Understanding Photobiomodulation: Laser Therapy and Its Benefits Explained

Oct 17, 2024 | Written by Solène Grosdidier, PharmD, PhD | Reviewed by Scott Sherr, MD and Marion Hall

Understanding Photobiomodulation: Laser Therapy and Its Benefits Explained

Red light therapy, also known as photobiomodulation (PBM) therapy, low-level laser therapy, or cold laser therapy, is a phototherapy technique that uses red (wavelengths between 600-700 nm) and near-infrared (wavelengths between 700-1440 nm) lights [1] from lasers or light-emitting diodes (LEDs). While lasers are commonly used in animal studies, LEDs are preferred in clinical trials due to their low price and easy assembly [2]. As the wavelength increases, the light penetrates deeper into tissues. Pulsed wave light also penetrates deeper into tissues than continuous wave light at the same wavelength in ex vivo experiments [3-6]. The pulsed wave frequency can be adjusted to trigger specific beneficial biological effects [7].

PBM was first described by Endre Mester in 1968 when he observed an unexpected acceleration of hair regeneration in mice exposed to low-energy lasers with a wavelength of 694 nm [8]. Since then, PBM has demonstrated beneficial effects in managing non-healing wounds, ulcers, scars, musculoskeletal disorders, persistent pain, and immunological regulation [9]. The scientific literature on PBM has grown significantly in recent years [10].
 
Read on to learn more about how PBM therapy works and its many benefits, but if you're already familiar, then check out this article on the synergistic effects of methylene blue combined with red light!

How does photobiomodulation or low-level laser therapy work?

The interaction of red light (RL) or near-infrared light (NIR) at specific wavelengths with tissues activates endogenous chromophores (groups of atoms that interact with light to create color). PBM triggers biological effects in a non-thermal and non-cytotoxic manner [11,12]. One of its most recognized mechanisms of action involves the mitochondria [13]. RL and NIR photons are absorbed at the cellular level by mitochondria, which contain sensitive photo-acceptors to the length used with PBM, like the cytochrome C oxidase enzyme. When tissues are stressed, mitochondria produce nitric oxide, which competes with oxygen at the cytochrome C oxidase site in the respiratory chain. This process reduces adenosine triphosphate (ATP) synthesis, increases oxidative stress, and triggers inflammation [14]. However, when exposed to RL or NIR photons, cytochrome C oxidase absorbs their energy, dissociating nitric oxide. This release enhances ATP production, reduces oxidative stress [15], and influences various cellular components, including the cytosol, membrane, and nucleus. Through these mechanisms, RL and NIR indirectly impact gene transcription, cell proliferation, migration, viability, and inflammation [16]. Systemic effects can also be observed via light activation of blood and lymph cells [17,18].

Therapeutic applications

The biological effects identified in preclinical and clinical studies include anti-inflammatory and analgesic actions, enhanced blood circulation, promotion of angiogenesis, and stimulation of tissue healing, regeneration, and proliferation [19-21]. This wide range of effects makes PBM suitable for various therapeutic applications, including pain management, neurological disorders, and radiation dermatitis.

PBM and pain management

Experiments in animals showed that, at optimal doses, PBM is as effective as non-steroidal anti-inflammatory drugs to treat pain [22].

Via its effects on the mitochondrial cytochrome C oxidase enzyme, PBM has been shown to increase cell proliferation and migration by fibroblasts, reduce cytokines, growth factors, and inflammatory mediators, while increasing tissue oxygenation, reducing pain, and improving wound healing in humans [23-25]. Clinical trials have confirmed that PBM also enhances wound healing in diabetic patients [26] and reduces the incidence of post-herpetic neuralgia when applied early to herpes zoster eruptions [27].
 
PBM has shown effectiveness in alleviating acute pain in various conditions, such as oral surgery [28,29], nipple pain from breastfeeding [30], plantar fasciitis [31], and carpal tunnel syndrome [32], with positive results in reducing pain intensity in women with chronic myofascial pain [33]. It has also alleviated pain after coronary artery bypass surgery [34] and improved sensory function in neuropathy [35]. PBM is commonly used to manage dental pain after oral surgery, reducing orthodontic pain for up to a week [36]. However, some studies have shown inconsistent results, such as those for plantar fasciitis [37] or knee osteoarthritis [38].
 
PBM offers a promising non-invasive option for pain management across a range of conditions.

PBM and neurological diseases

Alzheimer’s disease

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by progressive memory loss and cognitive decline, primarily associated with the accumulation of amyloid-β (Aβ) peptide and tau protein tangles in the brain. Mitochondrial dysfunction is related to the main pathological feature of AD: neuronal dysfunction [39].
 
In a rat model of AD, transcranial PBM at 808 nm was found to have suppressed Aβ aggregation, improving long-term spatial and recognition memory [40]. PBM at 808 nm in a transgenic mouse model of Aβ peptide amyloidosis showed an increase in soluble Aβ peptide, mitochondrial function, and ATP with a decrease in inflammatory markers, suggesting an overall improvement in neurological function [41]. Similarly, transcranial PBM at 633 nm reduced Aβ production and plaque formation by shifting amyloid precursor protein (APP) processing toward the nonamyloidogenic pathway in a mouse model of AD, resulting in memory and cognitive improvements [42]. At 633 nm, PBM also inhibited the activity of c-Jun N-terminal kinase 3 (a signal molecule related to neurodegeneration), a possible target of AD treatment [43]. Recent evidence suggests an involvement of the gut-brain axis in AD: gut microbiota produce peripheral inflammation and immune modulation that may contribute to brain amyloidosis, neurodegeneration, and cognitive impairment in AD. Simultaneous pulsed wave PBM to the head and abdomen for 10 min triggered neuroprotective effects via normalization of all modified behavioral and biochemical parameters in a β25-35 peptide-induced toxicity experiment in mice [44].
 
