Abstract
Photobiomodulation using light in the red or near-infrared region is an innovative treatment strategy for a wide range of neurological and psychological conditions. Photobiomodulation can promote neurogenesis and elicit anti-apoptotic, anti-inflammatory and antioxidative responses. Its therapeutic effects have been demonstrated in studies on neurological diseases, peripheral nerve injuries, pain relief and wound healing. We conducted a comprehensive literature review of the application of photobiomodulation in patients with central nervous system diseases in February 2019. The NCBI PubMed database, EMBASE database, Cochrane Library and ScienceDirect database were searched. We reviewed 95 papers and analyzed. Photobiomodulation has wide applicability in the treatment of stroke, traumatic brain injury, Parkinson’s disease, Alzheimer’s disease, major depressive disorder, and other diseases. Our analysis provides preliminary evidence that PBM is an effective therapeutic tool for the treatment of central nervous system diseases. However, additional studies with adequate sample size are needed to optimize treatment parameters.
Keywords: Alzheimer's disease, central nervous system diseases, major depressive disorder, Parkinson's disease, photobiomodulation, stroke, traumatic brain injury
Introduction
Photobiomodulation (PBM), an innovative therapeutical approach, utilizes light in the red (with wavelengths usually in the range of 600 to 700 nm) or near-infrared region (780 to 1100 nm), at a relatively low power density to minimize tissue damage (McGuff et al., 1965; Hennessy and Hamblin, 2017; Gordon and Johnstone, 2019). The photons can cause chemical changes within the cells and provoke various reactions, including the triggering of neuroprotective responses, improving blood flow, inducing metabolic changes and neurogenesis (Mitrofanis and Henderson, 2020). In 1967, Dr. Endre Mester first proposed the medical benefits of low-level laser therapy. Numerous studies thereafter investigated the medical application of low-level laser therapy and PBM. The therapeutic effects of PBM have been demonstrated in many studies on neurological diseases (McGuff et al., 1965), peripheral nerve injuries, pain relief (De Freitas and Hamblin, 2016) and wound healing (Houreld, 2014).
While the mechanisms underlying the therapeutic effects of PBM remain unclear, it has been thought that the photons induce the production of reactive oxygen species, increase electron transport, and trigger a series of downstream reactions. The resulting products, including nitric oxide (NO), reactive oxygen species, cyclic AMP and Ca 2+, are second messengers that can activate transcription factors and impact the expression of genes related to cell proliferation and migration, inflammation and apoptosis (Avci et al. 2013; De Freitas and Hamblin, 2016). PBM can increase cerebral blood flow (CBF), enhance cellular metabolism, and prevent neurodegeneration (Rojas et al., 2012; Salehpour et al., 2018). Transcranial PBM refers to near-infrared light (NIR) applied to the head to treat neurological diseases. Research on transcranial PBM is still in infancy, but the limited studies in humans have shown encouraging outcomes in the treatment of stroke, traumatic brain injury (TBI), Parkinson’s disease (PD), Alzheimer’s disease (AD) and major depressive disorder (MDD). However, its clinical application still remains controversial. Overall, the results are not yet consistent as parameters has been continuously tested and optimized. Therefore, to assess the therapeutic potential of PBM, we conducted this review to summarize existing studies on PBM in the central nervous system (CNS) diseases.
Literature Search
To evaluate the current application of PBM in CNS diseases, we conducted a literature review of all published original research studies involving PBM in subjects with CNS diseases. Articles involving treatment for stroke, TBI, PD, AD and MDD were included.
The literature search was conducted up to January 2019 using the NCBI PubMed database, EMBASE database, Cochrane Library and ScienceDirect database using the following search terms: (“transcranial photobiomodulation”) OR ((photobiomodulation OR “low level laser therapy”) AND brain) OR ((photobiomodulation OR “low level laser therapy”) AND (brain injury OR stroke OR cerebrovascular disease OR depressive disorder OR neurodegenerative disease)). Only English language articles published in peer-reviewed journals were included. The details of the included studies are presented in Tables 1–6. In total, we identified 95 published papers relating to stroke, TBI, PD, AD and MDD.
Table 1.
Animal studies | Animals | Modeling method | Wavelength (nm) | Irradiation parameters | Power density/energy density |
Lapchak et al. (2004) | 14 Male New Zealand white rabbits | Microclots were prepared from blood drawn from a donor rabbit and allowed to clot at 37°C | 808 | CW | 7 mW/cm2 for 2 min (0.84 J/cm2) or 25 mW/cm2 for 10 min (15 J/cm2) |
Lapchak et al. (2007) | Male New Zealand white rabbits | Injection of clot particle suspension | 808 | PW at 100 Hz or 1000 Hz, or CW | 7.5 mW/cm2, 0.9–1.2 J |
DeTaboada et al. (2006) | 169 Atherothrombotic model rats | / | 808 | / | 7.5 mW/cm2 at brain tissue level, 0.9 J/cm2 per site (in total 2 sites) |
Oron et al. (2006) | 43 Adult male Sprague-Dawley rats; 18 male Wistar rats | (1) Permanent occlusion of the middle cerebral artery through a craniotomy or (2) insertion of a filament | 808 | PW at 70 Hz or CW | 7.5 mW/cm2 at brain tissue level, 0.9 J/cm2 per site (in total 2 sites) |
Yang et al. (2018) | Male Sprague-Dawley rats | / | 808±3.0 | 25 mW/cm2 at cerebral cortex tissue level, 350 mW/cm2 on the scalp | |
Leung et al. (2002) | Male adult Sprague-Dawley rats | Unilateral occlusion of middle cerebral artery | 660 | PW at 10 Hz | 8.8 mW, 2.64, 13.2, or 26.4 J/cm2 |
Lapchak et al. (2008) | 89 Male New Zealand white rabbits | Injection of emboli | 808 | CW | 10 mW/cm2 |
Lapchak and De Taboada (2010) | 24 Male New Zealand white rabbits | Injection of emboli | 808 | PW at 100 Hz or CW | 7.5, 37.5, or 262.5 mW/cm2; 0.9, 4.5, or 31.5 J/cm2 |
Yip et al. (2011) | 12 Male Sprague-Dawley rats | Occlusion of right middle cerebral artery for 1 h | 606 | PW at 10 Hz | 8.8 mW, 2.64, 13.20, or 26.40 J/cm2 |
Choi et al. (2012) | Male Wistar rats | Occlusion of the right middle cerebral artery | 710 | CW | 0.042 mW/cm2, 1.796 J/cm2 |
Huisa et al. (2013) | Male New Zealand white rabbits | Injection of microemboli | 808.5 | CW | 7.5, 10.8, or 20 mW/cm2 |
Fukuzaki et al. (2015) | Adult FVB mice | Occlusion of bilateral common carotid artery | 532 | CW | 845 mW/cm2, 30.4 × 102 J/cm2 |
Lapchak and Boitano (2016) | 60 Male New Zealand white rabbits | Injection of emboli | 808 | CW | 7.5 mW/cm2, 0.9 J/cm2 |
Lee et al. (2016) | Male mice (C57BL/6J) | Photothrombosis of the cortical microvessels | 610 | CW | 1.7 mW/cm2, 2 J/cm2 |
Meyer et al. (2016) | One male New Zealand white rabbits | Injection of emboli | 808.5 | CW or PW at 10 or 100 Hz | 7.5–333 mW/cm2 |
Lee et al. (2017a) | Mouse photothrombotic cerebral focal ischemia model | / | 610 | CW | 1.7 mW/cm2, 2 J/cm2 |
Lee et al. (2017b) | 17 Male C57BL/6J wild-type and eNOS mice | Occlusion of the right middle cerebral artery | 610 | CW | 1.7 mW/cm2, 2 J/cm2 |
Yun et al. (2017) | 24 Male Sprague-Dawley rats | Occlusion of the left middle cerebral artery | 650 | PW at 100 Hz | 30 mW |
Argibay et al. (2019) | Male Sprague-Dawley rats | Occlusion of the middle cerebral artery | 830 | CW | 0.28 J / cm2 |
Clinical studies | Subjects | Wavelength (nm) | Irradiation parameters | Power density/energy density | |
Lampl et al. (2007) | 120 Patients | 808 | CW | 10 mW/cm2, 1.2 J/cm2 | |
Zivin et al. (2009) | 660 Patients | 808 | CW | 10 mW/cm2, 1.2 J/cm2 | |
Zivin et al. (2014) | 630,316 Patients were allocated to treatment group versus 314 allocated to controls | 808 | CW | 10 mW/cm2, 1.2 J/cm2 | |
Boonswang et al. (2012) | A 29-year-old woman with brainstem stroke | 660 and 850 | CW | 1400 mW, 2.95 J/cm2 | |
das Neves et al. (2016) | 15 Subjects (6 males and 9 females) with cerebrovascular accident and spastic hemiparesis | 808 | CW | 3.18 W/cm2, 127.39 J/cm2 | |
Jan et al. (2017) | 38 Patients; LASER group (20 patients) and interferential current group (18 patients). | 905 | CW | 400 mW, 6 J/cm2 |
CW: Continuous wave; eNOS: endothelial nitric oxide synthase; PW: pulsed wave.
