Feasibility and preliminary efficacy for morning bright light therapy to improve sleep and plasma biomarkers in US Veterans with TBI. A prospective, open-label, single-arm trial

Jonathan E Elliott, Alisha A McBride, Nadir M Balba, Stanley V Thomas, Cassandra L Pattinson, Benjamin J Morasco, Andrea Wilkerson, Jessica M Gill, Miranda M Lim, Jonathan E Elliott, Alisha A McBride, Nadir M Balba, Stanley V Thomas, Cassandra L Pattinson, Benjamin J Morasco, Andrea Wilkerson, Jessica M Gill, Miranda M Lim

Abstract

Mild traumatic brain injury (TBI) is associated with persistent sleep-wake dysfunction, including insomnia and circadian rhythm disruption, which can exacerbate functional outcomes including mood, pain, and quality of life. Present therapies to treat sleep-wake disturbances in those with TBI (e.g., cognitive behavioral therapy for insomnia) are limited by marginal efficacy, poor patient acceptability, and/or high patient/provider burden. Thus, this study aimed to assess the feasibility and preliminary efficacy of morning bright light therapy, to improve sleep in Veterans with TBI (NCT03578003). Thirty-three Veterans with history of TBI were prospectively enrolled in a single-arm, open-label intervention using a lightbox (~10,000 lux at the eye) for 60-minutes every morning for 4-weeks. Pre- and post-intervention outcomes included questionnaires related to sleep, mood, TBI, post-traumatic stress disorder (PTSD), and pain; wrist actigraphy as a proxy for objective sleep; and blood-based biomarkers related to TBI/sleep. The protocol was rated favorably by ~75% of participants, with adherence to the lightbox and actigraphy being ~87% and 97%, respectively. Post-intervention improvements were observed in self-reported symptoms related to insomnia, mood, and pain; actigraphy-derived measures of sleep; and blood-based biomarkers related to peripheral inflammatory balance. The severity of comorbid PTSD was a significant positive predictor of response to treatment. Morning bright light therapy is a feasible and acceptable intervention that shows preliminary efficacy to treat disrupted sleep in Veterans with TBI. A full-scale randomized, placebo-controlled study with longitudinal follow-up is warranted to assess the efficacy of morning bright light therapy to improve sleep, biomarkers, and other TBI related symptoms.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. CONSORT diagram.
Fig 1. CONSORT diagram.
The CONSORT diagram outlining the recruitment, flow, data processing and final n’s for select key outcome measures.
Fig 2. Sleep related questionnaire data.
Fig 2. Sleep related questionnaire data.
Pre- and post-morning bright light therapy questionnaire scores for the (A) Sleep Hygiene Index (SHI; range = 0–52; higher = worse sleep hygiene) and (C) Insomnia Severity Index (ISI; range = 0–28; higher = worse insomnia) displayed as truncated violin plots with moderate smoothing (dashed line = median score; dotted lines = 25% and 75% quartiles). Percent change in each Individual for (B) SHI and (D) ISI, normalized to the maximum score of each questionnaire, plotted with the mean and standard deviation overlaid. Shaded symbols are subjects with co-morbid post-traumatic stress disorder (PTSD), open symbols are subjects without co-morbid PTSD. * = p <0.05 vs pre; paired two-tail t-test.
Fig 3. TBI-related symptom severity.
Fig 3. TBI-related symptom severity.
Pre- and post-morning bright light therapy questionnaire scores for the (A) Neurobehavioral Symptom Inventory (NSI; range = 0–88; higher = worse brain injury related symptom severity) and (C) Post-traumatic stress disorder (PTSD) checklist-5 (PCL-5; range = 0–80; higher = worse PTSD symptom severity) displayed as truncated violin plots with moderate smoothing (dashed line = median score; dotted lines = 25% and 75% quartiles). Percent change in each Individual for (B) NSI and (D) PCL-5 normalized to the maximum score of each questionnaire, plotted with the mean and standard deviation overlaid. Shaded symbols are subjects with co-morbid post-traumatic stress disorder (PTSD), open symbols are subjects without co-morbid PTSD. * = p <0.05 vs pre; paired two-tail t-test.
Fig 4. Mood and anxiety.
Fig 4. Mood and anxiety.