In clinical trials, transcranial PBM treatment for 28 days at a wavelength between 1060 and 1080 nm improved cognition and memory in AD patients [45]. Other PBM treatments in patients with progressive neurodegenerative diseases improved cognition, mood, and sleep [45-47].
 
These findings suggest that PBM presents potential in mitigating AD, shedding light on possible new therapies.

Parkinson's disease

Parkinson’s disease (PD) is a progressive neurodegenerative disease characterized by the loss of dopaminergic neurons in the substantia nigra and the presence of Lewy bodies in the brain. The exact cause of PD remains unknown, but evidence suggests that mitochondrial dysfunction and oxidative stress contribute to its pathogenesis.
 
In animal models of PD, transcranial PBM treatment at 670 and 810 nm alleviated PD pathology in rats, mice, and monkeys [48-52]. PBM has also shown neuroprotective effects and prevented cerebrovascular leakage in the substantia nigra pars compacta region [53]. Additionally, in a rat model of PD, PBM significantly reduced serum levels of pro-inflammatory cytokines [49].
 
In clinical trials, transcranial PBM at 635 and 810 nm safely improved motor symptoms in PD patients [54], and combinations of other wavelengths (e.g., 670, 810, and 850 nm) also showed benefits for both motor and non-motor symptoms [55]. Due to the exponential attenuation of light through the skull and tissue, various strategies — such as applying PBM to the neck, intranasal passages, and the abdomen combined with transcranial PBM — have been developed and demonstrated effectiveness [56-58].
 
Overall, current research suggests the great potential of PBM in PD treatment.

Traumatic brain injury

A traumatic brain injury (TBI) is a trauma caused to the brain by an external force and can result in either complete recovery, permanent disability, or death. The pathophysiology of TBI is highly complex and involves inflammation, oxidative stress, mitochondrial dysfunction, and excitotoxic damage.
 
Experiments in mice showed that transcranial PBM mitigates the death of brain neurons by decreasing neuroinflammation, increasing blood flow [59], and improving brain self-repair abilities via stimulation of synapse formation and proliferation of nerve cells [60,61]. Laser treatment 4 h after TBI significantly improved the neurological severity score (a scale evaluating motor, sensory, and reflex functions) in moderate-to-severe TBI mouse models [62,63]. Combined with lactic acid or pyruvate, two energy metabolism regulators, PBM increased ATP levels more effectively and reduced neuroinflammation, preventing neuronal damage in the injured zone of the brain [64,65]. However, too many PBM treatments in TBI mouse models can temporarily inhibit brain repair, highlighting the importance of developing optimal protocols [66].
 
In clinical trials, PBM treatment significantly enhanced cognitive performance in mild TBI patients at 633 and 870 nm [67,68], while improving sleep duration at 633 and 810 nm [69] in chronic TBI patients commonly suffering from sleep disturbances. Another study combining intranasal and transcranial PBM treatments at 810 nm for eight weeks showed an increased brain volume and improvements in cerebral perfusion, functional connectivity, and neuropsychological test scores in a 23-year-old TBI patient [70].
 
Transcranial PBM demonstrates high potential for the clinical treatment of TBI.

Depression

Depression is a disease characterized by a lack of energy, depressed mood, and poor concentration. Typically treated with medication, these drugs often have low efficacy and can cause significant side effects. The pathophysiology of depression involves a complex interplay between inflammation, oxidative stress, DNA damage, impaired DNA repair, and mitochondrial dysfunction [71].
 
Experiments in rat models showed that PBM at 810 nm promoted neuroprotection effects from stress [72], underwater trauma [73], reserpine [74], and early AD-associated depression [75]. PBM treatment at 808 nm was as effective as citalopram in improving depressive-like behaviors in a rat model of chronic mild stress-induced depression [76].
 
In clinical trials, transcranial PBM at 810 nm significantly improved the Hamilton depression rating scale and anxiety rating scale scores in depressive patients [77], while demonstrating safety and effectiveness in major depressive disorder patients at 823 nm [78].
 
Although further research is needed, PBM could be a promising approach for the treatment of depression.

Aging

Aging is characterized by a gradual functional decline at the molecular, cellular, and tissue levels, linked to oxidative stress and mitochondrial dysfunction [79], contributing to cognitive impairment [80].
 
Comparing aged rats to young rats revealed that aging significantly decreases brain cytochrome C oxidase activity, an effect that PBM treatment at 810 nm reversed [81]. PBM (at 660 or 810 nm) also mitigated inflammatory responses and altered intracellular signaling pathways linked to glucose metabolism, vascular function, and cell survival in the aging brain of rats. PBM treatment (at 660 and 810 nm) also significantly alleviated cognitive and memory decline, while improving mitochondrial function and reducing oxidative stress and cell death in a D-galactose-induced aging mouse model [82,83]. In addition, treatment at 660 nm alleviated cognitive impairment and mitochondrial dysfunction in 18-month-old aging mice [84], while PBM at 670 nm showed beneficial effects on astrocytes and microglia proliferation [85].
 
In clinical trials, PBM (at 633 and 870 nm) improved depressive symptoms and cognitive function in both healthy participants [86], and those with mild cognitive impairment [87], as assessed by the Eriksen flanker category fluency tests and visual memory span test, respectively. PBM treatment at 1064 nm in elderly participants also improved attention and memory as assessed by psychomotor vigilance tasks and delayed match to sample. These improvements came together with an increased prefrontal blood-oxygen-level-dependent-fMRI activity and elevated levels of cytochrome C oxidase enzymes during the light exposure periods [88]. Beneficial effects on working memory were documented and lasted at least three weeks [89].
 