Table 6.
Studies | Animals/subjects | Modeling method | Wavelength (nm) | Irradiation parameters | Power density/energy density |
---|---|---|---|---|---|
Muili et al. (2012) | 17 C57BL/6 mouse model of multiple sclerosis | Induction with myelin oligodendrocyte glycoprotein | 670 | CW | 5 J/cm2 |
Leisman et al. (2018) | 40 Patients with autism spectrum disorder | 635 | CW | A power output of 15 mW | |
Yang et al. (2019) | Sprague-Dawley rats with neonatal hypoxic ischemic encephalopathy | Ligation of the right common carotid artery | 808 | CW | 100 mW/cm2; 12 J/cm2 |
CW: Continuous wave.
Photobiomodulation for Stroke
As summarized in Table 1, PBM has been evaluated in stroke animal models and patients. Lapchak et al. (2004) investigated the efficacy of laser therapy for stroke in a rabbit small clot embolic stroke model (RSCEM). They found that PBM improved behavioral performance and had long-term benefits. They also compared the effects of continuous wave (CW) or pulse wave (PW) PBM, and concluded that PW provides better outcome (Lapchak et al., 2007). In another study, 169 rats were irradiated ipsilaterally, contralaterally and on both sides, and all treated groups showed significant improvement (DeTaboada et al., 2006). The significant functional improvement provided by PBM may be associated with the induction of neurogenesis (Oron, 2006). Studies on C17.2 immortalized mouse neural progenitor cell lines show that PBM significantly increases cellular proliferation (Argibay et al., 2019). Yang et al. (2018) investigated the effect of PBM on neurogenesis. PBM promoted the proliferation and differentiation of neural progenitor cells in the peri-infarct zone and the switch from an M1 microglial phenotype to an anti-inflammatory M2 phenotype, thereby improving microenvironment and mitochondrial function.
Despite the encouraging results in animal stroke studies, laser therapy has limited success in humans. Early studies were not successful. A series of three clinical trials termed “NeuroThera Effectiveness and Safety Trials” (NEST-1 (Lampl, 2007), NEST-2 (Zivin, 2009), and NEST-3 (Zivin et al., 2014)) have evaluated the efficacy of PBM in stroke patients. Lampl et al. (2007)recruited 120 ischemic stroke patients, with 79 patients in the experimental group and 41 in the control group. More patients (70%) in the experimental group had favorable outcomes than controls (51%), as assessed with the National Institutes of Health Stroke Scale (NIHSS) and modified Rankin Scale (mRS). In NEST-2 with 660 patients, the group given transcranial laser therapy showed slightly, but not significantly better outcome than the control group. There were no significant differences in mortality rates or serious adverse events in term of safety data (Zivin, 2009). NEST-3 was prematurely terminated for futility (an expected lack of statistical significance) (Zivin et al., 2014). Researchers tend to attribute this failure to the violation of RIGOR guidelines (Lapchak and Boitano, 2016). In a case study, a 29-year-old woman who suffered a brainstem stroke showed improvement in both cognitive state and motor recovery after 8 weeks of PBM (Boonswang et al., 2012). The accelerated recovery in motor functions was also observed in a study of 15 patients with post-stroke spasticity (das Neves et al., 2016). After three consecutive phases, the group treated with PBM showed significant reduction in pain intensity. PBM was also effective in ameliorating post-stroke shoulder pain (Jan et al., 2017).
Photobiomodulation for Traumatic Brain Injury
We identified 21 papers reporting on PBM for TBI, including 15 animal studies and 6 clinical studies (Table 2). Oron et al. (2007) investigated the therapeutic effectiveness of PBM in mice with traumatic brain injury (TBI). They evaluated the effects of two PBM modes (PW versus CW), and found a substantial improvement and better outcome with pulsed laser mode at 100 Hz (Oron et al., 2012). Ando et al. (2011) found that 10-Hz pulse frequency was more effective than CW and 100-Hz mode with a wavelength of 810 nm. The effectiveness of 810 nm is also supported by another study (Wu et al., 2012). Anders et al. (2014) proposed that the parameters can be optimized with in vitro models, and then followed by in vivo research and clinical application.
Table 2.
Animal studies | Animal models | Modeling method | Wavelength (nm) | Irradiation parameters | Power density/energy density |
---|---|---|---|---|---|
Oron et al. (2007) | 24 Mice | Weight-drop device | 808 | CW | 10 or 20 mW/cm2, 1.2 or 2.4 J/cm2 |
Oron et al. (2012) | / | Weight-drop device | 808 | PW at 100 Hz or CW | / |
Ando et al. (2011) | 40 Mice | Controlled cortical impact | 810 | CW; PW at 10 Hz and 100 Hz | 50 mW/cm2, 36 J/cm2 |
Wu et al. (2012) | 28 Adult male BALB/c mice | Controlled weight drop onto the skull | 665, 730, 810, or 980 | CW | 150 mW/cm2, 36 J/cm2 |
Anders et al. (2014) | 22 New Zealand white rabbits | Controlled cortical impact | 810 and 980 | CW | 10 mW/cm2; 2–200 mJ/cm2 |
Moreira et al. (2009) | 51 Adult male Wistar rats | Cryogenic brain injury | 660 or 780 | CW | 40 mW, 3 or 5 J/cm2 per site (2 sites in total) |
Moreira et al. (2011) | Forty adult male Wistar rats (Rattus norvegicus albinus) | Cryogenic brain injury | 780 | CW | 40 mW, 3 J/cm2 |
Khuman et al. (2012) | 239 Male C57BL/6 mice | Controlled cortical impact | 800 | CW | 500 mW/cm2, 60 J/cm2 |
Quirk et al. (2012) | 104 Sprague-Dawley rats | Controlled cortical impact | 670 | CW | 50 mW/cm2, 15 J/cm2 |
Xuan et al. (2013) | 144 Adult male BALB/c mice | Cortical impact; the bone flap was removed and mice were subjected to controlled cortical impact using a pneumatic impact device | 810 | CW | 25 mW/cm2, 18 J/cm2 |
Xuan et al. (2014) | 64 Young adult male BALB/c mice | Controlled cortical impact | 810 | CW | 25 mW/cm2, 18 J/cm2 |
Xuan et al. (2015) | 40 Male BALB/c mice | Controlled cortical impact | 810 | CW | 50 mW/cm2, 36 J/cm2 |
Xuan et al. (2016) | 96 Male BALB/c mice | Cortical impact; the bone flap was removed and mice were subjected to controlled cortical impact using a pneumatic impact device | 810 | CW | 25 mW/cm2, 18 J/cm2 |
Zhang et al. (2014) | Wild-type mice and IEX-1 knockout mice on 129Sv/C57BL/6 background | Controlled cortical impact | 810 | PW at 10 Hz | 150 mW/cm2, 36 J/cm2 |
Dong et al. (2015) | C57BL/6 mice | Controlled cortical impact | 810 | PW at 10 Hz | 150 mW/cm2; 36 J/cm2 |
Clinical studies | Subjects | Wavelength (nm) | Irradiation parameters | Power density/energy density | |
Naeser et al. (2011) | Two chronic, traumatic brain injury cases | 633 and 870 | CW | 19.39 mW/cm2 and 22.48 mW/cm2, 13.3 J/cm2 | |
Naeser et al. (2014) | Eleven chronic, mild traumatic brain injury participants | 633 and 870 | CW | 500 mW, 22.48 mW/cm2, 13 J/cm2 | |
Nawashiro et al. (2012) | Patients in a persistent vegetative state | 850 | CW | 11.4 mW/cm2; the energy density 20.5 J/cm2 | |
Henderson et al. (2015) | A patient with moderate traumatic brain injury | 810 and 980 | CW | 10–15 W | |
Hipskind et al. (2018) | Twelve symptomatic military veterans with chronic traumatic brain injury > 18 months post-trauma | 220 | CW | 6.4 mW/cm2 for 20 min | |
Morries et al. (2015) | Ten patients with chronic traumatic brain injury | 810 and 980 | PW at 10 Hz | 10 and 15 W, 14.8–28.3 J/cm2 |
CW: Continuous wave; PW: pulsed wave.
