Photobiomodulation with near infrared light mitigates Alzheimer's disease-related pathology in cerebral cortex - evidence from two transgenic mouse models

Sivaraman Purushothuman, Daniel M Johnstone, Charith Nandasena, John Mitrofanis, Jonathan Stone, Sivaraman Purushothuman, Daniel M Johnstone, Charith Nandasena, John Mitrofanis, Jonathan Stone

Abstract

Introduction: Previous work has demonstrated the efficacy of irradiating tissue with red to infrared light in mitigating cerebral pathology and degeneration in animal models of stroke, traumatic brain injury, parkinsonism and Alzheimer's disease (AD). Using mouse models, we explored the neuroprotective effect of near infrared light (NIr) treatment, delivered at an age when substantial pathology is already present in the cerebral cortex.

Methods: We studied two mouse models with AD-related pathologies: the K369I tau transgenic model (K3), engineered to develop neurofibrillary tangles, and the APPswe/PSEN1dE9 transgenic model (APP/PS1), engineered to develop amyloid plaques. Mice were treated with NIr 20 times over a four-week period and histochemistry was used to quantify AD-related pathological hallmarks and other markers of cell damage in the neocortex and hippocampus.

Results: In the K3 mice, NIr treatment was associated with a reduction in hyperphosphorylated tau, neurofibrillary tangles and oxidative stress markers (4-hydroxynonenal and 8-hydroxy-2'-deoxyguanosine) to near wildtype levels in the neocortex and hippocampus, and with a restoration of expression of the mitochondrial marker cytochrome c oxidase in surviving neurons. In the APP/PS1 mice, NIr treatment was associated with a reduction in the size and number of amyloid-β plaques in the neocortex and hippocampus.

Conclusions: Our results, in two transgenic mouse models, suggest that NIr may have potential as an effective, minimally-invasive intervention for mitigating, and even reversing, progressive cerebral degenerations.