Overall, PBM is a promising candidate for age-related cognitive improvement.

PBM and radiation dermatitis

One of the best-studied applications of PBM is the treatment of radiation dermatitis, a skin condition secondary to oncology therapies. Multiple controlled trials showed that PBM efficiently reduces radiation dermatitis severity, worsening, and pain [90-92]. However, other studies following breast cancer radiotherapy provide contradictory results [92,93]. Overall, two recent meta-analyses, including five and seven clinical trials, concluded that PBM presents promising properties to prevent radiation dermatitis severity [94,95].

Conclusion

PBM is a non-invasive therapy that uses red or near-infrared light to penetrate human tissues. By targeting the mitochondrial enzyme cytochrome C oxidase, PBM enhances ATP production and reduces oxidative stress, indirectly influencing gene transcription, cell proliferation, migration, viability, and inflammation. Clinical trials have demonstrated its potential benefits in various applications, including pain management, Alzheimer's disease, Parkinson's disease, traumatic brain injury, and radiation dermatitis, among others. PBM is a cost-effective therapy [96] with a favorable safety profile [97], erythema being the most common and self-limiting adverse effect [1]. It can also be used alongside pharmacological treatments.

References

[1] Maghfour, J., Ozog, D.M., Mineroff, J., Jagdeo, J., Kohli, I. and Lim, H.W. (2024) Photobiomodulation CME Part I: Overview and Mechanism of Action. Journal of the American Academy of Dermatology, S0190962224001865. https://doi.org/10.1016/j.jaad.2023.10.073
[2] Lin, H., Li, D., Zhu, J., Liu, S., Li, J., Yu, T. et al. (2024) Transcranial photobiomodulation for brain diseases: review of animal and human studies including mechanisms and emerging trends. Neurophotonics, 11. https://doi.org/10.1117/1.NPh.11.1.010601
[3] Hashmi, J.T., Huang, Y., Sharma, S.K., Kurup, D.B., De Taboada, L., Carroll, J.D. et al. (2010) Effect of pulsing in low‐level light therapy. Lasers in Surgery and Medicine, 42, 450–66. https://doi.org/10.1002/lsm.20950
[4] Brondon, P., Stadler, I. and Lanzafame, R.J. (2009) Pulsing influences photoradiation outcomes in cell culture. Lasers in Surgery and Medicine, 41, 222–6. https://doi.org/10.1002/lsm.20740
[5] Shariati B K, B., Khatami, S.S., Ansari, M.A., Jahangiri, F., Latifi, H. and Tuchin, V.V. (2022) Method for tissue clearing: temporal tissue optical clearing. Biomedical Optics Express, 13, 4222. https://doi.org/10.1364/BOE.461115
[6] Galiakhmetova, D., Dremin, V., Koviarov, A., Stoliarov, D., Ngum, N., Murugesan, R.C. et al. (2023) Ultra-Short Laser Pulses Propagation Through Mouse Head Tissues: Experimental and Computational Study. IEEE Journal of Selected Topics in Quantum Electronics, 29, 1–11. https://doi.org/10.1109/JSTQE.2022.3214788
[7] Tao, L., Liu, Q., Zhang, F., Fu, Y., Zhu, X., Weng, X. et al. (2021) Microglia modulation with 1070-nm light attenuates Aβ burden and cognitive impairment in Alzheimer’s disease mouse model. Light: Science & Applications, 10, 179. https://doi.org/10.1038/s41377-021-00617-3
[8] Mester, E., Szende, B. and Gärtner, P. (1968) [The effect of laser beams on the growth of hair in mice]. Radiobiologia, Radiotherapia, 9, 621–6.
[9] Hernández-Bule, M.L., Naharro-Rodríguez, J., Bacci, S. and Fernández-Guarino, M. (2024) Unlocking the Power of Light on the Skin: A Comprehensive Review on Photobiomodulation. International Journal of Molecular Sciences, 25, 4483. https://doi.org/10.3390/ijms25084483
[10] Perrier, Q., Moro, C. and Lablanche, S. (2024) Diabetes in spotlight: current knowledge and perspectives of photobiomodulation utilization. Frontiers in Endocrinology, 15, 1303638. https://doi.org/10.3389/fendo.2024.1303638
[11] Kuffler, D.P. (2016) Photobiomodulation in Promoting Wound Healing: A Review. Regenerative Medicine, 11, 107–22. https://doi.org/10.2217/rme.15.82
[12] Shaikh-Kader, A. and Houreld, N.N. (2022) Photobiomodulation, Cells of Connective Tissue and Repair Processes: A Look at In Vivo and In Vitro Studies on Bone, Cartilage and Tendon Cells. Photonics, 9, 618. https://doi.org/10.3390/photonics9090618
[13] Pastore, M. Greco, S. Passarella, D. (2000) Specific helium-neon laser sensitivity of the purified cytochrome c oxidase. International Journal of Radiation Biology, 76, 863–70. https://doi.org/10.1080/09553000050029020
[14] Palacios-Callender, M., Quintero, M., Hollis, V.S., Springett, R.J. and Moncada, S. (2004) Endogenous NO regulates superoxide production at low oxygen concentrations by modifying the redox state of cytochrome c oxidase. Proceedings of the National Academy of Sciences, 101, 7630–5. https://doi.org/10.1073/pnas.0401723101