Several studies have investigated the underlying mechanisms. Moreira et al. (2009) found that PBM affected local and systemic immune functions following cryogenic brain injury by modulating tumor necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1β) and interleukin-6 (IL-6) levels. They also showed that PBM prevented neuronal death and severe astrogliosis, thereby promoting wound healing (Moreira et al., 2011). Reduced microgliosis was also observed in the PBM-treated group in another study (Khuman et al., 2012). In addition, PBM may exert neuroprotective effects by upregulating mitochondrial function and decreasing oxidative stress (Quirk et al., 2012). Xuan et al. (2013) found that mice in the treatment group had smaller lesion size at 28 days and fewer degenerating neurons, suggesting that PBM therapy may encourage neurogenesis. They further discovered that laser therapy promoted neurogenesis in the hippocampus and subventricular zone by upregulating brain-derived neurotrophic factor, which may stimulate synaptogenesis and at least partially account for the improved memory and learning function (Xuan et al., 2014, 2015). Xuan et al. (2014, 2015) observed an interesting biphasic dose-response relationship in which the effect of PBM seemed to decline with increasing laser exposure. They designed another study with two groups given 3 or 14 sessions daily of PBM treatment, and found that the negative effect of excessive PBM was temporary and might be caused by temporary induction of reactive gliosis. With longer follow-up time, mice given 14 sessions started to show steady improvement (Xuan et al., 2016). Zhang et al. (2014) investigated the effect of PBM on secondary brain injury in mice lacking immediate early responsive gene X-1 (IEX-1). Laser therapy regulated proinflammatory mediators and increased ATP levels, promoting brain recovery. The recovery of learning and memory function was associated with reduced loss of hippocampal tissue compared with the control group (Dong et al., 2015).
Six human studies, all case series, with 37 patients in total have been done in traumatic brain injury with various results. Naeser et al. (2011) reported two cases with closed-head TBI that showed significant cognitive improvement and reduced cost of treatment. They then conducted a study in eleven chronic TBI patients. They found improvement in learning ability, which was positively correlated with treatment duration (Naeser et al., 2014). In other case reports, clinical symptoms, including depression, anxiety, headache and insomnia, were reduced after laser therapy, which might be associated with increased regional cerebral blood flow (Nawashiro et al., 2012; Henderson and Morries, 2015). Hipskind et al. (2018) investigated its effect on cognitive functional improvement and regional cerebral blood flow in 12 symptomatic military veterans diagnosed with chronic TBI.
Photobiomodulation for Parkinson’s Disease
In vitro studies have provided preliminarily support for a protective effect of PBM against 1-methyl-4-phenylpyridinium ion (MPTP)-induced neurotoxicity, supporting its application in in vivo studies (Dilworth et al., 1975; Liang et al., 2008; Ying et al., 2008; Trimmer et al., 2009). Peoples et al. (2012) found that laser therapy given concomitantly or after chronic MPTP administration protected dopaminergic cells from degeneration in the MPTP mouse model of PD (Table 3). The effect was long lasting, even after minimal exposure (Shaw et al., 2012). Moro et al. (2013) contributed greatly to the assessment of the efficacy and safety of laser treatment. They found higher numbers of tyrosine hydroxylase (TH)-positive cells in the laser-treated groups in both C57BL/6 (pigmented) and Balb/c (albino) mice. The albino mice showed better outcome because of greater penetration of NIR through the skin and fur. They then investigated its safety in MPTP-treated mice (Moro et al., 2014) and monkeys (Moro et al., 2016). NIR caused no observable behavioral deficits, nor was there evidence of tissue necrosis, suggesting NIR can be applied intracranially. Its effects on monkey PD models have also been investigated, and this primate model might be more suitable for pre-clinical studies (Shaw et al., 2010a; Darlot et al., 2016).
Table 3.
Animal studies | Animals | Modeling method | Wavelength (nm) | Irradiation parameters | Power density/energy density |
---|---|---|---|---|---|
Peoples et al. (2012) | 80 Male albino BALB/c mice | Injection of MPTP | 670 | CW | 5 J/cm2; 90 s |
Shaw et al. (2012) | 96 Male albino BALB/c mice | Injection of MPTP | 670 | CW | 0.5 J/cm2 |
Moro et al. (2013) | 40 Male BALB/c (albino) and 40 C57BL/6 (pigmented) mice | Injection of MPTP | CW | / | |
Moro et al. (2014) | 36 Male BALB/c mice and 3 Sprague-Dawley rats | Injection of MPTP | 670 | PW, CW | 1.5 mW/cm2 (PW) or 14.5 mW/cm2 (CW) |
Moro et al. (2016) | 15 Monkeys | Injection of MPTP | 670 | PW with 5 s ON/60 s OFF | Lower doses (25 J or 35 J); higher dose (125 J) |
Darlot et al. (2015) | A monkey | Injection of MPTP | 670 | PW with 5 s ON/60 s OFF | 10 mW; 25 or 35 J |
Shaw et al. (2014) | 12 Adult male macaque monkeys (Macaca fascicularis, Mauritius); 30 adult male albino BALB/c mice | Injection of MPTP | CW | / | |
Reinhart et al. (2015) | Male BALB/c mice | Injection of MPTP | 810 | / | 5.3 mW/cm3 |
Reinhart et al. (2016) | 147 Male BALB/c mice | Injection of MPTP | 670 | CW | 5.3 mW/cm2, 0.5 J/cm2 |
Reinhart et al. (2016) | 62 Male BALB/c mice | Injection of MPTP | 670 and/or 810 | CW | 15 or 30 mW |
El Massri et al. (2014) | 130 Male BALB/c mice | Injection of MPTP | 670 | CW | 5.3 mW/cm2 |
Purushothuman et al. (2013) | K3 transgenic mouse model (K369I tau transgenic model (K3)) | Transgenic mouse model | 670 | CW | 80 J/cm2 |
Vos et al. (2013) | Pink1 null mutants | Rotenone treatment | 808 | CW | 10–25 mW/cm2 |
Johnstone et al. (2014) | 143 Male BALB/c mice | Injection of MPTP | 670 | CW | 50 mW/cm2, 4 J/cm2; 90 s |
El Massri et al. (2016) | 24 Adult Macaque monkeys (Macaca fascicularis) | Injection of MPTP | 670 | PW with 5 s ON/60 s OFF | 10 mW; 25 or 35 J over 7 days |
El Massri et al. (2017) | 17 Balb/c mice, 15 Wistar rats and 16 macaque monkeys (Macaca fascicularis) | Injection of MPTP | 670 | CW | 0.16 mW for mouse and rat, and 10 mW for monkey |
El Massri et al. (2018) | 12 Macaque monkeys (Macaca fascicularis) | Injection of MPTP | 670 | / | / |
Kim et al. (2018) | 10 Male C57BL/6 mice/group | Injection of MPTP | 670 | CW | 50 mW/cm2, 3 min |
Oueslati et al. (2015) | 23 Sprague-Dawley female rats (Charles River Laboratories) | Injection of 2 μL of viral suspension | 808 | / | 2.5 mW/cm2 (n = 7) and 5 mW/cm2 (n = 7) |
Ganeshan et al. (2019) | 62 Male BALB/c mice | Injection of MPTP | 670 | CW | 50mW/cm2; 4J/cm2 per day |
Reinhart et al. (2016) | 61 Male Wistar rats | Injection of 6-OHDA | 670 | PW, CW | 333 nW or 0.16 mW, 634 mJ or 304 J |
Shaw et al. (2010b) | BALB/c albino mice | Injections of MPTP | 670 | CW | 40 mW/cm2 at scalp, 5.3 mW/cm2 inside skull, 0.47 J/cm2 |
CW: Continuous wave; PW: pulsed wave.
Reinhart et al. (2015) evaluated the impact of different treatment parameters. They showed that 810 nm laser therapy had a more immediate therapeutic effect than 670 nm (Reinhart et al., 2015). They also investigated the effects of laser therapy before, at the same time, and after injection of MPTP. These investigators found that all three treatments produced similar outcomes in their PD model (Reinhart et al., 2016a). In addition, exposure to 670 nm and 810 nm NIR either together or sequentially produced better results than either alone, especially together (Reinhart et al., 2016b). El Massri et al. (2016a) investigated the effect of different doses of NIR. The positive effect of PBM seemed to be dose-dependent—exposure to higher doses of NIR had a longer protective effect and was associated with reduced astrogliosis. Further studies are needed to optimize treatment parameters.