Figures

Figure 1
Figure 1
Time course of the natural development of cortical pathology in K3 and APP/PS1 mice. (A), (B), (C), (D), (E), (F) Micrographs of hyperphosphorylated tau labelling (red), using the AT8 antibody, in the neocortex (A to C) and hippocampus (D to F) of untreated K3 mice at 3 months (A, D), 6 months (B, E) and 12 months (C, F) of age. (G), (H), (I) Micrographs of amyloid-beta (Aβ) labelling (green), using the 6E10 antibody, in neocortex of untreated APP/PS1 mice at 3 months (G), 4.5 months (H) and 12 months (I) of age. Arrowheads indicate intraneuronal Aβ labelling, arrows indicate extracellular plaques. Comparable immunolabelling was achieved with the 4G8 antibody. Sections were co-labelled for glial fibrillary acidic protein (red), a marker of astrocytes. For all sections, nuclei were labelled with bisbenzimide (blue). Scale in (H) applies to (A) to (G).
Figure 2
Figure 2
Effect of near-infrared light treatment on hyperphosphorylated tau and neurofibrillary tangles in the neocortex of K3 mice. (A), (B) Quantification of tau AT8 immunolabelling, based on average labelling intensity (A) and labelled area (B). All error bars indicate standard error of mean. *P < 0.05, **P < 0.01. (C), (D), (E) Representative photomicrographs of sections stained with Bielschowsky silver stain to demonstrate neurofibrillary tangles (NFTs). Arrows indicate axonal swellings and NFTs. (F), (G), (H) Representative micrographs of AT8 (red) labelling within neurons of the neocortex retrosplenial area. Nuclei were labelled with bisbenzimide (blue). Scale bars = 50 μm; scale in (E) applies to (C) and (D), scale in (H) applies to (F) and (G). NIr, near-infrared light; WT, wildtype.
Figure 3
Figure 3
Effect of near-infrared light treatment on hyperphosphorylated tau and neurofibrillary tangles in the hippocampus of K3 mice. (A), (B) Quantification of tau AT8 immunolabelling, based on average labelling intensity (A) and labelled area (B). All error bars indicate standard error of mean. **P < 0.01, ***P < 0.001. (C), (D), (E) Representative photomicrographs of sections of the hippocampal subiculum area, stained with Bielschowsky silver stain to demonstrate neurofibrillary tangles (NFTs). Arrows indicate axonal swellings and NFTs. The classical silver stain also labelled axons in the white matter core (WM). (F), (G), (H) Representative micrographs of AT8 (red) labelling within hippocampal CA1 pyramidal neurons. Nuclei were labelled with bisbenzimide (blue). Scale bars = 50 μm; scale in (E) applies to (C) and (D), scale in (H) applies to (F) and (G). NIr, near-infrared light; WT, wildtype.
Figure 4
Figure 4
Effect of near-infrared light treatment on oxidative stress markers in the neocortex of K3 mice. (A), (B), (F), (G) Quantification of immunolabelling of two oxidative stress markers, 4-hydroxynonenal (4-HNE; A, B) and 8-hydroxy-2′-deoxyguanosine (8-OHDG; F, G), based on average labelling intensity (A, F) and labelled area (B, G). All error bars indicate standard error of the mean. *P < 0.05, **P < 0.01, ***P < 0.001. (C), (D), (E), (H), (I), (J) Representative micrographs of 4-HNE (red) labelling (C, D, E) and 8-OHDG (red) labelling (H, I, J) within layers IV and V of the neocortical retrosplenial area. Nuclei were labelled with bisbenzimide (blue). Scale bar = 50 μm; scale in (J) applies to all other micrographs. NIr, near-infrared light; WT, wildtype.
Figure 5
Figure 5
Effect of near-infrared light treatment on cytochrome c oxidase labelling in the neocortex and hippocampus of K3 mice. (A), (B), (F), (G) Quantification of immunolabelling of the mitochondrial marker cytochrome c oxidase (COX) in the neocortex retrosplenial area (A, B) and hippocampal CA1 layer (F, G), based on average labelling intensity (A, F) and labelled area (B, G). All error bars indicate standard error of the mean. *P < 0.05, ***P < 0.001. (C), (D), (E), (H), (I), (J) Representative micrographs of COX (red) labelling in the neocortex retrosplenial area (C, D, E) and hippocampal CA1 layer (H, I, J). Nuclei were labelled with bisbenzimide (blue). Scale bar = 50 μm; scale in (J) applies to all other micrographs. NIr, near-infrared light; WT, wildtype.
Figure 6
Figure 6
Effect of near-infrared light on amyloid-beta and plaque pathology in APP/PS1 mice. (A), (B), (C), (D), (E), (F) Quantification of amyloid-beta (Aβ) 4G8 immunolabelling of amyloid plaques in the neocortex (A, B, C) and hippocampus (D, E, F), based on plaque burden (A, D), plaque size (B, E) and number of plaques (C, F). All error bars indicate standard error of the mean. *P < 0.05, ***P < 0.001, ****P < 0.0001. (H), (I), (J), (K), (L), (M) Representative micrographs showing Aβ labelling with the 4G8 antibody (brown) in the neocortex (H, I, J) and hippocampus (K, L, M). Arrows indicate plaques. Scale bar = 100 μm; scale in (M) applies to all other micrographs. DG, dentate gyrus of hippocampus; NIr, near-infrared light; SLM, stratum lacunosum moleculare; WT, wildtype.
Figure 7
Figure 7
Effect of near-infrared light on Congo red-positive plaque numbers in APP/PS1 mice. (A), (B) Quantification of Congo red-positive plaque counts in the neocortex (A) and hippocampus (B). All error bars indicate standard error of the mean. (C), (D), (E), (F), (G), (H) Representative micrographs showing Congo red staining of plaques in the neocortex (C, E, G) and hippocampus (D, F, H). Arrows indicate plaques. Scale bar = 50 μm; scale in (H) applies to all other micrographs. DG, dentate gyrus of hippocampus; NIr, near-infrared light; SLM, stratum lacunosum moleculare; WT, wildtype.