[15] Sharma, S.K., Kharkwal, G.B., Sajo, M., Huang, Y., De Taboada, L., McCarthy, T. et al. (2011) Dose response effects of 810 nm laser light on mouse primary cortical neurons. Lasers in Surgery and Medicine, 43, 851–9. https://doi.org/10.1002/lsm.21100
[16] Chung, H., Dai, T., Sharma, S.K., Huang, Y.-Y., Carroll, J.D. and Hamblin, M.R. (2012) The Nuts and Bolts of Low-level Laser (Light) Therapy. Annals of Biomedical Engineering, 40, 516–33. https://doi.org/10.1007/s10439-011-0454-7
[17] Stergioulas, A. (2004) Low-Level Laser Treatment Can Reduce Edema in Second Degree Ankle Sprains. Journal of Clinical Laser Medicine & Surgery, 22, 125–8. https://doi.org/10.1089/104454704774076181
[18] Omar, M.T.A., Shaheen, A.A.M. and Zafar, H. (2012) A systematic review of the effect of low-level laser therapy in the management of breast cancer-related lymphedema. Supportive Care in Cancer, 20, 2977–84. https://doi.org/10.1007/s00520-012-1546-0
[19] R Hamblin, M., 1 Wellman Center for Photomedicine, Massachusetts General Hospital, BAR414, 40 Blossom Street, Boston, MA 02114, USA, 2 Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA and 3 Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA. (2017) Mechanisms and applications of the anti-inflammatory effects of photobiomodulation. AIMS Biophysics, 4, 337–61. https://doi.org/10.3934/biophy.2017.3.337
[20] Dompe, C., Moncrieff, L., Matys, J., Grzech-Leśniak, K., Kocherova, I., Bryja, A. et al. (2020) Photobiomodulation—Underlying Mechanism and Clinical Applications. Journal of Clinical Medicine, 9, 1724. https://doi.org/10.3390/jcm9061724
[21] Zein, R., Selting, W. and Hamblin, M.R. (2018) Review of light parameters and photobiomodulation efficacy: dive into complexity. Journal of Biomedical Optics, 23, 1. https://doi.org/10.1117/1.JBO.23.12.120901
[22] Bjordal, J.M., Johnson, M.I., Iversen, V., Aimbire, F. and Lopes-Martins, R.A.B. (2006) Low-Level Laser Therapy in Acute Pain: A Systematic Review of Possible Mechanisms of Action and Clinical Effects in Randomized Placebo-Controlled Trials. Photomedicine and Laser Surgery, 24, 158–68. https://doi.org/10.1089/pho.2006.24.158
[23] Ojea, A.R., Madi, O., Neto, R.M.L., Lima, S.E., De Carvalho, B.T., Ojea, M.J.M.R. et al. (2016) Beneficial Effects of Applying Low-Level Laser Therapy to Surgical Wounds After Bariatric Surgery. Photomedicine and Laser Surgery, 34, 580–4. https://doi.org/10.1089/pho.2016.4149
[24] Cullum, N., Nelson, E.A., Flemming, K. and Sheldon, T. (2001) Systematic reviews of wound care management: (5) beds; (6) compression; (7) laser therapy, therapeutic ultrasound, electrotherapy and electromagnetic therapy. Health Technology Assessment (Winchester, England), 5, 1–221. https://doi.org/10.3310/hta5090
[25] Posten, W., Wrone, D.A., Dover, J.S., Arndt, K.A., Silapunt, S. and Alam, M. (2006) Low-Level Laser Therapy for Wound Healing: Mechanism and Efficacy. Dermatologic Surgery, 31, 334–40. https://doi.org/10.1111/j.1524-4725.2005.31086
[26] Sousa, R.G.D. and Batista, K.D.N.M. (2016) Laser therapy in wound healing associated with diabetes mellitus - Review. Anais Brasileiros de Dermatologia, 91, 489–93. https://doi.org/10.1590/abd1806-4841.20163778
[27] Chen, Y.-T., Wang, H.-H., Wang, T.-J., Li, Y.-C. and Chen, T.-J. (2016) Early application of low-level laser may reduce the incidence of postherpetic neuralgia (PHN). Journal of the American Academy of Dermatology, 75, 572–7. https://doi.org/10.1016/j.jaad.2016.03.050
[28] Pol, R., Gallesio, G., Riso, M., Ruggiero, T., Scarano, A., Mortellaro, C. et al. (2016) Effects of Superpulsed, Low-Level Laser Therapy on Neurosensory Recovery of the Inferior Alveolar Nerve. Journal of Craniofacial Surgery, 27, 1215–9. https://doi.org/10.1097/SCS.0000000000002757
[29] Pol, R., Ruggiero, T., Gallesio, G., Riso, M., Bergamasco, L., Mortellaro, C. et al. (2016) Efficacy of Anti-Inflammatory and Analgesic of Superpulsed Low Level Laser Therapy After Impacted Mandibular Third Molars Extractions. Journal of Craniofacial Surgery, 27, 685–90. https://doi.org/10.1097/SCS.0000000000002523
[30] Coca, K.P., Marcacine, K.O., Gamba, M.A., Corrêa, L., Aranha, A.C.C. and Abrão, A.C.F.D.V. (2016) Efficacy of Low-Level Laser Therapy in Relieving Nipple Pain in Breastfeeding Women: A Triple-Blind, Randomized, Controlled Trial. Pain Management Nursing, 17, 281–9. https://doi.org/10.1016/j.pmn.2016.05.003
[31] Macias, D.M., Coughlin, M.J., Zang, K., Stevens, F.R., Jastifer, J.R. and Doty, J.F. (2015) Low-Level Laser Therapy at 635 nm for Treatment of Chronic Plantar Fasciitis: A Placebo-Controlled, Randomized Study. The Journal of Foot and Ankle Surgery, 54, 768–72. https://doi.org/10.1053/j.jfas.2014.12.014