Several studies have investigated the mechanisms underlying the therapeutic effects of laser therapy. Purushothuman et al. (2013) found that NIR treatment reduced oxidative stress and inhibited neurodegeneration. Mitochondrial dysfunction has been observed in PD animal models and patients. PBM can improve mitochondrial function and cellular metabolism (Vos et al., 2013). Interestingly, it has been observed that unilateral exposure to NIR can have a bilateral effect. Indirect light may rescue TH+ cells in the substantia nigra pars compacta, possibly via unidentified mediators. This indirect effect is diminished by high-dose MPTP exposure (Johnstone et al., 2014).
El Massri et al. (2016b) discovered changes in the glial response, especially in astrocytes, after laser therapy in a monkey model of PD. These investigators further found that trophic factors, such as glial-derived neurotrophic factor, in the striatum may also play a role during NIR therapy (El Massri et al., 2017). In a subsequent study, their research group focused on encephalopsin, which is expressed by two populations of striatal interneurons constituting complex networks. Although PBM seemed to have no notable effect, external light seemed to exert an effect on the network of encephalopsin-expressing cells (El Massri et al., 2018).
A number of recent studies have examined the indirect effects of PBM. For example, PBM applied distally can trigger brain protective mechanisms, saving crucial neurons in PD (Kim et al., 2018). Consistent with previous studies (Purushothuman et al., 2013; Oueslati et al., 2015; Vos et al., 2016), remote PBM was demonstrated to modulate a variety of signaling pathways, thereby upregulating cell signaling and migration, including CXCR4+ stem cells, adipocytokine signaling and nuclear factor erythroid 2-related factor 2 expression, in turn modulating cellular oxidative stress response pathways. In addition, PBM affects the blood-brain barrier and might reduce damage to the brain (Ganeshan et al., 2019).
Photobiomodulation for Alzheimer’s Disease
Aβ plaques and hyperphosphorylated tau are observed in patients with AD. NIR was shown to reduce Aβ plaques in the brain of a transgenic AD mouse model in a dose-dependent manner (De Taboada et al., 2011; Grillo et al., 2013) (Table 4). Grillo et al. (2013) reported upregulation of heat shock proteins in an AD model; however, a significant downregulation of heat shock proteins was observed after treatment with 1072-nm NIR. Purushothuman et al. (2014) used two different mouse models of AD: the K369I tau transgenic model (K3) that develops neurofibrillary tangles, and the APPswe/PSEN1dE9 transgenic model (APP/PS1) that develops Aβ plaques. Both of these characteristic features of AD were reduced after NIR treatment (Purushothuman et al., 2014). These investigators subsequently examined the therapeutic effects of NIR treatment on the cerebellum (Purushothuman et al., 2015). A recent study demonstrated that PBM improves spatial memory and behavioral performance (da Luz Eltchechem et al., 2017). As mentioned above, PBM can impact signaling pathways, and thereby regulate cell proliferation, migration and apoptosis. In an AD model, NIR induces proliferation of CD11b-positive monocytes, which appear to remove plaques by phagocytosis (Farfara et al., 2015). Because inflammatory responses and oxidative stress are associated with the development of AD (De Felice and Ferreira, 2014; Urrutia et al., 2014), PBM may ameliorate mitochondrial dysfunction in the disease. Indeed, Lu et al. (2017) showed that PBM inhibits G6PDH and NADPH oxidase activities, thereby reducing reactive oxygen species production and oxidative stress.
Table 4.
Studies | Animals/Subjects | Modeling method | Wavelength (nm) | Irradiation parameters | Power density/energy density |
De Taboada et al. (2011) | One hundred male transgenic Aβ PP mice | Microinjection of human Aβ PP gene | 808 | PW at 100 Hz, or CW | 10 mW/cm2; 1.2, 6, or 12 J/cm2 |
Grillo et al. (2013) | TASTPM mice | Transgenic mouse model | 1072 | PW at 600 Hz | 5 mW/cm2, 1.8 J/cm2 |
Purushothuman et al. (2014) | 15 K3 mice or 18 APP/PS1 mice | Transgenic mouse model | 670 | CW | 4 J/cm2; 90-second treatment equates to 4 J/cm2; a total of 80 J/cm2 was delivered to the skull over the 4 weeks; 90 seconds |
Purushothuman et al. (2015) | 10 K3 and 12 APP/PS1 transgenic mice | Transgenic mouse model | 670 | CW | 4 J/cm2 |
da Luz Eltchechem et al. (2017) | 60 Male Wistar rats (Rattus Norvegicus) | Transgenic mouse model | 627 | / | 7 J/cm2, 70 mW |
Farfara et al. (2015) | 5XFAD transgenic male mice (Tg6799) | Transgenic mouse model | / | CW | 400 mW, 1 J/cm2 |
Lu et al. (2017) | 12 Male Sprague-Dawley rats | Transgenic mouse model | 808 | CW | 25 mW/cm2, 3 J/cm2 |
Saltmarche et al. (2017) | Five participants with dementia or Alzheimer‘s disease | / | 810 | PW at 10 Hz | 14.2 mW/cm2; 10.65 J/cm2 |
Berman et al. (2017) | 11 Participants | / | 1072 | PW at 10 Hz | / |
Chao (2019) | 8 Participants with dementia | / | 810 | PW at 40 Hz | 75 mW/cm2, 45 J/cm2 |
CW: Continuous wave; PW: pulsed wave.
Human studies on the effects of PBM are still limited. Saltmarche et al. (2017) reported a case series of five patients given PBM therapy. The subjects showed cognitive improvement and better emotional control after a 4-week treatment period. No side effects were observed. In another controlled trial with 11 participants, no significant difference was found between the PBM group and controls, possibly because of small sample size (Berman et al., 2017). Chao (2019) found increased cerebral perfusion in eight participants diagnosed with dementia after 12 weeks of PBM. Given the encouraging outcomes in animal studies, further well-designed clinical trials with larger sample size and long-term follow-up are warranted.
Photobiomodulation for Major Depressive Disorder
Major depressive disorder (MDD) is one of the most common psychiatric disorders. PBM has been found to be potentially effective in the treatment of MDD (Table 5). In studies investigating PBM for TBI, immobility time in the forced swim test was reported to be decreased in the treatment group, suggesting an anti-depressive effect of PBM (Ando et al., 2011; Wu et al., 2012). Salehpour and Rasta (2017) assessed the effects of low-level laser therapy (10 Hz PW, 810 nm) in the chronic mild stress model of depression, compared with citalopram. Immobility time was significantly decreased in both groups; however, no significant reduction in anxiety-like behavior was detected in the elevated plus maze test. An antidepressant-like effect of PBM was also observed in the model of reserpine-induced depression, as evaluated by forced swim test and electrocorticography (Mohammed, 2016). Xu et al. (2017) reported that the NIR-treated group showed better outcomes in behavioral despair tests, and found that this improvement was associated with the modulation of neurotransmitter levels and improved mitochondrial function in the prefrontal cortex. Furthermore, PBM has been shown to reduce oxidative stress and superoxide anion levels (Salehpour et al., 2019). In a randomized double-blind controlled study with 30 patients with depression, a significant difference was observed in Beck Depression Inventory scores between the laser therapy and control groups (Quah-Smith et al., 2005). Schiffer et al. (2009) used the Hamilton Depression Rating Scale (HAM-D) and Hamilton Anxiety Rating Scale (HAM-A) to evaluate the efficacy of PBM in 10 patients. Cassano et al. (2015) investigated the safety of 700 mW/cm 2NIR, and reported that no serious adverse events were observed. High power NIR provides persistent and better results compared with low power NIR (Henderson and Morries, 2017). In addition, PBM can be used in combination with other treatment modalities to enhance therapeutic effectiveness. For example, laser therapy combined with attention bias modification can enhance cognitive improvement (Disner et al., 2016). A case report of a 76-year-old white woman diagnosed with MDD with anxious distress showed steady improvement (Caldieraro et al., 2018).
Table 5.