Pre- and post-morning bright light therapy questionnaire scores for the (A) Patient Health Questionnaire-9 (PHQ-9; range = 0–27; higher = worse depressive symptom severity) and (C) NIH PROMIS Emotional-Distress and Anxiety (EDA; range 4–20; higher = worse symptom severity) displayed as truncated violin plots with moderate smoothing (dashed line = median score; dotted lines = 25% and 75% quartiles). Percent change in each Individual for (B) PHQ-9 and (D) EDA normalized to the maximum score of each questionnaire, plotted with the mean and standard deviation overlaid. Shaded symbols are subjects with co-morbid post-traumatic stress disorder (PTSD), open symbols are subjects without co-morbid PTSD. * = p <0.05 vs pre; paired two-tail t-test.
Fig 5. Pain interference and intensity.
Fig 5. Pain interference and intensity.
Pre- and post-morning bright light therapy questionnaire scores for the (A) NIH PROMIS Pain Interference (range = 4–20; higher = worse pain interference) and (C) NIH PROMIS Pain Intensity (range = 3–15; higher = pain intensity) displayed as truncated violin plots with moderate smoothing (dashed line = median score; dotted lines = 25% and 75% quartiles). Percent change in each Individual for (B) pain interference and (D) pain intensity, normalized to the maximum score of each questionnaire, plotted with the mean and standard deviation overlaid. Shaded symbols are subjects with co-morbid post-traumatic stress disorder (PTSD), open symbols are subjects without co-morbid PTSD. * = p <0.05 vs pre; paired two-tail t-test.
Fig 6. Quality of life.
Fig 6. Quality of life.
Pre- and post-morning bright light therapy questionnaire scores for the (A) World Health Organization Disability Assessment Schedule 2.0 (WHO-DAS 2.0; range = 0–100%; higher = worse quality of life/greater disability) is displayed as a truncated violin plot with moderate smoothing (dashed line = median score; dotted lines = 25% and 75% quartiles). Percent change in each Individual for (B) WHO-DAS 2.0 is normalized to the maximum score of this questionnaire, plotted with the mean and standard deviation overlaid. Shaded symbols are subjects with co-morbid post-traumatic stress disorder (PTSD), open symbols are subjects without co-morbid PTSD. * = p <0.05 vs pre; paired two-tail t-test.
Fig 7. Neuronal injury and neuroinflammation biomarkers.
Fig 7. Neuronal injury and neuroinflammation biomarkers.
Pre- and post-morning bright light therapy biomarkers for (A) Glial Fibrillary Acidic Protein (GFAP), (C) Neurofilament Light Chain (NfL), and (E) Tau, displayed as truncated violin plots with moderate smoothing (dashed line = median score; dotted lines = 25% and 75% quartiles). Percent change in each Individual for (B) GFAP, (D) NfL, and (F) Tau, plotted with the mean and standard deviation overlaid. Shaded symbols are subjects with co-morbid post-traumatic stress disorder (PTSD), open symbols are subjects without co-morbid PTSD. * = p <0.05 vs pre; paired two-tail t-test.
Fig 8. Peripheral cytokines.
Fig 8. Peripheral cytokines.
Pre- and post-morning bright light therapy biomarkers for (A) Interleukin-6 (IL-6), (C) Tumor Necrosis Factor Alpha (TNF-ɑ), and (E) Interleukin-10 (IL-10) displayed as truncated violin plots with moderate smoothing (dashed line = median score; dotted lines = 25% and 75% quartiles). Percent change in each Individual for (B) IL-6, (D) TNF-ɑ, and (F) IL-10, plotted with the mean and standard deviation overlaid. Shaded symbols are subjects with co-morbid post-traumatic stress disorder (PTSD), open symbols are subjects without co-morbid PTSD. * = p <0.05 vs pre; paired two-tail t-test.

References

    1. Hoge CW, McGurk D, Thomas JL, Cox AL, Engel CC, Castro CA. Mild Traumatic Brain Injury in U.S. Soldiers Returning from Iraq. N Engl J Med. 2008;358: 2543–2551. doi: 10.1056/NEJMoa072972
    1. Ouellet M-C, Savard J, Morin CM. Insomnia following Traumatic Brain Injury: A Review. Neurorehabil Neural Repair. 2004;18: 187–198. doi: 10.1177/1545968304271405
    1. Castriotta RJ, Wilde MC, Lai JM, Atanasov S, Masel BE, Kuna ST. Prevalence and consequences of sleep disorders in traumatic brain injury. J Clin Sleep Med. 2007;3: 349–356.