[32] Li, Z.-J., Wang, Y., Zhang, H.-F., Ma, X.-L., Tian, P. and Huang, Y. (2016) Effectiveness of low-level laser on carpal tunnel syndrome: A meta-analysis of previously reported randomized trials. Medicine, 95, e4424. https://doi.org/10.1097/MD.0000000000004424
[33] Magri, L.V., Carvalho, V.A., Rodrigues, F.C.C., Bataglion, C. and Leite-Panissi, C.R.A. (2017) Effectiveness of low-level laser therapy on pain intensity, pressure pain threshold, and SF-MPQ indexes of women with myofascial pain. Lasers in Medical Science, 32, 419–28. https://doi.org/10.1007/s10103-016-2138-x
[34] Fernandes, G., Araújo Júnior, R.B., Lima, A.G., Gonzaga, I.A., De Oliveira, R. and Nicolau, R. (2017) Low-intensity laser (660 NM) has analgesic effects on sternotomy of patients who underwent coronary artery bypass grafts. Annals of Cardiac Anaesthesia, 20, 52. https://doi.org/10.4103/0971-9784.197836
[35] Fallah, A., Mirzaei, A., Gutknecht, N. and Demneh, A.S. (2017) Clinical effectiveness of low-level laser treatment on peripheral somatosensory neuropathy. Lasers in Medical Science, 32, 721–8. https://doi.org/10.1007/s10103-016-2137-y
[36] Eslamipour, F., Motamedian, S.R. and Bagheri, F. (2017) Ibuprofen and Low-level Laser Therapy for Pain Control during Fixed Orthodontic Therapy: A Systematic Review of Randomized Controlled Trials and Meta-analysis. The Journal of Contemporary Dental Practice, 18, 527–33. https://doi.org/10.5005/jp-journals-10024-2078
[37] Basford, J.R., Malanga, G.A., Krause, D.A. and Harmsen, W.S. (1998) A randomized controlled evaluation of low-intensity laser therapy: Plantar fasciitis. Archives of Physical Medicine and Rehabilitation, 79, 249–54. https://doi.org/10.1016/S0003-9993(98)90002-8
[38] Jorge, A.E.S., Dantas, L.O., Aburquerque-Sendín, F., Ferrari, A.V., Cunha, J.E., Dantas, G.A.D.F. et al. (2023) Photobiomodulation does not provide incremental benefits to patients with knee osteoarthritis who receive a strengthening exercises program: a randomized controlled trial. Brazilian Journal of Physical Therapy, 27, 100519. https://doi.org/10.1016/j.bjpt.2023.100519
[39] Ashleigh, T., Swerdlow, R.H. and Beal, M.F. (2023) The role of mitochondrial dysfunction in Alzheimer’s disease pathogenesis. Alzheimer’s & Dementia, 19, 333–42. https://doi.org/10.1002/alz.12683
[40] Lu, Y., Wang, R., Dong, Y., Tucker, D., Zhao, N., Ahmed, M.E. et al. (2017) Low-level laser therapy for beta amyloid toxicity in rat hippocampus. Neurobiology of Aging, 49, 165–82. https://doi.org/10.1016/j.neurobiolaging.2016.10.003
[41] De Taboada, L., Yu, J., El-Amouri, S., Gattoni-Celli, S., Richieri, S., McCarthy, T. et al. (2011) Transcranial Laser Therapy Attenuates Amyloid-β Peptide Neuropathology in Amyloid-β Protein Precursor Transgenic Mice. Journal of Alzheimer’s Disease, 23, 521–35. https://doi.org/10.3233/JAD-2010-100894
[42] Zhang, Z., Shen, Q., Wu, X., Zhang, D. and Xing, D. (2020) Activation of PKA/SIRT1 signaling pathway by photobiomodulation therapy reduces Aβ levels in Alzheimer’s disease models. Aging Cell, 19, e13054. https://doi.org/10.1111/acel.13054
[43] Shen, Q., Liu, L., Gu, X. and Xing, D. (2021) Photobiomodulation suppresses JNK3 by activation of ERK/MKP7 to attenuate AMPA receptor endocytosis in Alzheimer’s disease. Aging Cell, 20, e13289. https://doi.org/10.1111/acel.13289
[44] Blivet, G., Meunier, J., Roman, F.J. and Touchon, J. (2018) Neuroprotective effect of a new photobiomodulation technique against Aβ 25–35 peptide–induced toxicity in mice: Novel hypothesis for therapeutic approach of Alzheimer’s disease suggested. Alzheimer’s & Dementia: Translational Research & Clinical Interventions, 4, 54–63. https://doi.org/10.1016/j.trci.2017.12.003
[45] Berman, M.H., Halper, J.P., Nichols, T.W., H, J., Lundy, A. and Huang, J.H. (2017) Photobiomodulation with Near Infrared Light Helmet in a Pilot, Placebo Controlled Clinical Trial in Dementia Patients Testing Memory and Cognition. Journal of Neurology and Neuroscience, 08. https://doi.org/10.21767/2171-6625.1000176
[46] Saltmarche, A.E., Naeser, M.A., Ho, K.F., Hamblin, M.R. and Lim, L. (2017) Significant Improvement in Cognition in Mild to Moderately Severe Dementia Cases Treated with Transcranial Plus Intranasal Photobiomodulation: Case Series Report. Photomedicine and Laser Surgery, 35, 432–41. https://doi.org/10.1089/pho.2016.4227
[47] Chao, L.L. (2019) Effects of Home Photobiomodulation Treatments on Cognitive and Behavioral Function, Cerebral Perfusion, and Resting-State Functional Connectivity in Patients with Dementia: A Pilot Trial. Photobiomodulation, Photomedicine, and Laser Surgery, 37, 133–41. https://doi.org/10.1089/photob.2018.4555