Animal studies | Animals | Wavelength (nm) | Irradiation parameters | |
Ando et al. (2011) | 40 Male BALB/c mice | Depression following traumatic brain injury | 810 | CW; PW at 10 Hz and 100 Hz |
Wu et al. (2012) | 32 Adult male BALB/c mice | Chronic mild stress | 810 | PW at 100 Hz |
Salehpour and Rasta (2016) | 50 Adult male BALB/c mice | Chronic mild stress | 630 or 810 | PW at 10 Hz |
Mohammed (2016) | 24 Adult male albino rats | Reserpine induced depression | 804 | CW |
Xu et al. (2016) | / | Depression induced by Ahi1 KO or space restriction | 808 | CW |
Salehpour et al. (2018) | 75 Adult male BALB/c mice | Sub-chronic restraint stress | 810 | PW at 10 Hz |
Clinical studies | Subjects | Wavelength (nm) | Irradiation parameters | Irradiation parameters |
Quah-Smith et al. (2005) | 30 Patients with elevated depressive symptoms | 804 | CW | / |
Schiffer et al. (2009) | 10 Patients with treatment-resistant major depressive disorder | 810 | CW | 250 mW/cm2, 60 J/cm2 |
Cassano et al. (2015) | 4 Patients with major depressive disorder | 808 | CW | 5 W, 700 mW/cm2, 84 J/cm2 |
Henderson et al. (2017) | 39 Patients with traumatic brain injury presenting with depressive symptoms | 810, 980 | CW | 8–15 W |
Disner et al. (2016) | Fifty-one adult participants with elevated symptoms of depression | 1064 | CW | 250 mW/cm2, 60 J/cm2 |
Caldieraro et al. (2018) | One patient with major depressive disorder with anxious distress | 830 | CW | 36mW/cm2; 80J/cm2 |
CW: Continuous wave; KO: knock-out; PW: pulsed wave.
Other Applications
PBM has been shown to be effective in other CNS diseases as well (Table 6). Muili et al. (2012) found amelioration of symptoms in a mouse model of multiple sclerosis. A study reported improvement of autism spectrum disorder in children and adolescents of 5–17 years of age after PBM treatment (Leisman et al., 2018). PBM can also prevent ischemic injury to neurons after global cerebral ischemia caused by cardiac arrest and neonatal hypoxic-ischemic encephalopathy (HIE) (Tucker et al., 2018; Yang et al., 2019). PBM attenuates hypoxic-ischemic brain injury by maintaining mitochondrial function, decreasing oxidative stress and inhibiting neuronal apoptosis.
Discussion
PBM with NIR delivered noninvasively to the deep brain tissue has wide application in the treatment of neurological diseases. Numerous studies have demonstrated its efficacy in stroke, TBI, PD, AD, MDD and other disorders. The low power density laser, insufficient to burn or damage tissue, has no adverse effects on non-human primates (Moro et al., 2017). Notably, no adverse events have been reported in clinical trials.
The parameters of PBM, including wavelength, operation mode, power density and treatment duration, are critical factors to optimize therapeutic effectiveness (Salehpour et al., 2018). The wavelengths affect the absorption and penetration depth. Light has been employed in recent studies with wavelengths in the red including 606, 627, 630, 632.8, 640, 660 and 670 nm, and in the NIR regions including 785, 800, 804, 808, 810, 830 and 850 nm. NIR wavelengths produce more favorable outcomes. PBM has CW and PW modes. Studies have shown that PW mode at 10, 40 and 100 Hz provides better outcomes compared with CW. Pulsed light at 10 or 40 Hz may better affect brain activity. In addition, PBM with energy densities of 0.1–15 J/cm2 is effective for neurons in animal models, whereas 10–84 J/cm2 is effective in humans. PBM treatment appears to observe a biphasic dose-response relationship that follows the Arndt-Schulz Law. It has a stimulatory effect at low doses, but after the peak, stronger stimuli are inhibitory, leading to a negative response (Huang et al., 2011). Therefore, treatment dose and duration are of great importance. However, optimal parameters have not yet been determined.
The application of 670 nm and 810 nm NIR together or sequentially provides better outcome than individually (Reinhart et al., 2017). PBM combined with intranasal and/or transcranial light-emitting diodes has notable advantages for long-term therapy in that it can be performed at home for long-term use (Naeser et al., 2011).
Given favorable outcomes in pre-clinical and clinical studies, the application of PBM in CNS diseases has a promising future. However, studies with larger sample size are needed for a consensus on treatment parameters. An improved apparatus with optimal parameters could enhance the efficacy and safety of PBM, and allow its application to be standardized to minimize side effects.
Footnotes
C-Editor: Zhao M; S-Editor: Song LP; L-Editor: Song LP; T-Editor: Jia Y
Conflicts of interest: The authors declare no conflicts of interest.
Financial support: None.
Copyright license agreement: The Copyright License Agreement has been signed by all authors before publication.
Plagiarism check: Checked twice by iThenticate.
Peer review: Externally peer reviewed.
References
- 1.Anders JJ, Moges H, Wu X, Erbele ID, Alberico SL, Saidu EK, Smith JT, Pryor BA. In vitro and in vivo optimization of infrared laser treatment for injured peripheral nerves. Lasers Surg Med. 2014;46:34–45. doi: 10.1002/lsm.22212. [DOI] [PubMed] [Google Scholar]
- 2.Ando T, Xuan W, Xu T, Dai T, Sharma SK, Kharkwal GB, Huang YY, Wu Q, Whalen MJ, Sato S, Obara M, Hamblin MR. Comparison of therapeutic effects between pulsed and continuous wave 810-nm wavelength laser irradiation for traumatic brain injury in mice. PLoS One. 2011;6:e26212. doi: 10.1371/journal.pone.0026212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Argibay B, Campos F, Perez-Mato M, Vieites-Prado A, Correa-Paz C, López-Arias E, Da Silva-Candal A, Moreno V, Montero C, Sobrino T, Castillo J, Iglesias-Rey R. Light-emitting diode photobiomodulation after cerebral Ischemia. Front Neurol. 2019;22:10–911. doi: 10.3389/fneur.2019.00911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Avci P, Nyame TT, Gupta GK, Sadasivam M, Hamblin MR. Low-level laser therapy for fat layer reduction: a comprehensive review. Lasers Surg Med. 2013;45:349–357. doi: 10.1002/lsm.22153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Berman MH, Halper JP, Nichols TW. Photobiomodulation with near infrared light helmet in a, placebo controlled clinical trial in dementia patients testing memory and cognition. J Neurol Neurosci. 2017;8:176. doi: 10.21767/2171-6625.1000176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Boonswang NA, Chicchi M, Lukachek A, Curtiss D. A new treatment protocol using photobiomodulation and muscle/bone/joint recovery techniques having a dramatic effect on a stroke patient’s recovery: a new weapon for clinicians. BMJ Case Rep. 2012;2012 doi: 10.1136/bcr.08.2011.4689. pii: bcr0820114689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Caldieraro MA, Sani G, Bui E, Cassano P. Long-term near-infrared photobiomodulation for anxious depression complicated by Takotsubo cardiomyopathy. J Clin Psychopharmacol. 2018;38:268–270. doi: 10.1097/JCP.0000000000000883. [DOI] [PubMed] [Google Scholar]
- 8.Cassano P, Cusin C, Mischoulon D, Hamblin MR, De Taboada L, Pisoni A, Chang T, Yeung A, Ionescu DF, Petrie SR, Nierenberg AA, Fava M, Iosifescu DV. Near-infrared transcranial radiation for major depressive disorder: proof of concept study. Psychiatry J. 2015;2015:352979. doi: 10.1155/2015/352979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chao LL. Effects of home photobiomodulation treatments on cognitive and behavioral, cerebral perfusion, and resting-state functional connectivity in patients with dementia: a pilot trial. Photobiomodul Photomed Laser Surg. 2019;37:133–141. doi: 10.1089/photob.2018.4555. [DOI] [PubMed] [Google Scholar]