    1. Duclos C, Dumont M, Wiseman-Hakes C, Arbour C, Mongrain V, Gaudreault P-O, et al.. Sleep and wake disturbances following traumatic brain injury. Pathol Biol. 2014;62: 252–261. doi: 10.1016/j.patbio.2014.05.014
    1. Kempf J, Werth E, Kaiser PR, Bassetti CL, Baumann CR. Sleep-wake disturbances 3 years after traumatic brain injury. J Neurol Neurosurg Psychiatry. 2010;81: 1402–1405. doi: 10.1136/jnnp.2009.201913
    1. Balba NM, Elliott JE, Weymann KB, Opel RA, Duke JW, Oken BS, et al.. Increased sleep disturbances and pain in Veterans with co-morbid TBI and PTSD. J Clin Sleep Med. 2018;14: 1865–1878. doi: 10.5664/jcsm.7482
    1. Elliott JE, Opel RA, Weymann KB, Chau AQ, Papesh MA, Callahan ML, et al.. Sleep Disturbances in Traumatic Brain Injury: Associations With Sensory Sensitivity. J Clin Sleep Med. 2018;14: 1177–1186. doi: 10.5664/jcsm.7220
    1. Elliott JE, Opel RA, Pleshakov D, Rachakonda T, Chau AQ, Weymann KB, et al.. Posttraumatic stress disorder increases the odds of REM sleep behavior disorder and other parasomnias in Veterans with and without comorbid traumatic brain injury. Sleep. 2020;43: zsz237. doi: 10.1093/sleep/zsz237
    1. Sandsmark DK, Elliott JE, Lim MM. Sleep-Wake Disturbances After Traumatic Brain Injury: Synthesis of Human and Animal Studies. Sleep. 2017;40: zsx044. doi: 10.1093/sleep/zsx044
    1. Cantu D, Walker K, Andresen L, Taylor-Weiner A, Hampton D, Tesco G, et al.. Traumatic Brain Injury Increases Cortical Glutamate Network Activity by Compromising GABAergic Control. Cereb Cortex. 2015;25: 2306–2320. doi: 10.1093/cercor/bhu041
    1. Elliott JE, De Luche SE, Churchill MJ, Moore C, Cohen AS, Meshul CK, et al.. Dietary therapy restores glutamatergic input to orexin/hypocretin neurons after traumatic brain injury in mice. Sleep. 2018;41: zsx212. doi: 10.1093/sleep/zsx212
    1. Baumann CR, Bassetti CL, Valko PO, Haybaeck J, Keller M, Clark E, et al.. Loss of hypocretin (orexin) neurons with traumatic brain injury. Ann Neurol. 2009;66: 555–559. doi: 10.1002/ana.21836
    1. Shekleton JA, Parcell DL, Redman JR, Phipps-Nelson J, Ponsford JL, Rajaratnam SMW. Sleep disturbance and melatonin levels following traumatic brain injury. Neurology. 2010;74: 1732–1738. doi: 10.1212/WNL.0b013e3181e0438b
    1. Dinges DF. Sleep in space flight breath easy—Sleep less? Am J Respir Crit Care Med. 2001;164: 337–338. doi: 10.1164/ajrccm.164.3.2105072a
    1. Banks S, Dinges DF. Behavioral and physiological consequences of sleep restriction. J Clin Sleep Med. 2007;3: 519–528.
