Novel Invisible Spectral Flicker Induces 40 Hz Neural Entrainment with Similar Spatial Distribution as 40 Hz Stroboscopic Light

Mikkel Pejstrup Agger, Marcus Schultz Carstensen, Mark Alexander Henney, Luna Skytte Hansen, Anders Ohlhues Baandrup, Mai Nguyen, Paul Michael Petersen, Kristoffer Hougaard Madsen, Troels Wesenberg Kjær, Mikkel Pejstrup Agger, Marcus Schultz Carstensen, Mark Alexander Henney, Luna Skytte Hansen, Anders Ohlhues Baandrup, Mai Nguyen, Paul Michael Petersen, Kristoffer Hougaard Madsen, Troels Wesenberg Kjær

Abstract

Background: Exposure to 40 Hz stroboscopic light, for one hour a day, has previously been published as a potential treatment option for Alzheimer's disease in animal models. However, exposure for an hour a day to 40 Hz stroboscopic light can be strenuous and examining other types of 40 Hz inducing stimuli is paramount if chronic treatment is wanted.

Objective: A core assumption behind ensuring a therapeutic outcome is that the visual stimuli can induce 40 Hz gamma entrainment. Here, we examine whether a specific visual stimulus, 40 Hz invisible spectral flicker (ISF), can induce gamma entrainment and how it differs from both continuous light (CON) and 40 Hz stroboscopic light (STROBE).

Methods: The study included non-simultaneous EEG-fMRI neuroimaging of 13 young healthy volunteers during light exposure. Each light condition (i.e., CON, ISF, or STROBE) was active for 30 seconds followed immediately by the next.

Results: Entrainment of 40 Hz neural activity were significantly higher signal-to-noise ratio during exposure to ISF (mean: 3.03, 95% CI 2.07 to 3.99) and STROBE (mean: 12.04, 95% CI 10.18 to 13.87) compared to CON. Additionally STROBE had a higher entrainment than ISF (mean: 9.01, 95% CI 7.16 to 12.14).

Conclusion: This study presents a novel method of 40 Hz entrainment using ISF. This enables the possibility of future randomized placebo-controlled clinical trials with acceptable double blinding due to the essentially imperceivable flicker, which is expected to substantially reduce discomfort compared to interventions with stroboscopic flicker.

Keywords: 40 Hz; Alzheimer’s disease; GENUS; electroencephalograph; functional MRI; gamma entrainment; invisible spectral flicker; light-based neurostimulation; steady state visually evoked potentials.

Conflict of interest statement

Authors’ disclosures available online (https://www.j-alz.com/manuscript-disclosures/22-0081r1).

Figures

Fig. 1
Fig. 1
Experimental design. A) Normalized spectral irradiance of the different light conditions used, top panel: non-flickering light (CON), middle panel: 40 Hz ISF, lower panel: 40 Hz stroboscopic light. B) Normalized spectral irradiance of the 1st and 2nd half cycle of the different conditions. C) Color temperature of the three light conditions used. D) Illustration of the AB and ABC paradigms.
Fig. 2
Fig. 2
Experimental setup in the MR scanner. Fiber optic cables projected light to the eyes of the subject within the MR scanner. Each fiber optic cable was positioned 5 cm from the medial corner of each eye to achieve equal light intensity at the level of the eyes to that used in the EEG setup.
Fig. 3
Fig. 3
40 Hz neural activity from the AB paradigm. A) Shows the PSD for both continuous non-flickering light (CON) and 40 Hz ISF (ISF). The lower panel is zoomed in to more accurately represents frequencies around 40 Hz. B) Boxplot of signal-to-noise ratios (SNR) for both conditions. C) Topographical distribution of 40 Hz activity during exposure to continuous non-flickering light. D) As C, but for exposure to 40 Hz ISF. Note the difference in color bar to better visualize the spatial distributions of the induced 40 Hz neural activity.
Fig. 4
Fig. 4
40 Hz neural activity from the ABC paradigm. A) Shows the PSD for both continuous non-flickering light (CON), 40 Hz ISF (ISF), and 40 Hz stroboscopic light. The lower panel is zoomed in to more accurately represent frequencies around 40 Hz. B) Boxplot of signal-to-noise ratios for all three conditions. C) Topographical distribution of 40 Hz activity during exposure to continuous non-flickering light. D) As C, but for exposure to 40 Hz ISF. E) As C and D, but for 40 Hz stroboscopic light. Note the difference in color bar to better visualize the spatial distributions of the induced 40 Hz neural activity.
Fig. 5
Fig. 5
Confusion matrices of the multivariate pattern analysis of within-subject comparison with 1000 permutations. A) AB paradigm comparing non-flickering continuous light to 40 Hz ISF. B) ABC paradigm comparing non-flickering continuous light to 40 Hz ISF. C) ABC paradigm comparing non-flickering continuous light to 40 Hz Stroboscopic light. D) Control paradigm testing no light stimulus against non-flickering lights.