[48] O’Brien, J.A. and Austin, P.J. (2019) Effect of Photobiomodulation in Rescuing Lipopolysaccharide-Induced Dopaminergic Cell Loss in the Male Sprague–Dawley Rat. Biomolecules, 9, 381. https://doi.org/10.3390/biom9080381
[49] Salgado, A. (2016) Effects of Light Emitting Diode and Low-intensity Light on the immunological process in a model of Parkinson’s disease. Medical Research Archives, 4. https://doi.org/10.18103/mra.v4i8.652
[50] Oueslati, A., Lovisa, B., Perrin, J., Wagnières, G., Van Den Bergh, H., Tardy, Y. et al. (2015) Photobiomodulation Suppresses Alpha-Synuclein-Induced Toxicity in an AAV-Based Rat Genetic Model of Parkinson’s Disease. Mosley RL, editor. PLOS ONE, 10, e0140880. https://doi.org/10.1371/journal.pone.0140880
[51] Purushothuman, S., Nandasena, C., Johnstone, D.M., Stone, J. and Mitrofanis, J. (2013) The impact of near-infrared light on dopaminergic cell survival in a transgenic mouse model of parkinsonism. Brain Research, 1535, 61–70. https://doi.org/10.1016/j.brainres.2013.08.047
[52] Darlot, F., Moro, C., El Massri, N., Chabrol, C., Johnstone, D.M., Reinhart, F. et al. (2016) Near‐infrared light is neuroprotective in a monkey model of P arkinson disease. Annals of Neurology, 79, 59–75. https://doi.org/10.1002/ana.24542
[53] San Miguel, M., Martin, K.L., Stone, J. and Johnstone, D.M. (2019) Photobiomodulation Mitigates Cerebrovascular Leakage Induced by the Parkinsonian Neurotoxin MPTP. Biomolecules, 9, 564. https://doi.org/10.3390/biom9100564
[54] McGee, C., Liebert, A., Bicknell, B., Pang, V., Isaac, V., McLachlan, C.S. et al. (2023) A Randomized Placebo-Controlled Study of a Transcranial Photobiomodulation Helmet in Parkinson’s Disease: Post-Hoc Analysis of Motor Outcomes. Journal of Clinical Medicine, 12, 2846. https://doi.org/10.3390/jcm12082846
[55] Hamilton, C.L., El Khoury, H., Hamilton, D., Nicklason, F. and Mitrofanis, J. (2019) “Buckets”: Early Observations on the Use of Red and Infrared Light Helmets in Parkinson’s Disease Patients. Photobiomodulation, Photomedicine, and Laser Surgery, 37, 615–22. https://doi.org/10.1089/photob.2019.4663
[56] Liebert, A., Bicknell, B., Laakso, E.-L., Jalilitabaei, P., Tilley, S., Kiat, H. et al. (2022) Remote Photobiomodulation Treatment for the Clinical Signs of Parkinson’s Disease: A Case Series Conducted During COVID-19. Photobiomodulation, Photomedicine, and Laser Surgery, 40, 112–22. https://doi.org/10.1089/photob.2021.0056
[57] Liebert, A., Bicknell, B., Laakso, E.-L., Heller, G., Jalilitabaei, P., Tilley, S. et al. (2021) Improvements in clinical signs of Parkinson’s disease using photobiomodulation: a prospective proof-of-concept study. BMC Neurology, 21, 256. https://doi.org/10.1186/s12883-021-02248-y
[58] Liebert, A., Bicknell, B., Laakso, E.-L., Tilley, S., Pang, V., Heller, G. et al. (2023) Improvements in the clinical signs of Parkinson’s disease using photobiomodulation: a 3-year follow-up case series. Medical Research Archives, 11. https://doi.org/10.18103/mra.v11i3.3690
[59] Shemesh, D., Aburus, O., Turgeman, G. and Abookasis, D. (2022) Quantification of the effectivity of laser therapy shortly following brain injury via dual-wavelength laser imaging. Optics & Laser Technology, 145, 107506. https://doi.org/10.1016/j.optlastec.2021.107506
[60] Xuan, W., Vatansever, F., Huang, L. and Hamblin, M.R. (2014) Transcranial low-level laser therapy enhances learning, memory, and neuroprogenitor cells after traumatic brain injury in mice. Journal of Biomedical Optics, 19, 108003. https://doi.org/10.1117/1.JBO.19.10.108003
[61] Xuan, W., Agrawal, T., Huang, L., Gupta, G.K. and Hamblin, M.R. (2015) Low-level laser therapy for traumatic brain injury in mice increases brain derived neurotrophic factor (BDNF) and synaptogenesis. Journal of Biophotonics, 8, 502–11. https://doi.org/10.1002/jbio.201400069
[62] Ando, T., Xuan, W., Xu, T., Dai, T., Sharma, S.K., Kharkwal, G.B. et al. (2011) Comparison of Therapeutic Effects between Pulsed and Continuous Wave 810-nm Wavelength Laser Irradiation for Traumatic Brain Injury in Mice. El Khoury J, editor. PLoS ONE, 6, e26212. https://doi.org/10.1371/journal.pone.0026212
[63] Wu, Q., Xuan, W., Ando, T., Xu, T., Huang, L., Huang, Y. et al. (2012) Low‐Level Laser Therapy for Closed‐Head Traumatic Brain Injury in Mice: Effect of Different Wavelengths. Lasers in Surgery and Medicine, 44, 218–26. https://doi.org/10.1002/lsm.22003
[64] Zhang, Q., Zhou, C., Hamblin, M.R. and Wu, M.X. (2014) Low-Level Laser Therapy Effectively Prevents Secondary Brain Injury Induced by Immediate Early Responsive Gene X-1 Deficiency. Journal of Cerebral Blood Flow & Metabolism, 34, 1391–401. https://doi.org/10.1038/jcbfm.2014.95