    1. Dinges DF, Douglas SD, Hamarman S, Zaugg L, Kapoor S. Sleep deprivation and human immune function. Adv Neuroimmunol. 1995;5: 97–110. doi: 10.1016/0960-5428(95)00002-j
    1. Durmer JS, Dinges DF. Neurocognitive Consequences of Sleep Deprivation. Semin Neurol. 2005;25: 117–129. doi: 10.1055/s-2005-867080
    1. Turk DC, Dworkin RH, Revicki D, Harding G, Burke LB, Cella D, et al.. Identifying important outcome domains for chronic pain clinical trials: An IMMPACT survey of people with pain. Pain. 2008;137: 276–285. doi: 10.1016/j.pain.2007.09.002
    1. Cappuccio FP, D’Elia L, Strazzullo P, Miller MA. Sleep duration and all-cause mortality: A systematic review and meta-analysis of prospective studies. Sleep. 2010;33: 585–592. doi: 10.1093/sleep/33.5.585
    1. Watson NF, Badr MS, Belenky G, Bliwise DL, Buxton OM, Buysse D, et al.. Joint Consensus Statement of the American Academy of Sleep Medicine and Sleep Research Society on the Recommended Amount of Sleep for a Healthy Adult: Methodology and Discussion. Sleep. 2015;38: 1161–1183. doi: 10.5665/sleep.4886
    1. Rao V, Spiro J, Vaishnavi S, Rastogi P, Mielke M, Noll K, et al.. Prevalence and types of sleep disturbances acutely after traumatic brain injury. Brain Inj. 2008;22: 381–386. doi: 10.1080/02699050801935260
    1. Makley MJ, Gerber D, Newman JK, Philippus A, Monden KR, Biggs J, et al.. Optimized Sleep After Brain Injury (OSABI): A Pilot Study of a Sleep Hygiene Intervention for Individuals With Moderate to Severe Traumatic Brain Injury. Neurorehabil Neural Repair. 2020;34: 111–121. doi: 10.1177/1545968319895478
    1. Makley MJ, Johnson-Greene L, Tarwater PM, Kreuz AJ, Spiro J, Rao V, et al.. Return of Memory and Sleep Efficiency Following Moderate to Severe Closed Head Injury. Neurorehabil Neural Repair. 2008;23: 320–326.
    1. Zollman FS, Larson EB, Wasek-Throm LK, Cyborski CM, Bode RK. Acupuncture for Treatment of Insomnia in Patients With Traumatic Brain Injury: A Pilot Intervention Study. J Head Trauma Rehabil. 2012;27: 135–142. doi: 10.1097/HTR.0b013e3182051397
    1. Viola-Saltzman M, Musleh C. Traumatic brain injury-induced sleep disorders. Neuropsychiatr Dis Treat. 2016;12: 339–348. doi: 10.2147/NDT.S69105
    1. Viola-Saltzman M, Watson NF. Traumatic Brain Injury and Sleep Disorders. Neurol Clin. 2012;30: 1299–1312. doi: 10.1016/j.ncl.2012.08.008
    1. Ayalon L, Borodkin K, Dishon L, Kanety H, Dagan Y. Circadian rhythm sleep disorders following mild traumatic brain injury. Neurology. 2007;68: 1136–1140. doi: 10.1212/01.wnl.0000258672.52836.30
    1. Grima NA, Ponsford JL, St Hilaire MA, Mansfield D, Rajaratnam SMW. Circadian Melatonin Rhythm Following Traumatic Brain Injury. Neurorehabil Neural Repair. 2016;30: 972–977. doi: 10.1177/1545968316650279
    1. LeGates TA, Altimus CM, Wang H, Lee H-K, Yang S, Zhao H, et al.. Aberrant light directly impairs mood and learning through melanopsin-expressing neurons. Nature. 2012;491: 594–598. doi: 10.1038/nature11673
    1. Lam RW, Levitt AJ, Levitan RD, Michalak EE, Cheung AH, Morehouse R, et al.. Efficacy of bright light treatment, fluoxetine, and the combination in patients with nonseasonal major depressive disorder a randomized clinical trial. JAMA Psychiatry. 2016;73: 56–63. doi: 10.1001/jamapsychiatry.2015.2235
    1. LeGates TA, Fernandez DC, Hattar S. Light as a central modulator of circadian rhythms, sleep and affect. Nat Rev Neurosci. 2014;15: 443–54. doi: 10.1038/nrn3743
    1. Raikes AC, Killgore WDS. Potential for the development of light therapies in mild traumatic brain injury. Concussion. 2018;3. doi: 10.2217/cnc-2018-0006
    1. Terman M, Terman JS. Light therapy for seasonal and nonseasonal depression: efficacy, protocol, safety, and side effects. CNS Spectr. 2005;10: 647–63. doi: 10.1017/s1092852900019611
    1. Riemersma-van der Lek RF, Swaab DF, Twisk J, Hol EM, Hoogendijk WJ, van Someren EJW. Effect of Bright Light and Melatonin on Cognitive and Noncognitive Function in Elderly Residents of Group Care Facilities. JAMA. 2008;299: 2642–2655. doi: 10.1001/jama.299.22.2642
    1. Lockley SW, Evans EE, Scheer FAJL, Brainard GC, Czeisler CA, Aeschbach D. Short-wavelength sensitivity for the direct effects of light on alertness, vigilance, and the waking electroencephalogram in humans. Sleep. 2006;29: 161–168.