- 10.Choi DH, Lim JH, Lee KH, Kim MY, Kim HY, Shin CY, Han SH, Lee J. Effect of 710-nm visible light irradiation on neuroprotection and immune function after stroke. Neuroimmunomodulation. 2012;19:267–276. doi: 10.1159/000335547. [DOI] [PubMed] [Google Scholar]
- 11.da Luz Eltchechem C, Salgado ASI, Zângaro RA, da Silva Pereira MC, Kerppers II, da Silva LA, Parreira RB. Transcranial LED therapy on amyloid-β toxin 25–35 in the hippocampal region of rats. Lasers Med Sci. 2017;32:749–756. doi: 10.1007/s10103-017-2156-3. [DOI] [PubMed] [Google Scholar]
- 12.Darlot F, Moro C, El Massri N, Chabrol C, Johnstone DM, Reinhart F, Agay D, Torres N, Bekha D, Auboiroux V, Costecalde T, Peoples CL, Anastascio HD, Shaw VE, Stone J, Mitrofanis J, Benabid AL. Near-infrared light is neuroprotective in a monkey model of Parkinson disease. Ann Neurol. 2016;79:59–75. doi: 10.1002/ana.24542. [DOI] [PubMed] [Google Scholar]
- 13.das Neves MF, Dos Reis MC, de Andrade EA, Lima FP, Nicolau RA, Arisawa EÂ, Andrade AO, Lima MO. Effects of low-level laser therapy (LLLT 808 nm) on lower limb spastic muscle activity in chronic stroke patients. Lasers Med Sci. 2016;31:1293–1300. doi: 10.1007/s10103-016-1968-x. [DOI] [PubMed] [Google Scholar]
- 14.De Felice FG, Ferreira ST. Inflammation, defective insulin signaling, and mitochondrial dysfunction as common molecular denominators connecting type 2 diabetes to Alzheimer disease. Diabetes. 2014;63:2262–2272. doi: 10.2337/db13-1954. [DOI] [PubMed] [Google Scholar]
- 15.De Freitas LF, Hamblin MR. Proposed mechanisms of potobiomodulation or low-level light therapy. IEEE J Sel Top Quant. 2016;22:1–17. doi: 10.1109/JSTQE.2016.2561201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.De Taboada L, Yu J, El-Amouri S, Gattoni-Celli S, Richieri S, McCarthy T, Streeter J, Kindy MS. Transcranial laser therapy attenuates amyloid-β peptide neuropathology in amyloid-β protein precursor transgenic mice. J Alzheimers Dis. 2011;23:521–535. doi: 10.3233/JAD-2010-100894. [DOI] [PubMed] [Google Scholar]
- 17.De Taboada L, Ilic S, Leichliter-Martha S, Oron U, Oron A, Streeter J. Transcranial application of low-energy laser irradiation improves neurological deficits in rats following acute stroke. Lasers Surg Med. 2006;38:70–73. doi: 10.1002/lsm.20256. [DOI] [PubMed] [Google Scholar]
- 18.Dilworth JA, Stewart P, Gwaltney JM, Jr, Hendley JO, Sande MA. Methods to improve detection of pneumococci in respiratory secretions. J Clin Microbiol. 1975;2:453–455. doi: 10.1128/jcm.2.5.453-455.1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Disner SG, Beevers CG, Gonzalez-Lima F. Transcranial laser stimulation as neuroenhancement for attention bias modification in adults with elevated depression symptoms. Brain Stimul. 2016;9:780–787. doi: 10.1016/j.brs.2016.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Dong T, Zhang Q, Hamblin MR, Wu MX. Low-level light in combination with metabolic modulators for effective therapy of injured brain. J Cereb Blood Flow Metab. 2015;35:1435–1444. doi: 10.1038/jcbfm.2015.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.El Massri N, Cullen KM, Stefani S, Moro C, Torres N, Benabid AL, Mitrofanis J. Evidence for encephalopsin immunoreactivity in interneurones and striosomes of the monkey striatum. Exp Brain Res. 2018;236:955–961. doi: 10.1007/s00221-018-5191-9. [DOI] [PubMed] [Google Scholar]
- 22.El Massri N, Johnstone DM, Peoples CL, Moro C, Reinhart F, Torres N, Stone J, Benabid AL, Mitrofanis J. The effect of different doses of near infrared light on dopaminergic cell survival and gliosis in MPTP-treated mice. Int J Neurosci. 2016a;126:76–87. doi: 10.3109/00207454.2014.994063. [DOI] [PubMed] [Google Scholar]
- 23.El Massri N, Moro C, Torres N, Darlot F, Agay D, Chabrol C, Johnstone DM, Stone J, Benabid AL, Mitrofanis J. Near-infrared light treatment reduces astrogliosis in MPTP-treated monkeys. Exp Brain Res. 2016b;234:3225–3232. doi: 10.1007/s00221-016-4720-7. [DOI] [PubMed] [Google Scholar]
- 24.El Massri N, Lemgruber AP, Rowe IJ, Moro C, Torres N, Reinhart F, Chabrol C, Benabid AL, Mitrofanis J. Photobiomodulation-induced changes in a monkey model of Parkinson’s disease: changes in tyrosine hydroxylase cells and GDNF expression in the striatum. Exp Brain Res. 2017;235:1861–1874. doi: 10.1007/s00221-017-4937-0. [DOI] [PubMed] [Google Scholar]
- 25.Farfara D, Tuby H, Trudler D, Doron-Mandel E, Maltz L, Vassar RJ, Frenkel D, Oron U. Low-level laser therapy ameliorates disease progression in a mouse model of Alzheimer’s disease. J Mol Neurosci. 2015;55:430–436. doi: 10.1007/s12031-014-0354-z. [DOI] [PubMed] [Google Scholar]
- 26.Fukuzaki Y, Shin H, Kawai HD, Yamanoha B, Kogure S. 532 nm low-power laser irradiation facilitates the migration of GABAergic neural stem/progenitor cells in mouse neocortex. PLoS One. 2015;10:e0123833. doi: 10.1371/journal.pone.0123833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ganeshan V, Skladnev NV, Kim JY, Mitrofanis J, Stone J, Johnstone DM. Pre-conditioning with remote photobiomodulation modulates the brain transcriptome and protects against MPTP insult in mice. Neuroscience. 2019;400:85–97. doi: 10.1016/j.neuroscience.2018.12.050. [DOI] [PubMed] [Google Scholar]
- 28.Gordon LC, Johnstone DM. Remote photobiomodulation: an emerging strategy for neuroprotection. Neural Regen Res. 2019;14:2086–2087. doi: 10.4103/1673-5374.262573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Grillo S, Duggett N, Ennaceur A, Chazot P. Non-invasive infrared therapy (1072 nm) reduces β-amyloid protein levels in the brain of an Alzheimer’s disease mouse model, TASTPM. J Photochem Photobiol B Biol. 2013;123:13–22. doi: 10.1016/j.jphotobiol.2013.02.015. [DOI] [PubMed] [Google Scholar]
- 30.Hamblin, Michael R. Photobiomodulation for traumatic brain injury and stroke. J Neurosci Res. 2017;2017 doi: 10.1002/jnr.24190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Henderson TA, Morries LD. SPECT perfusion imaging demonstrates improvement of traumatic brain injury with transcranial near-infrared laser phototherapy. Adv Mind Body Med. 2015;29:27–33. [PubMed] [Google Scholar]
- 32.Henderson TA, Morries LD. Multi-watt near-infrared phototherapy for the treatment of comorbid depression: an open-label single-arm study. Front Psychiatry. 2017;8:187. doi: 10.3389/fpsyt.2017.00187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hennessy M, Hamblin MR. Photobiomodulation and the brain: a new paradigm. J Opt. 2017;19:013003. doi: 10.1088/2040-8986/19/1/013003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hipskind SG, Grover FL, Jr, Fort TR, Helffenstein D, Burke TJ, Quint SA, Bussiere G, Stone M, Hurtado T. Pulsed transcranial red/near-infrared light therapy using light-emitting diodes improves cerebral blood flow and cognitive function in veterans with chronic traumatic brain injury: a case series. Photomed Laser Surg. 2018;28 doi: 10.1089/pho.2018.4489. [DOI] [PubMed] [Google Scholar]
- 35.Houreld NN. Shedding light on a new treatment for diabetic wound healing: a review on phototherapy. Sci World J. 2014;2014:1–13. doi: 10.1155/2014/398412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Huang YY, Sharma SK, Carroll J, Hamblin MR. Biphasic dose response in low level light therapy - an update. Dose Response. 2011;9:602–618. doi: 10.2203/dose-response.11-009.Hamblin. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Huisa BN, Chen Y, Meyer BC, Tafreshi GM, Zivin JA. Incremental treatments with laser therapy augments good behavioral outcome in the rabbit small clot embolic stroke model. Lasers Med Sci. 2013;28:1085–1089. doi: 10.1007/s10103-012-1193-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Jan F, Naeem A, Malik AN, Amjad I, Malik T. Comparison of low level laser therapy and interferential current on post stroke shoulder pain. J Pak Med Assoc. 2017;67:788–789. [PubMed] [Google Scholar]