[65] Dong, T., Zhang, Q., Hamblin, M.R. and Wu, M.X. (2015) Low-Level Light in Combination with Metabolic Modulators for Effective Therapy of Injured Brain. Journal of Cerebral Blood Flow & Metabolism, 35, 1435–44. https://doi.org/10.1038/jcbfm.2015.87
[66] Xuan, W., Huang, L. and Hamblin, M.R. (2016) Repeated transcranial low‐level laser therapy for traumatic brain injury in mice: biphasic dose response and long‐term treatment outcome. Journal of Biophotonics, 9, 1263–72. https://doi.org/10.1002/jbio.201500336
[67] Naeser, M.A., Zafonte, R., Krengel, M.H., Martin, P.I., Frazier, J., Hamblin, M.R. et al. (2014) Significant Improvements in Cognitive Performance Post-Transcranial, Red/Near-Infrared Light-Emitting Diode Treatments in Chronic, Mild Traumatic Brain Injury: Open-Protocol Study. Journal of Neurotrauma, 31, 1008–17. https://doi.org/10.1089/neu.2013.3244
[68] Naeser, M.A., Saltmarche, A., Krengel, M.H., Hamblin, M.R. and Knight, J.A. (2011) Improved Cognitive Function After Transcranial, Light-Emitting Diode Treatments in Chronic, Traumatic Brain Injury: Two Case Reports. Photomedicine and Laser Surgery, 29, 351–8. https://doi.org/10.1089/pho.2010.2814
[69] Naeser, M.A., Martin, P.I., Ho, M.D., Krengel, M.H., Bogdanova, Y., Knight, J.A. et al. (2016) Transcranial, Red/Near-Infrared Light-Emitting Diode Therapy to Improve Cognition in Chronic Traumatic Brain Injury. Photomedicine and Laser Surgery, 34, 610–26. https://doi.org/10.1089/pho.2015.4037
[70] Chao, L.L., Barlow, C., Karimpoor, M. and Lim, L. (2020) Changes in Brain Function and Structure After Self-Administered Home Photobiomodulation Treatment in a Concussion Case. Frontiers in Neurology, 11, 952. https://doi.org/10.3389/fneur.2020.00952
[71] Czarny, P., Wigner, P., Galecki, P. and Sliwinski, T. (2018) The interplay between inflammation, oxidative stress, DNA damage, DNA repair and mitochondrial dysfunction in depression. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 80, 309–21. https://doi.org/10.1016/j.pnpbp.2017.06.036
[72] Wu, X., Alberico, S.L., Moges, H., De Taboada, L., Tedford, C.E. and Anders, J.J. (2012) Pulsed Light Irradiation Improves Behavioral Outcome in a Rat Model of Chronic Mild Stress. Lasers in Surgery and Medicine, 44, 227–32. https://doi.org/10.1002/lsm.22004
[73] Li, Y., Dong, Y., Yang, L., Tucker, L., Yang, B., Zong, X. et al. (2021) Transcranial photobiomodulation prevents PTSD-like comorbidities in rats experiencing underwater trauma. Translational Psychiatry, 11, 270. https://doi.org/10.1038/s41398-021-01389-5
[74] Mohammed, H.S. (2016) Transcranial low-level infrared laser irradiation ameliorates depression induced by reserpine in rats. Lasers in Medical Science, 31, 1651–6. https://doi.org/10.1007/s10103-016-2033-5
[75] Yang, L., Wu, C., Tucker, L., Dong, Y., Li, Y., Xu, P. et al. (2021) Photobiomodulation Therapy Attenuates Anxious-Depressive-Like Behavior in the TgF344 Rat Model. Journal of Alzheimer’s Disease, 83, 1415–29. https://doi.org/10.3233/JAD-201616
[76] Salehpour, F., Rasta, S.H., Mohaddes, G., Sadigh-Eteghad, S. and Salarirad, S. (2016) Therapeutic effects of 10-HzPulsed wave lasers in rat depression model: A comparison between near-infrared and red wavelengths: TRANSCRANIAL LOW-LEVEL LASER THERAPY IMPROVES DEPRESSION. Lasers in Surgery and Medicine, 48, 695–705. https://doi.org/10.1002/lsm.22542
[77] Schiffer, F., Johnston, A.L., Ravichandran, C., Polcari, A., Teicher, M.H., Webb, R.H. et al. (2009) 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. Behavioral and Brain Functions, 5, 46. https://doi.org/10.1186/1744-9081-5-46
[78] Cassano, P., Petrie, S.R., Mischoulon, D., Cusin, C., Katnani, H., Yeung, A. et al. (2018) Transcranial Photobiomodulation for the Treatment of Major Depressive Disorder. The ELATED-2 Pilot Trial. Photomedicine and Laser Surgery, 36, 634–46. https://doi.org/10.1089/pho.2018.4490
[79] Payne, B.A.I. and Chinnery, P.F. (2015) Mitochondrial dysfunction in aging: Much progress but many unresolved questions. Biochimica et Biophysica Acta (BBA) - Bioenergetics, 1847, 1347–53. https://doi.org/10.1016/j.bbabio.2015.05.022
[80] Grady, C. (2012) The cognitive neuroscience of ageing. Nature Reviews Neuroscience, 13, 491–505. https://doi.org/10.1038/nrn3256
[81] Cardoso, F.D.S., Barrett, D.W., Wade, Z., Gomes Da Silva, S. and Gonzalez-Lima, F. (2022) Photobiomodulation of Cytochrome c Oxidase by Chronic Transcranial Laser in Young and Aged Brains. Frontiers in Neuroscience, 16, 818005. https://doi.org/10.3389/fnins.2022.818005
[82] Salehpour, F., Ahmadian, N., Rasta, S.H., Farhoudi, M., Karimi, P. and Sadigh-Eteghad, S. (2017) Transcranial low-level laser therapy improves brain mitochondrial function and cognitive impairment in D-galactose–induced aging mice. Neurobiology of Aging, 58, 140–50. https://doi.org/10.1016/j.neurobiolaging.2017.06.025