References

    1. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;6:353–356. doi: 10.1126/science.1072994.
    1. Braak H, Braak E. Staging of Alzheimer’s disease-related neurofibrillary changes. Neurobiol Aging. 1995;6:271–278. doi: 10.1016/0197-4580(95)00021-6.
    1. Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, Jones PK, Ghanbari H, Wataya T, Shimohama S, Chiba S, Atwood CS, Petersen RB, Smith MA. Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol. 2001;6:759–767.
    1. Yao J, Irwin RW, Zhao L, Nilsen J, Hamilton RT, Brinton RD. Mitochondrial bioenergetic deficit precedes Alzheimer’s pathology in female mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA. 2009;6:14670–14675. doi: 10.1073/pnas.0903563106.
    1. Stone J. What initiates the formation of senile plaques? The origin of Alzheimer-like dementias in capillary haemorrhages. Med Hypotheses. 2008;6:347–359. doi: 10.1016/j.mehy.2008.04.007.
    1. Calabrese V, Guagliano E, Sapienza M, Panebianco M, Calafato S, Puleo E, Pennisi G, Mancuso C, Butterfield DA, Stella AG. Redox regulation of cellular stress response in aging and neurodegenerative disorders: role of vitagenes. Neurochem Res. 2007;6:757–773. doi: 10.1007/s11064-006-9203-y.
    1. Whelan HT, Smits RL Jr, Buchman EV, Whelan NT, Turner SG, Margolis DA, Cevenini V, Stinson H, Ignatius R, Martin T, Martin T, Cwiklinski J, Philippi AF, Graf WR, Hodgson B, Gould L, Kane M, Chen G, Caviness J. Effect of NASA light-emitting diode irradiation on wound healing. J Clin Laser Med Surg. 2001;6:305–314. doi: 10.1089/104454701753342758.
    1. Desmet KD, Paz DA, Corry JJ, Eells JT, Wong-Riley MT, Henry MM, Buchmann EV, Connelly MP, Dovi JV, Liang HL, Henshel DS, Yeager RL, Millsap DS, Lim J, Gould LJ, Das R, Jett M, Hodgson BD, Margolis D, Whelan HT. Clinical and experimental applications of NIR-LED photobiomodulation. Photomed Laser Surg. 2006;6:121–128. doi: 10.1089/pho.2006.24.121.
    1. Eells JT, Wong-Riley MT, VerHoeve J, Henry M, Buchman EV, Kane MP, Gould LJ, Das R, Jett M, Hodgson BD, Margolis D, Whelan HT. Mitochondrial signal transduction in accelerated wound and retinal healing by near-infrared light therapy. Mitochondrion. 2004;6:559–567. doi: 10.1016/j.mito.2004.07.033.
    1. 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;6:1801–1822.
    1. Xuan W, Vatansever F, Huang L, Wu Q, Xuan Y, Dai T, Ando T, Xu T, Huang YY, Hamblin MR. Transcranial low-level laser therapy improves neurological performance in traumatic brain injury in mice: effect of treatment repetition regimen. PLoS One. 2013;6:e53454. doi: 10.1371/journal.pone.0053454.
    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;6:651–656. doi: 10.1089/neu.2006.0198.
    1. Moro C, Torres N, El Massri N, Ratel D, Johnstone DM, Stone J, Mitrofanis J, Benabid AL. Photobiomodulation preserves behaviour and midbrain dopaminergic cells from MPTP toxicity: evidence from two mouse strains. BMC Neurosci. 2013;6:40. doi: 10.1186/1471-2202-14-40.
    1. Shaw VE, Spana S, Ashkan K, Benabid AL, Stone J, Baker GE, Mitrofanis J. Neuroprotection of midbrain dopaminergic cells in MPTP-treated mice after near-infrared light treatment. J Comp Neurol. 2010;6:25–40. doi: 10.1002/cne.22207.
    1. Peoples C, Spana S, Ashkan K, Benabid AL, Stone J, Baker GE, Mitrofanis J. Photobiomodulation enhances nigral dopaminergic cell survival in a chronic MPTP mouse model of Parkinson’s disease. Parkinsonism Relat Disord. 2012;6:469–476. doi: 10.1016/j.parkreldis.2012.01.005.