    1. Ancoli-israel S, Gehrman P, Martin JL, Shochat T, Marler M, Corey-bloom J, et al.. Increased Light Exposure Consolidates Sleep and Strengthens Circadian Rhythms in Severe Alzheimer’s Disease Patients Increased Light Exposure Consolidates Sleep and Strengthens Circadian Rhythms in Severe Alzheimer’s Disease Patients. Behav Sleep Med. 2003;1: 22–36. doi: 10.1207/S15402010BSM0101_4
    1. Chellappa S, Gordijn MCM, Cajochen C. Can light make us bright? Effects of light on cognition sleep. Prog Brain Res. 2011;190: 119–133. doi: 10.1016/B978-0-444-53817-8.00007-4
    1. Killgore WDS, Vanuk JR, Shane BR, Weber M, Bajaj S. A randomized, double-blind, placebo-controlled trial of blue wavelength light exposure on sleep and recovery of brain structure, function, and cognition following mild traumatic brain injury. Neurobiol Dis. 2020;134: 104679. doi: 10.1016/j.nbd.2019.104679
    1. Raikes AC, Dailey NS, Shane BR, Forbeck B, Alkozei A, Killgore WDS. Daily Morning Blue Light Therapy Improves Daytime Sleepiness, Sleep Quality, and Quality of Life Following a Mild Traumatic Brain Injury. J Head Trauma Rehabil. 2020.
    1. Sinclair KL, Ponsford JL, Taffe J, Lockley SW, Rajaratnam SMW. Randomized Controlled Trial of Light Therapy for Fatigue Following Traumatic Brain Injury. Neurorehabil Neural Repair. 2013;28: 303–313. doi: 10.1177/1545968313508472
    1. Quera Salva M-A, Azabou E, Hartley S, Sauvagnac R, Leotard A, Vaugier I, et al.. Blue-Enriched White Light Therapy Reduces Fatigue in Survivors of Severe Traumatic Brain Injury: A Randomized Controlled Trial. J Head Trauma Rehabil. 2020;35: E78–E85. doi: 10.1097/HTR.0000000000000500
    1. Youngstedt SD, Kline CE, Reynolds AM, Crowley SK, Burch JB, Khan N, et al.. Bright Light Treatment of Combat-related PTSD: A Randomized Controlled Trial. Mil Med. 2021;00: 1–10.
    1. McInnes K, Friesen CL, MacKenzie DE, Westwood DA, Boe SG. Mild Traumatic Brain Injury (mTBI) and chronic cognitive impairment: A scoping review. PLoS One. 2017;12.
    1. Chesson AL, Littner M, Davila D, Anderson WM, Grigg-damberger M, Hartse K, et al.. Practice Parameters for the Use of Light Therapy in the Treatment of Sleep Disorders. Sleep. 1999;22: 641–660. doi: 10.1093/sleep/22.5.641
    1. Penders TM, Stanciu CN, Schoemann AM, Ninan PT, Bloch R, Saeed SA. Bright Light Therapy as Augmentation of Pharmacotherapy for Treatment of Depression. Prim Care Companion CNS Disord. 2016. doi: 10.4088/PCC.15r01906
    1. Anderson JL, Glod CA, Dai J, Cao Y, Lockley SW. Lux vs. wavelength in light treatment of seasonal affective disorder. Acta Psychiatr Scand. 2009;120: 203–212. doi: 10.1111/j.1600-0447.2009.01345.x
    1. Cifu D, Hurley R, Peterson M, Cornis-Pop M, Rikli PA, Ruff RL, et al.. VA/DoD Clinical practice guideline: Management of Concussion/Mild Traumatic Brain Injury. J Rehabil Res Dev. 2009;46: CP1–68.