[83] Hosseini, L., Farazi, N., Erfani, M., Mahmoudi, J., Akbari, M., Hosseini, S.H. et al. (2022) Effect of transcranial near-infrared photobiomodulation on cognitive outcomes in D-galactose/AlCl3 induced brain aging in BALB/c mice. Lasers in Medical Science, 37, 1787–98. https://doi.org/10.1007/s10103-021-03433-8
[84] Salehpour, F., De Taboada, L., Cassano, P., Kamari, F., Mahmoudi, J., Ahmadi-Kandjani, S. et al. (2018) A Protocol for Transcranial Photobiomodulation Therapy in Mice. Journal of Visualized Experiments, 59076. https://doi.org/10.3791/59076
[85] El Massri, N., Weinrich, T.W., Kam, J.H., Jeffery, G. and Mitrofanis, J. (2018) Photobiomodulation reduces gliosis in the basal ganglia of aged mice. Neurobiology of Aging, 66, 131–7. https://doi.org/10.1016/j.neurobiolaging.2018.02.019
[86] Chan, A.S., Lee, T.L., Yeung, M.K. and Hamblin, M.R. (2019) Photobiomodulation improves the frontal cognitive function of older adults. International Journal of Geriatric Psychiatry, 34, 369–77. https://doi.org/10.1002/gps.5039
[87] Chan, A.S., Lee, T., Hamblin, M.R. and Cheung, M. (2021) Photobiomodulation Enhances Memory Processing in Older Adults with Mild Cognitive Impairment: A Functional Near-Infrared Spectroscopy Study. Journal of Alzheimer’s Disease, 83, 1471–80. https://doi.org/10.3233/JAD-201600
[88] Saucedo, C.L., Courtois, E.C., Wade, Z.S., Kelley, M.N., Kheradbin, N., Barrett, D.W. et al. (2021) Transcranial laser stimulation: Mitochondrial and cerebrovascular effects in younger and older healthy adults. Brain Stimulation, 14, 440–9. https://doi.org/10.1016/j.brs.2021.02.011
[89] Qu, X., Li, L., Zhou, X., Dong, Q., Liu, H., Liu, H. et al. (2022) Repeated transcranial photobiomodulation improves working memory of healthy older adults: behavioral outcomes of poststimulation including a three-week follow-up. Neurophotonics, 9. https://doi.org/10.1117/1.NPh.9.3.035005
[90] Strouthos, I., Chatzikonstantinou, G., Tselis, N., Bon, D., Karagiannis, E., Zoga, E. et al. (2017) Photobiomodulation therapy for the management of radiation-induced dermatitis: A single-institution experience of adjuvant radiotherapy in breast cancer patients after breast conserving surgery. Strahlentherapie Und Onkologie, 193, 491–8. https://doi.org/10.1007/s00066-017-1117-x
[91] Costa, M.M., Silva, S.B., Quinto, A.L.P., Pasquinelli, P.F.S., De Queiroz Dos Santos, V., De Cássia Santos, G. et al. (2014) Phototherapy 660 nm for the prevention of radiodermatitis in breast cancer patients receiving radiation therapy: study protocol for a randomized controlled trial. Trials, 15, 330. https://doi.org/10.1186/1745-6215-15-330
[92] Robijns, J., Lodewijckx, J., Claes, S., Van Bever, L., Pannekoeke, L., Censabella, S. et al. (2021) Photobiomodulation therapy for the prevention of acute radiation dermatitis in head and neck cancer patients (DERMISHEAD trial). Radiotherapy and Oncology, 158, 268–75. https://doi.org/10.1016/j.radonc.2021.03.002
[93] Fife, D., Rayhan, D.J., Behnam, S., Ortiz, A., Elkeeb, L., Aquino, L. et al. (2010) A Randomized, Controlled, Double-Blind Study of Light Emitting Diode Photomodulation for the Prevention of Radiation Dermatitis in Patients with Breast Cancer. Dermatologic Surgery, 36, 1921–7. https://doi.org/10.1111/j.1524-4725.2010.01801.x
[94] Gobbo, M., Rico, V., Marta, G.N., Caini, S., Ryan Wolf, J., Van Den Hurk, C. et al. (2023) Photobiomodulation therapy for the prevention of acute radiation dermatitis: a systematic review and meta-analysis. Supportive Care in Cancer, 31, 227. https://doi.org/10.1007/s00520-023-07673-y
[95] Aguiar, B.R.L.D., Guerra, E.N.S., Normando, A.G.C., Martins, C.C., Reis, P.E.D.D. and Ferreira, E.B. (2021) Effectiveness of photobiomodulation therapy in radiation dermatitis: A systematic review and meta-analysis. Critical Reviews in Oncology/Hematology, 162, 103349. https://doi.org/10.1016/j.critrevonc.2021.103349
[96] Tunér, J. (2020) Is Photobiomodulation Therapy Cost Effective? Photobiomodulation, Photomedicine, and Laser Surgery, 38, 193–4. https://doi.org/10.1089/photob.2020.4805
[97] Glass, G.E. (2023) Photobiomodulation: A Systematic Review of the Oncologic Safety of Low-Level Light Therapy for Aesthetic Skin Rejuvenation. Aesthetic Surgery Journal, 43, NP357–71. https://doi.org/10.1093/asj/sjad018

Comments (0)

There are no comments for this article. Be the first one to leave a message!

Leave a comment

Please note: comments must be approved before they are published

AI-generated responses are for informational purposes only and do not constitute medical advice. Accuracy, completeness, or timeliness are not guaranteed. Use at your own risk.

Trixie - AI assistant

close