    1. Grillo SL, Duggett NA, Ennaceur A, Chazot PL. Non-invasive infra-red therapy (1072 nm) reduces beta-amyloid protein levels in the brain of an Alzheimer’s disease mouse model, TASTPM. J Photochem Photobiol B. 2013;6:13–22.
    1. De Taboada L, Yu J, El-Amouri S, Gattoni-Celli S, Richieri S, McCarthy T, Streeter J, Kindy MS. Transcranial laser therapy attenuates amyloid-beta peptide neuropathology in amyloid-beta protein precursor transgenic mice. J Alzheimers Dis. 2011;6:521–535.
    1. Lampl Y, Zivin JA, 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;6:1843–1849. doi: 10.1161/STROKEAHA.106.478230.
    1. Schiffer F, Johnston AL, Ravichandran C, Polcari A, Teicher MH, Webb RH, Hamblin MR. 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;6:46. doi: 10.1186/1744-9081-5-46.
    1. Tuby H, Hertzberg E, Maltz L, Oron U. Long-term safety of low-level laser therapy at different power densities and single or multiple applications to the bone marrow in mice. Photomed Laser Surg. 2013;6:269–273. doi: 10.1089/pho.2012.3395.
    1. Ittner LM, Fath T, Ke YD, Bi M, van Eersel J, Li KM, Gunning P, Gotz J. Parkinsonism and impaired axonal transport in a mouse model of frontotemporal dementia. Proc Natl Acad Sci USA. 2008;6:15997–16002. doi: 10.1073/pnas.0808084105.
    1. van Eersel J, Ke YD, Liu X, Delerue F, Kril JJ, Gotz J, Ittner LM. Sodium selenate mitigates tau pathology, neurodegeneration, and functional deficits in Alzheimer’s disease models. Proc Natl Acad Sci USA. 2010;6:13888–13893. doi: 10.1073/pnas.1009038107.
    1. Jankowsky JL, Fadale DJ, Anderson J, Xu GM, Gonzales V, Jenkins NA, Copeland NG, Lee MK, Younkin LH, Wagner SL, Younkin SG, Borchelt DR. Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase. Hum Mol Genet. 2004;6:159–170.
    1. Garcia-Alloza M, Robbins EM, Zhang-Nunes SX, Purcell SM, Betensky RA, Raju S, Prada C, Greenberg SM, Bacskai BJ, Frosch MP. Characterization of amyloid deposition in the APPswe/PS1dE9 mouse model of Alzheimer disease. Neurobiol Dis. 2006;6:516–524. doi: 10.1016/j.nbd.2006.08.017.
    1. Cao D, Lu H, Lewis TL, Li L. Intake of sucrose-sweetened water induces insulin resistance and exacerbates memory deficits and amyloidosis in a transgenic mouse model of Alzheimer disease. J Biol Chem. 2007;6:36275–36282. doi: 10.1074/jbc.M703561200.
    1. Truett GE, Heeger P, Mynatt RL, Truett AA, Walker JA, Warman ML. Preparation of PCR-quality mouse genomic DNA with hot sodium hydroxide and tris (HotSHOT) Biotechniques. 2000;6:52–54.
    1. 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;6:61–70.
    1. Purushothuman S, Marotte L, Stowe S, Johnstone DM, Stone J. The response of cerebral cortex to haemorrhagic damage: experimental evidence from a penetrating injury model. PLoS One. 2013;6:e59740. doi: 10.1371/journal.pone.0059740.
    1. Wilcock DM, Gordon MN, Morgan D. Quantification of cerebral amyloid angiopathy and parenchymal amyloid plaques with Congo red histochemical stain. Nat Protoc. 2006;6:1591–1595. doi: 10.1038/nprot.2006.277.
    1. Yan Q, Zhang J, Liu H, Babu-Khan S, Vassar R, Biere AL, Citron M, Landreth G. Anti-inflammatory drug therapy alters beta-amyloid processing and deposition in an animal model of Alzheimer’s disease. J Neurosci. 2003;6:7504–7509.
    1. Augustinack JC, Schneider A, Mandelkow EM, Hyman BT. Specific tau phosphorylation sites correlate with severity of neuronal cytopathology in Alzheimer’s disease. Acta Neuropathol. 2002;6:26–35. doi: 10.1007/s004010100423.
    1. Bruck W, Bitsch A, Kolenda H, Bruck Y, Stiefel M, Lassmann H. Inflammatory central nervous system demyelination: correlation of magnetic resonance imaging findings with lesion pathology. Ann Neurol. 1997;6:783–793. doi: 10.1002/ana.410420515.