References

    1. GBD 2016 Dementia Collaborators (2019) Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990-2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol 18, 88–106.
    1. Long JM, Holtzman DM (2019) Alzheimer disease: An update on pathobiology and treatment strategies. Cell 179, 312–339.
    1. Congdon EE, Sigurdsson EM (2018) Tau-targeting therapies for Alzheimer disease. Nat Rev Neurol 14, 399–415.
    1. Iaccarino HF, Singer AC, Martorell AJ, Rudenko A, Gao F, Gillingham TZ, Mathys H, Seo J, Kritskiy O, Abdurrob F, Adaikkan C, Canter RG, Rueda R, Brown EN, Boyden ES, Tsai L-H (2016) Gamma frequency entrainment attenuates amyloid load and modifies microglia. Nature 540, 230–235.
    1. Martorell AJ, Paulson AL, Suk H-J, Abdurrob F, Drummond GT, Guan W, Young JZ, Kim DN-W, Kritskiy O, Barker SJ, Mangena V, Prince SM, Brown EN, Chung K, Boyden ES, Singer AC, Tsai L-H (2019) Multi-sensory gamma stimulation ameliorates Alzheimer’s-associated pathology and improves cognition. Cell 177, 256–271.e22.
    1. Adaikkan C, Middleton SJ, Marco A, Pao P-C, Mathys H, Kim DN-W, Gao F, Young JZ, Suk H-J, Boyden ES, McHugh TJ, Tsai L-H (2019) Gamma entrainment binds higher-order brain regions and offers neuroprotection. Neuron 102, 929–943.e8.
    1. Herrmann CS (2001) Human EEG responses to 1-100 Hz flicker: Resonance phenomena in visual cortex and their potential correlation to cognitive phenomena. Exp Brain Res 137, 346–353.
    1. Jones M, McDermott B, Oliveira BL, O’Brien A, Coogan D, Lang M, Moriarty N, Dowd E, Quinlan L, Mc Ginley B, Dunne E, Newell D, Porter E, Elahi MA, O’ Halloran M, Shahzad A (2019) Gamma band light stimulation in human case studies: Groundwork for potential Alzheimer’s disease treatment. J Alzheimers Dis 70, 171–185.
    1. Ismail R, Hansen AK, Parbo P, Brændgaard H, Gottrup H, Brooks DJ, Borghammer P (2018) The effect of 40-Hz light therapy on amyloid load in patients with prodromal and clinical Alzheimer’s disease. Int J Alzheimers Dis 2018, 6852303.
    1. Chan D, Suk H-J, Jackson B, Milman NP, Stark D, Klerman EB, Kitchener E, Avalos VSF, Banerjee A, Beach SD, Blanchard J, Stearns C, Boes A, Uitermarkt B, Gander P, Howard M, Sternberg EJ, Nieto-Castanon A, Anteraper S, Whitfield-Gabrieli S, Brown EN, Boyden ES, Dickerson B, Tsai L-H (2021) 40Hz sensory stimulation induces gamma entrainment and affects brain structure, sleep and cognition in patients with Alzheimer’s dementia. medRxiv, 2021.03.01.21252717.
    1. Cimenser A, Hempel E, Travers T, Strozewski N, Martin K, Malchano Z, Hajós M (2021) Sensory-evoked 40-Hz gamma oscillation improves sleep and daily living activities in Alzheimer’s disease patients. Front Syst Neurosci 15, 746859.
    1. Carstensen MS, Lindén J, Nguyen NM, Hansen HE, Feijóo Carrillo GM, Hansen LS, Corell DD, Broeng J, Kriegsfeld LJ, Petersen PM (2020) 40 Hz invisible spectral flicker and its potential use in Alzheimer’s light therapy treatment. In Mechanisms of Photobiomodulation Therapy XV, Hamblin MR, Carroll JD, Arany P, eds. SPIE, San Francisco, p. 20.
    1. Terman M, Terman JS (2005) Light therapy for seasonal and nonseasonal depression: Efficacy, protocol, safety, and side effects. CNS Spectr 10, 647–663.
    1. Liu C-R, Liou YM, Jou J-H (2021) Pilot study of the effects of bright ambient therapy on dementia symptoms and cognitive function. Front Psychol 12, 782160.
