlled, Single-Blind Pilot Study in Participants with Dementia (Abstract)
    1. Maksimovich I.V. Dementia and cognitive impairment reduction after laser transcatheter treatment of Alzheimer's disease. World J. Neurosci. 2015;5
    1. Johnstone D.M., Moro C., Stone J., Benabid A.L., Mitrofanis J. Turning on lights to stop neurodegeneration: the potential of near infrared light therapy in Alzheimer's and Parkinson's disease. Front. Neurosci. 2015;9:500.
    1. Shaw V.E., Spana S., Ashkan K., Benabid A.L., Stone J., Baker G.E., Mitrofanis J. Neuroprotection of midbrain dopaminergic cells in MPTP-treated mice after near-infrared light treatment. J. Comp. Neurol. 2010;518:25–40.
    1. Barcia C. Who else was intoxicated with MPTP in Santa Clara? Parkinsonism Relat. Disord. 2012;18:1005–1006.
    1. Peoples C., Spana S., Ashkan K., Benabid A.L., Stone J., Baker G.E., Mitrofanis J. Photobiomodulation enhances nigral dopaminergic cell survival in a chronic MPTP mouse model of Parkinson's disease. Parkinsonism Relat. Disord. 2012;18:469–476.
    1. Purushothuman S., Nandasena C., Johnstone D.M., 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.
    1. Moro C., Massri N.E., Torres N., Ratel D., De Jaeger X., Chabrol C., Perraut F., Bourgerette A., Berger M., Purushothuman S., Johnstone D., Stone J., Mitrofanis J., Benabid A.L. 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. Neurosurg. 2014;120:670–683.
    1. El Massri N., Moro C., Torres N., Darlot F., Agay D., Chabrol C., Johnstone D.M., Stone J., Benabid A.L., Mitrofanis J. Near-infrared light treatment reduces astrogliosis in MPTP-treated monkeys. Exp. Brain Res. 2016
    1. Moro C., Massri N.E., Darlot F., Torres N., Chabrol C., Agay D., Auboiroux V., Johnstone D.M., Stone J., Mitrofanis J., Benabid A.L. Effects of a higher dose of near-infrared light on clinical signs and neuroprotection in a monkey model of Parkinson's disease. Brain Res. 2016
    1. 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 (abstract) Lasers Surg. Med. 2010;185
    1. Willner P. Validity, reliability and utility of the chronic mild stress model of depression: a 10-year review and evaluation. Psychopharmacology. 1997;134:319–329.
    1. Anisman H., Matheson K. Stress, depression, and anhedonia: caveats concerning animal models. Neurosci. Biobehav. Rev. 2005;29:525–546.
    1. Wu X., Alberico S.L., Moges H., De Taboada L., Tedford C.E., Anders J.J. Pulsed light irradiation improves behavioral outcome in a rat model of chronic mild stress. Lasers Surg. Med. 2012;44:227–232.
    1. Salehpour F., Rasta S.H., Mohaddes G., Sadigh-Eteghad S., Salarirad S. Therapeutic effects of 10-HzPulsed wave lasers in rat depression model: a comparison between near-infrared and red wavelengths. Lasers Surg. Med. 2016
    1. Schiffer F., Johnston A.L., Ravichandran C., Polcari A., Teicher M.H., Webb R.H., Hamblin M.R. 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.
    1. Cassano P., Cusin C., Mischoulon D., Hamblin M.R., De Taboada L., Pisoni A., Chang T., Yeung A., Ionescu D.F., Petrie S.R., Nierenberg A.A., Fava M., Iosifescu D.V. Near-infrared transcranial radiation for major depressive disorder: proof of concept study. Psychiatry J. 2015;2015:352979.
    1. Michalikova S., Ennaceur A., van Rensburg R., Chazot P.L. Emotional responses and memory performance of middle-aged CD1 mice in a 3D maze: effects of low infrared light. Neurobiol. Learn. Mem. 2008;89:480–488.
    1. Gonzalez-Lima F., Barrett D.W. Augmentation of cognitive brain functions with transcranial lasers. Front. Syst. Neurosci. 2014;8:36.
    1. Rojas J.C., Bruchey A.K., Gonzalez-Lima F. Low-level light therapy improves cortical metabolic capacity and memory retention. J. Alzheimers Dis. 2012;32:741–752.
    1. Blanco N.J., Maddox W.T., Gonzalez-Lima F. Improving executive function using transcranial infrared laser stimulation. J. Neuropsychol. 2015
    1. Disner S.G., Beevers C.G., Gonzalez-Lima F. Transcranial laser stimulation as neuroenhancement for attention bias modification in adults with elevated depression symptoms. Brain Stimul. 2016
    1. Salgado A.S., Zangaro R.A., Parreira R.B. Kerppers, II, the effects of transcranial LED therapy (TCLT) on cerebral blood flow in the elderly women. Lasers Med. Sci. 2015;30:339–346.
    1. Kalmar S. The importance of neuropsychopharmacology in the development of psychiatry. Neuropsychopharmacol. Hung. 2014;16:149–156.
    1. Sindi S., Mangialasche F., Kivipelto M. Advances in the prevention of Alzheimer's disease. F1000Prime Rep. 2015;7:50.
    1. Bellou V., Belbasis L., Tzoulaki I., Evangelou E., Ioannidis J.P. Environmental risk factors and Parkinson's disease: an umbrella review of meta-analyses. Parkinsonism Relat. Disord. 2016;23:1–9.
    1. Liebert A.D., Chow R.T., Bicknell B.T., Varigos E. Neuroprotective effects against POCD by photobiomodulation: evidence from assembly/disassembly of the cytoskeleton. J. Exp. Neurosci. 2016;10:1–19.
    1. Ando T., Xuan W., Xu T., Dai T., Sharma S.K., Kharkwal G.B., Huang Y.Y., Wu Q., Whalen M.J., Sato S., Obara M., Hamblin M.R. Comparison of therapeutic effects between pulsed and continuous wave 810-nm wavelength laser irradiation for traumatic brain injury in mice. PLoS One. 2011;6:e26212–e26220.

Source: PubMed

3
Abonneren