- 39.Johnstone DM, el Massri N, Moro C, Spana S, Wang XS, Torres N, Chabrol C, De Jaeger X, Reinhart F, Purushothuman S, Benabid AL, Stone J, Mitrofanis J. Indirect application of near infrared light induces neuroprotection in a mouse model of parkinsonism - an abscopal neuroprotective effect. Neuroscience. 2014;274:93–101. doi: 10.1016/j.neuroscience.2014.05.023. [DOI] [PubMed] [Google Scholar]
- 40.Khuman J, Zhang J, Park J, Carroll JD, Donahue C, Whalen MJ. Low-level laser light therapy improves cognitive deficits and inhibits microglial activation after controlled cortical impact in mice. J Neurotrauma. 2012;29(2):408–417. doi: 10.1089/neu.2010.1745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kim B, Mitrofanis J, Stone J, Johnstone DM. Remote tissue conditioning is neuroprotective against MPTP insult in mice. IBRO Rep. 2018;31(4):14–17. doi: 10.1016/j.ibror.2018.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.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;38:1843–1849. doi: 10.1161/STROKEAHA.106.478230. [DOI] [PubMed] [Google Scholar]
- 43.Lapchak P, Salgado K, Chao C, Zivin J. Transcranial near-infrared light therapy improves motor function following embolic strokes in rabbits: an extended therapeutic window study using continuous and pulse frequency delivery modes. Neuroscience. 2007;148:907–914. doi: 10.1016/j.neuroscience.2007.07.002. [DOI] [PubMed] [Google Scholar]
- 44.Lapchak PA, Boitano PD. A novel method to promote behavioral improvement and enhance mitochondrial function following an embolic stroke. Brain Res. 2016;1646:125–131. doi: 10.1016/j.brainres.2016.04.039. [DOI] [PubMed] [Google Scholar]
- 45.Lapchak PA, De Taboada L. Transcranial near infrared laser treatment (NILT) increases cortical adenosine-5’-triphosphate (ATP) content following embolic strokes in rabbits. Brain Res. 2010;1306:100–105. doi: 10.1016/j.brainres.2009.10.022. [DOI] [PubMed] [Google Scholar]
- 46.Lapchak PA, Han MK, Salgado KF, Streeter J, Zivin JA. Safety profile of transcranial near-infrared laser therapy administered in combination with thrombolytic therapy to embolized rabbits. Stroke. 2008;39:3073–3078. doi: 10.1161/STROKEAHA.108.516393. [DOI] [PubMed] [Google Scholar]
- 47.Lapchak PA, Wei J, Zivin JA. Transcranial infrared laser therapy improves clinical rating scores after embolic strokes in rabbits. Stroke. 2004;35:1985–1988. doi: 10.1161/01.STR.0000131808.69640.b7. [DOI] [PubMed] [Google Scholar]
- 48.Lee HI, Lee SW, Kim NG, Park KJ, Choi BT, Shin YI, Shin HK. a Low-level light emitting diode (LED) therapy suppresses inflammasome-mediated brain damage in experimental ischemic. J Biophotonics. 2017a;10:1502–1513. doi: 10.1002/jbio.201600244. [DOI] [PubMed] [Google Scholar]
- 49.Lee HI, Lee SW, Kim SY, Kim NG, Park KJ, Choi BT, Shin YI, Shin HK. b Pretreatment with light-emitting diode therapy reduces ischemic brain injury in mice through endothelial nitric oxide synthase-dependent. Biochem Biophys Res Commun. 2017b;486:945–950. doi: 10.1016/j.bbrc.2017.03.131. [DOI] [PubMed] [Google Scholar]
- 50.Lee HI, Park JH, Park MY, Kim NG, Park KJ, Choi BT, Shin YI, Shin HK. Pre-conditioning with transcranial low-level light therapy reduces neuroinflammation and protects blood-brain barrier after focal cerebral ischemia in mice. Restor Neurol Neurosci. 2016;34:201–214. doi: 10.3233/RNN-150559. [DOI] [PubMed] [Google Scholar]
- 51.Leisman G, Machado C, Machado Y, Chinchilla-Acosta M. Effects of low-level laser therapy in autism spectrum disorder. Adv Exp Med Biol. 2018;1116:111–130. doi: 10.1007/5584_2018_234. [DOI] [PubMed] [Google Scholar]
- 52.Leung MC, Lo SC, Siu FK, So KF. Treatment of experimentally induced transient cerebral ischemia with low energy laser inhibits nitric oxide synthase activity and up-regulates the expression of transforming growth factor-beta 1. Lasers Surg Med. 2002;31:283–288. doi: 10.1002/lsm.10096. [DOI] [PubMed] [Google Scholar]
- 53.Liang HL, Whelan HT, Eells JT, Wong-Riley MT. Near-infrared light via light-emitting diode treatment is therapeutic against rotenone-and 1-methyl-4-phenylpyridinium ion-induced neurotoxicity. Neuroscience. 2008;153:963–974. doi: 10.1016/j.neuroscience.2008.03.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lu Y, Wang R, Dong Y, Tucker D, Zhao N, Ahmed ME, Zhu L, Liu TC, Cohen RM, Zhang Q. Low-level laser therapy for beta amyloid toxicity in rat hippocampus. Neurobiol Aging. 2017;49:165–182. doi: 10.1016/j.neurobiolaging.2016.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.McGuff PE, Deterling RA, Jr, Gottlieb LS. Tumoricidal effect of laser energy on experimental and human malignant tumors. N Engl J Med. 1965;273:490–492. doi: 10.1056/NEJM196508262730906. [DOI] [PubMed] [Google Scholar]
- 56.Meyer DM, Chen Y, Zivin JA. Dose-finding study of phototherapy on stroke outcome in a rabbit model of ischemic stroke. Neurosci Lett. 2016;630:254–258. doi: 10.1016/j.neulet.2016.06.038. [DOI] [PubMed] [Google Scholar]
- 57.Mitrofanis J, Henderson LA. How and why does photobiomodulation change brain activity. Neural Regen Res. 2020;15:2243–2244. doi: 10.4103/1673-5374.284989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Mohammed HS. Transcranial low-level infrared laser irradiation ameliorates depression induced by reserpine in rats. Lasers Med Sci. 2016;31:1651–1656. doi: 10.1007/s10103-016-2033-5. [DOI] [PubMed] [Google Scholar]
- 59.Moreira MS, Velasco IT, Ferreira LS, Ariga SK, Abatepaulo F, Grinberg LT, Marques MM. Effect of laser phototherapy on wound healing following cerebral ischemia by cryogenic injury. J Photochem Photobiol B. 2011;105:207–215. doi: 10.1016/j.jphotobiol.2011.09.005. [DOI] [PubMed] [Google Scholar]
- 60.Moreira MS, Velasco IT, Ferreira LS, Ariga SK, Barbeiro DF, Meneguzzo DT, Abatepaulo F, Marques MM. Effect of phototherapy with low intensity laser on local and systemic immunomodulation following focal brain damage in rat. J Photochem Photobiol B. 2009;97:145–151. doi: 10.1016/j.jphotobiol.2009.09.002. [DOI] [PubMed] [Google Scholar]
- 61.Moro C, El Massri N, Darlot F, Torres N, Chabrol C, Agay D, Auboiroux V, Johnstone DM, Stone J, Mitrofanis J, Benabid AL. Effects of a higher dose of near-infrared light on clinical signs and neuroprotection in a monkey model of Parkinson’s disease. Brain Res. 2016;1648(Pt A):19–26. doi: 10.1016/j.brainres.2016.07.005. [DOI] [PubMed] [Google Scholar]
- 62.Moro C, Massri NE, Torres N, Ratel D, De Jaeger X, Chabrol C, Perraut F, Bourgerette A, Berger M, Purushothuman S, Johnstone D, Stone J, Mitrofanis J, Benabid AL. Photobiomodulation inside the brain: a novel method of applying near-infrared light intracranially and its impact on dopaminergic cell survival in MPTP-treated mice. J Neurosurg. 2014;120:670–683. doi: 10.3171/2013.9.JNS13423. [DOI] [PubMed] [Google Scholar]
- 63.Moro C, Torres N, Arvanitakis K, Cullen K, Chabrol C, Agay D, Darlot F, Benabid AL, Mitrofanis J. No evidence for toxicity after long-term photobiomodulation in normal non-human primates. Exp Brain Res. 2017;235:3081–3092. doi: 10.1007/s00221-017-5048-7. [DOI] [PubMed] [Google Scholar]