    1. Menon V, Crottaz-Herbette S (2005) Combined EEG and fMRI studies of human brain function. Int Rev Neurobiol 66, 291–321.
    1. Lachaux J-P, Fonlupt P, Kahane P, Minotti L, Hoffmann D, Bertrand O, Baciu M (2007) Relationship between task-related gamma oscillations and BOLD signal: New insights from combined fMRI and intracranial EEG. Hum Brain Mapp 28, 1368–1375.
    1. Leicht G, Björklund J, Vauth S, Mußmann M, Haaf M, Steinmann S, Rauh J, Mulert C (2021) Gamma-band synchronisation in a frontotemporal auditory information processing network. Neuroimage 239, 118307.
    1. Mulert C, Leicht G, Hepp P, Kirsch V, Karch S, Pogarell O, Reiser M, Hegerl U, Jäger L, Moller HJ, McCarley RW (2010) Single-trial coupling of the gamma-band response and the corresponding BOLD signal. Neuroimage 49, 2238–2247.
    1. Bayram A, Bayraktaroglu Z, Karahan E, Erdogan B, Bilgic B, Özker M, Kasikci I, Duru AD, Ademoglu A, Öztürk C, Arikan K, Tarhan N, Demiralp T (2011) Simultaneous EEG/fMRI analysis of the resonance phenomena in steady-state visual evoked responses. Clin EEG Neurosci 42, 98–106.
    1. Emir UE, Bayraktaroglu Z, Ozturk C, Ademoglu A, Demiralp T (2008) Changes in BOLD transients with visual stimuli across 1–44Hz. Neurosci Lett 436, 185–188.
    1. Murta T, Chaudhary UJ, Tierney TM, Dias A, Leite M, Carmichael DW, Figueiredo P, Lemieux L (2017) Phase–amplitude coupling and the BOLD signal: A simultaneous intracranial EEG (icEEG) - fMRI study in humans performing a finger-tapping task. NeuroImage 146, 438–451.
    1. Stern JM, Engel J (2013) Atlas of EEG patterns chapter 4, Wolters Kluwer Lippincott Williams & Wilkins, Philadelphia.
    1. Gramfort A (2013) MEG and EEG data analysis with MNE-Python. Front Neurosci 7, 267.
    1. Yao D (2001) A method to standardize a reference of scalp EEG recordings to a point at infinity. Physiol Meas 22, 693–711.
    1. Perrin F, Pernier J, Bertrand O, Echallier JF (1989) Spherical splines for scalp potential and current density mapping. Electroencephalogr Clin Neurophysiol 72, 184–187.
    1. Meigen T, Bach M (1999) On the statistical significance of electrophysiological steady-state responses. Doc Ophthalmol Adv Ophthalmol 98, 207–232.
    1. Norcia AM, Appelbaum LG, Ales JM, Cottereau BR, Rossion B (2015) The steady-state visual evoked potential in vision research: A review. J Vis 15, 4.
    1. Ashburner J, Friston KJ (2005) Unified segmentation. Neuroimage 26, 839–851.
    1. Evans AC, Collins DL, Mills SR, Brown ED, Kelly RL, Peters TM (1993) 3D statistical neuroanatomical models from 305 MRI volumes. In 1993 IEEE Conference Record Nuclear Science Symposium and Medical Imaging Conference IEEE, San Francisco, CA, USA, pp. 1813–1817.
    1. (2007), Statistical Parametric Mapping, Elsevier.
    1. Rivolta D, Woolgar A, Palermo R, Butko M, Schmalzl L, Williams MA (2014) Multi-voxel pattern analysis (MVPA) reveals abnormal fMRI activity in both the “core” and “extended” face network in congenital prosopagnosia. Front Hum Neurosci 8, 925.
    1. Haxby JV, Gobbini MI, Furey ML, Ishai A, Schouten JL, Pietrini P (2001) Distributed and overlapping representations of faces and objects in ventral temporal cortex. Science 293, 2425–2430.