    1. Sayre LM, Zelasko DA, Harris PL, Perry G, Salomon RG, Smith MA. 4-Hydroxynonenal-derived advanced lipid peroxidation end products are increased in Alzheimer’s disease. J Neurochem. 1997;6:2092–2097.
    1. Mecocci P, MacGarvey U, Beal MF. Oxidative damage to mitochondrial DNA is increased in Alzheimer’s disease. Ann Neurol. 1994;6:747–751. doi: 10.1002/ana.410360510.
    1. Trinchese F, Liu S, Battaglia F, Walter S, Mathews PM, Arancio O. Progressive age-related development of Alzheimer-like pathology in APP/PS1 mice. Ann Neurol. 2004;6:801–814. doi: 10.1002/ana.20101.
    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;6:2620–2624. doi: 10.1161/01.STR.0000242775.14642.b8.
    1. Blanchard V, Moussaoui S, Czech C, Touchet N, Bonici B, Planche M, Canton T, Jedidi I, Gohin M, Wirths O, Bayer TA, Langui D, Duyckaerts C, Tremp G, Pradier L. Time sequence of maturation of dystrophic neurites associated with Aβ deposits in APP/PS1 transgenic mice. Exp Neurol. 2003;6:247–263. doi: 10.1016/S0014-4886(03)00252-8.
    1. Karu T. Mitochondrial mechanisms of photobiomodulation in context of new data about multiple roles of ATP. Photomed Laser Surg. 2010;6:159–160. doi: 10.1089/pho.2010.2789.
    1. Wilden L, Karthein R. Import of radiation phenomena of electrons and therapeutic low-level laser in regard to the mitochondrial energy transfer. J Clin Laser Med Surg. 1998;6:159–165.
    1. Wong-Riley MT, Bai X, Buchmann E, Whelan HT. Light-emitting diode treatment reverses the effect of TTX on cytochrome oxidase in neurons. Neuroreport. 2001;6:3033–3037. doi: 10.1097/00001756-200110080-00011.
    1. Hou JF, Zhang H, Yuan X, Li J, Wei YJ, Hu SS. In vitro effects of low-level laser irradiation for bone marrow mesenchymal stem cells: proliferation, growth factors secretion and myogenic differentiation. Lasers Surg Med. 2008;6:726–733. doi: 10.1002/lsm.20709.
    1. 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;6:682–688. doi: 10.1002/lsm.20377.
    1. 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;6:401–409. doi: 10.1002/lsm.21063.
    1. Stamer K, Vogel R, Thies E, Mandelkow E, Mandelkow EM. Tau blocks traffic of organelles, neurofilaments, and APP vesicles in neurons and enhances oxidative stress. J Cell Biol. 2002;6:1051–1063. doi: 10.1083/jcb.200108057.
    1. Aliev G, Smith MA, Seyidov D, Neal ML, Lamb BT, Nunomura A, Gasimov EK, Vinters HV, Perry G, LaManna JC, Friedland RP. The role of oxidative stress in the pathophysiology of cerebrovascular lesions in Alzheimer’s disease. Brain Pathol. 2002;6:21–35.
    1. Hamel E, Nicolakakis N, Aboulkassim T, Ongali B, Tong XK. Oxidative stress and cerebrovascular dysfunction in mouse models of Alzheimer’s disease. Exp Physiol. 2008;6:116–120.
    1. Zhu X, Perry G, Moreira PI, Aliev G, Cash AD, Hirai K, Smith MA. Mitochondrial abnormalities and oxidative imbalance in Alzheimer disease. J Alzheimers Dis. 2006;6:147–153.
    1. Zhang X, Le W. Pathological role of hypoxia in Alzheimer’s disease. Exp Neurol. 2010;6:299–303. doi: 10.1016/j.expneurol.2009.07.033.
    1. Wen Y, Yang S, Liu R, Brun-Zinkernagel AM, Koulen P, Simpkins JW. Transient cerebral ischemia induces aberrant neuronal cell cycle re-entry and Alzheimer’s disease-like tauopathy in female rats. J Biol Chem. 2004;6:22684–22692. doi: 10.1074/jbc.M311768200.
    1. Uttara B, Singh AV, Zamboni P, Mahajan RT. Oxidative stress and neurodegenerative diseases: a review of upstream and downstream antioxidant therapeutic options. Curr Neuropharmacol. 2009;6:65–74. doi: 10.2174/157015909787602823.

Source: PubMed

3
Suscribir