    1. Thabane L, Hopewell S, Lancaster GA, Bond CM, Coleman CL, Campbell MJ, et al.. Methods and processes for development of a CONSORT extension for reporting pilot randomized controlled trials. Pilot feasibility Stud. 2016;2: 25. Available: .
    1. Eldridge SM, Chan CL, Campbell MJ, Bond CM, Hopewell S, Thabane L, et al.. CONSORT 2010 statement: extension to randomised pilot and feasibility trials. BMJ. 2016;355: i5239. Available: .
    1. Eldridge SM, Lancaster GA, Campbell MJ, Thabane L, Hopewell S, Coleman CL, et al.. Defining feasibility and pilot studies in preparation for randomised controlled trials: Development of a conceptual framework. PLoS One. 2016;11: 1–22. doi: 10.1371/journal.pone.0150205
    1. Spitschan M, Stefani O, Blattner P, Gronfier C, Lockley S, Lucas R. How to Report Light Exposure in Human Chronobiology and Sleep Research Experiments. Clocks & Sleep. 2019;1: 280–289. doi: 10.3390/clockssleep1030024
    1. Jean-Louis G, Kripke DF, Mason WJ, Elliott JA, Youngstedt SD. Sleep estimation from wrist movement quantified by different actigraphic modalities. J Neurosci Methods. 2001;105: 185–191. doi: 10.1016/s0165-0270(00)00364-2
    1. Morin CM. Insomnia. Psychological assessment and management. New York: Guilford Press; 1993.
    1. Bastien CH, Vallières A, Morin CM. Validation of the insomnia severity index as an outcome measure for insomnia research. Sleep Med. 2001;2: 297–307. doi: 10.1016/s1389-9457(00)00065-4
    1. Mastin DF, Bryson J, Corwyn R. Assessment of sleep hygiene using the sleep hygiene index. J Behav Med. 2006;29: 223–227. doi: 10.1007/s10865-006-9047-6
    1. Belanger HG, Kretzmer T, Vanderploeg RD, French LM. Symptom complaints following combat-related traumatic brain injury: Relationship to traumatic brain injury severity and posttraumatic stress disorder. J Int Neuropsychol Soc. 2009;16: 194–199. doi: 10.1017/S1355617709990841
    1. Bulson R, Jun W, Hayes J. Visual symptomatology and referral patterns for Operation Iraqi Freedom and Operation Enduring Freedom veterans with traumatic brain injury. J Rehabil Res Dev. 2012;49: 1075–1082. doi: 10.1682/jrrd.2011.02.0017
    1. Vanderploeg RD, Cooper DB, Belanger HG, Donnell AJ, Kennedy JE, Hopewell CA, et al.. Screening for Postdeployment Conditions: Development and Cross-Validation of an Embedded Validity Scale in the Neurobehavioral Symptom Inventory. J Head Trauma Rehabil. 2014;29: 1–10. doi: 10.1097/HTR.0b013e318281966e
    1. Blevins CA, Weather FW, Davis MT, Witte TK, Domino JL. The Posttraumatic Stress Disorder Checklist for DSM-5 (PCL-5): Development and Initial Psychometric Evaluation. J Trauma Stress. 2015;28: 489–498. doi: 10.1002/jts.22059
    1. Edwards KA, Gill JM, Pattinson CL, Lai C, Brière M, Rogers NJ, et al.. Interleukin-6 is associated with acute concussion in military combat personnel. BMC Neurol. 2020;20: 209. doi: 10.1186/s12883-020-01760-x
    1. Guedes VA, Kenney K, Shahim P, Qu B, Lai C, Devoto C, et al.. Exosomal neurofilament light A prognostic biomarker for remote symptoms after mild traumatic brain injury? Neurology. 2020.
    1. Mondello S, Buki A, Barzo P, Randall J, Provuncher G, Hanlon D, et al.. CSF and Plasma Amyloid-β Temporal Profiles and Relationships with Neurological Status and Mortality after Severe Traumatic Brain Injury. Sci Rep. 2014;3: 2–7. doi: 10.1038/srep06446
    1. Korley FK, Yue JK, Wilson DH, Hrusovsky K, Diaz-Arrastia R, Ferguson AR, et al.. Performance Evaluation of a Multiplex Assay for Simultaneous Detection of Four Clinically Relevant Traumatic Brain Injury Biomarkers. J Neurotrauma. 2019;36: 182–187.