    1. Benjamini Y, Hochberg Y (1995) Controlling the false discovery rate: A practical and powerful approach to multiple testing. J R Stat Soc Ser B Methodol 57, 289–300.
    1. Pedregosa F, Varoquaux G, Gramfort A, Michel V, Thirion B, Grisel O, Blondel M, Prettenhofer P, Weiss R, Dubourg V, Vanderplas J, Passos A, Cournapeau D, Brucher M, Perrot M, Duchesnay É (2011) Scikit-learn: Machine learning in Python. J Mach Learn Res 12, 2825–2830.
    1. Abraham A, Pedregosa F, Eickenberg M, Gervais P, Mueller A, Kossaifi J, Gramfort A, Thirion B, Varoquaux G (2014) Machine learning for neuroimaging with scikit-learn. Front Neuroinformatics 8, 14.
    1. Laconte S, Strother S, Cherkassky V, Anderson J, Hu X (2005) Support vector machines for temporal classification of block design fMRI data. Neuroimage 26, 317–329.
    1. Ojala M, Garriga GC (2009) Permutation tests for studying classifier performance. In 2009 Ninth IEEE International Conference on Data Mining IEEE, Miami Beach, FL, USA, pp. 908–913.
    1. Nichols TE, Holmes AP (2002) Nonparametric permutation tests for functional neuroimaging: A primer with examples. Hum Brain Mapp 15, 1–25.
    1. RStudio Team (2015) RStudio: Integrated Development Environment for R, Boston MA.
    1. Bates D, Mächler M, Bolker B, Walker S (2015) Fitting Linear Mixed-Effects Models Using lme4. J Stat Softw 67, 1–48.
    1. Kuznetsova A, Brockhoff PB, Christensen RHB (2017) lmerTest Package: Tests in linear mixed effects models. J Stat Softw 82, 1–26.
    1. Adaikkan C, Tsai L-H (2020) Gamma entrainment: Impact on neurocircuits, glia, and therapeutic opportunities. Trends Neurosci 43, 24–41.
    1. Suzuki Y, Minami T, Nakauchi S (2019) Pupil constriction in the glare illusion modulates the steady-state visual evoked potentials. Neuroscience 416, 221–228.
    1. Zibrandtsen IC, Agger M, Kjaer TW (2020) Gamma entrainment in a large retrospective cohort: Implications for photic stimulation therapy for Alzheimer’s disease. J Alzheimers Dis 75, 1181–1190.
    1. Vialatte F-B, Maurice M, Dauwels J, Cichocki A (2010) Steady-state visually evoked potentials: Focus on essential paradigms and future perspectives. Prog Neurobiol 90, 418–438.
    1. Josephs O, Henson RNA (1999) Event-related functional magnetic resonance imaging: Modelling, inference and optimization. Philos Trans R Soc Lond B Biol Sci 354, 1215–1228.
    1. Sohal VS (2016) How close are we to understanding what (if anything) oscillations do in cortical circuits? J Neurosci 36, 10489–10495.
    1. Buzsáki G, Wang X-J (2012) Mechanisms of gamma oscillations. Annu Rev Neurosci 35, 203–225.
    1. Cardin JA, Carlén M, Meletis K, Knoblich U, Zhang F, Deisseroth K, Tsai L-H, Moore CI (2009) Driving fast-spiking cells induces gamma rhythm and controls sensory responses. Nature 459, 663–667.
    1. Sohal VS, Zhang F, Yizhar O, Deisseroth K (2009) Parvalbumin neurons and gamma rhythms enhance cortical circuit performance. Nature 459, 698–702.
    1. Siegle JH, Pritchett DL, Moore CI (2014) Gamma-range synchronization of fast-spiking interneurons can enhance detection of tactile stimuli. Nat Neurosci 17, 1371–1379.
    1. Kim H, Ährlund-Richter S, Wang X, Deisseroth K, Carlén M (2016) Prefrontal parvalbumin neurons in control of attention. Cell 164, 208–218.
    1. Rizzo M (2000) Vision and cognition in Alzheimer’s disease. Neuropsychologia 38, 1157–1169.

Source: PubMed

3
Suscribir