    1. van Maanen A, Meijer AM, van der Heijden KB, Oort FJ. The effects of light therapy on sleep problems: A systematic review and meta-analysis. Sleep Med Rev. 2016;29: 52–62. doi: 10.1016/j.smrv.2015.08.009
    1. Lewy AJ, Wehr TA, Goodwin FK, Newsome DA, Markey SP. Light Suppresses Melatonin Secretion in Humans. Science. 1980;210: 1267–1269. doi: 10.1126/science.7434030
    1. Czeisler CA, Shanahan TL, Klerman EB, Martens H, Brotman DJ, Emens JS, et al.. Suppression of Melatonin Secretion in Some Blind Patients by Exposure to Bright Light. N Engl J Med. 1995;332: 6–11. doi: 10.1056/NEJM199501053320102
    1. Kripke DF, Elliott JA, Youngstedt SD, Rex KM. Circadian phase response curves to light in older and young women and men. J Circadian Rhythms. 2007;5: 1–13.
    1. Hattar S, Liao HW, Takao M, Berson DM, Yau KW. Melanopsin-containing retinal ganglion cells: architecture, projections, and intrinsic photosensitivity. Science. 2002;295: 1065–1070. doi: 10.1126/science.1069609
    1. Schmidt TM, Chen SK, Hattar S. Intrinsically photosensitive retinal ganglion cells: Many subtypes, diverse functions. Trends Neurosci. 2011;34: 572–580. doi: 10.1016/j.tins.2011.07.001
    1. Van Someren EJ, Kessler A, Mirmiran M, Swaab DF. Indirect bright light improves circadian rest-activity rhythm disturbances in demented patients. Biol Psychiatry. 1997;41: 955–963. doi: 10.1016/S0006-3223(97)89928-3
    1. Lockley SW, Gooley JJ. Circadian Photoreception: Spotlight on the Brain. Curr Biol. 2006;16: 795–797.
    1. Borbely AA. A two process model of sleep regulation. Hum Neurobiol. 1982;1: 195–204.
    1. Jindal RD, Thase ME. Treatment of insomnia associated with clinical depression. Sleep Med Rev. 2004;8: 19–30. doi: 10.1016/S1087-0792(03)00025-X
    1. Kantermann T, Burgess HJ. Average mid-sleep time as a proxy for circadian phase. PsyCh J. 2017;6: 290–291. doi: 10.1002/pchj.182
    1. Crowley SJ, Wolfson AR, Tarokh L, Carskadon MA. An update on adolescent sleep: New evidence informing the perfect storm model. J Adolesc. 2018;67: 55–65. doi: 10.1016/j.adolescence.2018.06.001
    1. Mattsson N, Cullen NC, Andreasson U, Zetterberg H, Blennow K. Association between Longitudinal Plasma Neurofilament Light and Neurodegeneration in Patients with Alzheimer Disease. JAMA Neurol. 2019;76: 791–799. doi: 10.1001/jamaneurol.2019.0765
    1. Pattinson CL, Shahim P, Taylor P, Dunbar K, Guedes VA, Motamedi V, et al.. Elevated tau in military personnel relates to chronic symptoms following traumatic brain injury. J Head Trauma Rehabil. 2020;35: 66–73. doi: 10.1097/HTR.0000000000000485
    1. Jin F, Liu J, Zhang X, Cai W, Zhang Y, Zhang W, et al.. Effect of continuous positive airway pressure therapy on inflammatory cytokines and atherosclerosis in patients with obstructive sleep apnea syndrome. Mol Med Rep. 2017;16: 6334–6339. doi: 10.3892/mmr.2017.7399
    1. Van Der Ploeg ES, O’Connor DW. Methodological challenges in studies of bright light therapy to treat sleep disorders in nursing home residents with dementia. Psychiatry Clin Neurosci. 2014;68: 777–784. doi: 10.1111/pcn.12192
    1. Ancoli-Israel S, Cole R, Alessi C, Chambers M, Moorcroft W, Pollak CP. The role of actigraphy in the study of sleep and circadian rhythms. Sleep. 2003;26: 342–392. doi: 10.1093/sleep/26.3.342

Source: PubMed

3
Subscribe