- 64.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;14:40. doi: 10.1186/1471-2202-14-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Morries LD, Cassano P, Henderson TA. Treatments for traumatic brain injury with emphasis on transcranial near-infrared laser phototherapy. Neuropsychiatr Dis Treat. 2015;11:2159–2175. doi: 10.2147/NDT.S65809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Muili KA, Gopalakrishnan S, Meyer SL, Eells JT, Lyons JA. Amelioration of experimental autoimmune encephalomyelitis in C57BL/6 mice by photobiomodulation induced by 670 nm light. PLoS One. 2012;7:e30655. doi: 10.1371/journal.pone.0030655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Naeser MA, Saltmarche A, Krengel MH, Hamblin MR, Knight JA. Improved cognitive function after, light-emitting diode treatments in chronic, traumatic brain injury: Two case reports. Photomed Laser Surg. 2011;29:351–358. doi: 10.1089/pho.2010.2814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Naeser MA, Saltmarche A, Krengel MH, Hamblin MR, Knight JA. Improved cognitive function after, light-emitting diode treatments in chronic, traumatic brain injury: two case reports. Photomed Laser Surg. 2011;29:351–358. doi: 10.1089/pho.2010.2814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Naeser MA, Zafonte R, Krengel MH, Martin PI, Frazier J, Hamblin MR, Knight JA, Meehan WP, III, Baker EH. Significant improvements in cognitive performance, red/near-infrared light-emitting diode treatments in chronic, mild traumatic brain injury: Open-protocol study. J Neurotrauma. 2014;31:1008–1017. doi: 10.1089/neu.2013.3244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Nawashiro H, Wada K, Nakai K, Sato S. Focal increase in cerebral blood flow after treatment with near-infrared light to the forehead in a patient in a persistent vegetative state. Photomed Laser Surg. 2012;30:231–233. doi: 10.1089/pho.2011.3044. [DOI] [PubMed] [Google Scholar]
- 71.Oron A, Oron U, Chen J, Eilam A, Zhang C, Sadeh M, Lampl Y, Streeter J, DeTaboada L, Chopp M. Low-level laser therapy applied transcranially to rats after induction of stroke significantly reduces long-term neurological deficits. Stroke. 2006;37:2620–2624. doi: 10.1161/01.STR.0000242775.14642.b8. [DOI] [PubMed] [Google Scholar]
- 72.Oron A, Oron U, Streeter J, De Taboada L, Alexandrovich A, Trembovler V, Shohami E. Near infrared transcranial laser therapy applied at various modes to mice following traumatic brain injury significantly reduces long-term neurological deficits. J Neurotrauma. 2012;29:401–407. doi: 10.1089/neu.2011.2062. [DOI] [PubMed] [Google Scholar]
- 73.Oron A, Oron U, Streeter J, Taboada LD, Alexandrovich A, Trembovler V, Shohami E. Low-level laser therapy applied transcranially to mice following traumatic brain injury significantly reduces long-term neurological deficits. J Neurotrauma. 2007;24:651–656. doi: 10.1089/neu.2006.0198. [DOI] [PubMed] [Google Scholar]
- 74.Oueslati A, Lovisa B, Perrin J, Wagnières G, van den Bergh H, Tardy Y, Lashuel HA. Photobiomodulation suppresses alpha-synuclein-induced toxicity in an AAV-based rat genetic model of Parkinson’s disease. PLoS One. 2015;10:e0140880. doi: 10.1371/journal.pone.0140880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.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;18:469–476. doi: 10.1016/j.parkreldis.2012.01.005. [DOI] [PubMed] [Google Scholar]
- 76.Purushothuman S, Johnstone DM, Nandasena C, Mitrofanis J, Stone J. Photobiomodulation with near infrared light mitigates Alzheimer’s disease-related pathology in cerebral cortex-evidence from two transgenic mouse models. Alzheimers Res Ther. 2014;6:2. doi: 10.1186/alzrt232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Purushothuman S, Johnstone DM, Nandasena C, van Eersel J, Ittner LM, Mitrofanis J, Stone J. Near infrared light mitigates cerebellar pathology in transgenic mouse models of dementia. Neurosci Lett. 2015;591:155–159. doi: 10.1016/j.neulet.2015.02.037. [DOI] [PubMed] [Google Scholar]
- 78.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;1535:61–70. doi: 10.1016/j.brainres.2013.08.047. [DOI] [PubMed] [Google Scholar]
- 79.Quah-Smith JI, Tang WM, Russell Laser acupuncture for mild to moderate depression in a primary care setting--a randomised controlled trial. Acupunct Med. 2005;23:103–111. doi: 10.1136/aim.23.3.103. [DOI] [PubMed] [Google Scholar]
- 80.Quirk BJ, Torbey M, Buchmann E, Verma S, Whelan HT. Near-infrared photobiomodulation in an animal model of traumatic brain injury: Improvements at the behavioral and biochemical levels. Photomed Laser Surg. 2012;30:523–529. doi: 10.1089/pho.2012.3261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Reinhart F, El Massri N, Johnstone DM, Stone J, Mitrofanis J, Benabid AL, Moro C. Near-infrared light (670 nm) reduces MPTP-induced parkinsonism within a broad therapeutic time window. Exp Brain Res. 2016a;234:178–1794. doi: 10.1007/s00221-016-4578-8. [DOI] [PubMed] [Google Scholar]
- 82.Reinhart F, Massri NE, Chabrol C, Cretallaz C, Johnstone DM, Torres N, Darlot F, Costecalde T, Stone J, Mitrofanis J, Benabid AL, Moro C. Intracranial application of near-infrared light in a hemi-parkinsonian rat model: the impact on behavior and cell survival. J Neurosurg. 2016b;124:1829–1841. doi: 10.3171/2015.5.JNS15735. [DOI] [PubMed] [Google Scholar]
- 83.Reinhart F, Massri NE, Darlot F, Torres N, Johnstone DM, Chabrol C, Costecalde T, Stone J, Mitrofanis J, Benabid AL, Moro C. 810nm near-infrared light offers neuroprotection and improves locomotor activity in MPTP-treated mice. Neurosci Res. 2015;92:86–90. doi: 10.1016/j.neures.2014.11.005. [DOI] [PubMed] [Google Scholar]
- 84.Reinhart F, Massri NE, Torres N, Chabrol C, Molet J, Johnstone DM, Stone J, Benabid AL, Mitrofanis J, Moro C. The behavioural and neuroprotective outcomes when 670 nm and 810 nm near infrared light are applied together in MPTP-treated mice. Neurosci Res. 2017;117:42–47. doi: 10.1016/j.neures.2016.11.006. [DOI] [PubMed] [Google Scholar]
- 85.Rojas JC, Bruchey AK, Gonzalez-Lima F. Low-level light therapy improves cortical metabolic capacity and memory retention. J Alzheimers Dis. 2012;32:741–752. doi: 10.3233/JAD-2012-120817. [DOI] [PubMed] [Google Scholar]
- 86.Salehpour F, Farajdokht F, Cassano P, Sadigh-Eteghad S, Erfani M, Hamblin MR, Salimi MM, Karimi P, Rasta SH, Mahmoudi J. Near-infrared photobiomodulation combined with coenzyme Q10 for depression in a mouse model of restraint stress: reduction in oxidative stress, neuroinflammation, and apoptosis. Brain Res Bull. 2019;144:213–222. doi: 10.1016/j.brainresbull.2018.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Salehpour F, Mahmoudi J, Kamari F, Sadigh-Eteghad S, Rasta SH, Hamblin MR. Brain photobiomodulation therapy: a narrative review. Mol Neurobiol. 2018;55:6601–6636. doi: 10.1007/s12035-017-0852-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Salehpour F, Rasta SH. The potential of transcranial photobiomodulation therapy for treatment of major depressive disorder. Rev Neurosci. 2017;28:441–453. doi: 10.1515/revneuro-2016-0087. [DOI] [PubMed] [Google Scholar]
- 89.Saltmarche AE, Naeser MA, Ho KF, Hamblin MR, Lim L. Significant improvement in cognition in mild to moderately severe dementia cases treated with transcranial plus intranasal photobiomodulation: case series report. Photomed Laser Surg. 2017;35:432–441. doi: 10.1089/pho.2016.4227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.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;5:46. doi: 10.1186/1744-9081-5-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Shaw VE, Keay KA, Ashkan K, Benabid AL, Mitrofanis J. Dopaminergic cells in the periaqueductal grey matter of MPTP-treated monkeys and mice; patterns of survival and effect of deep brain stimulation and lesion of the subthalamic nucleus. Parkinsonism Relat Disord. 2010a;16:338–344. doi: 10.1016/j.parkreldis.2010.02.008. [DOI] [PubMed] [Google Scholar]
- 92.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. 2010b;518:25–40. doi: 10.1002/cne.22207. [DOI] [PubMed] [Google Scholar]
- 93.Shaw VE, Peoples C, Spana S, Ashkan K, Benabid AL, Stone J, Baker GE, Mitrofanis J. Patterns of cell activity in the subthalamic region associated with the neuroprotective action of near-infrared light treatment in MPTP-treated mice. Parkinsons Dis. 2012;2012:296875. doi: 10.1155/2012/296875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Trimmer PA, Schwartz KM, Borland MK, De Taboada L, Streeter J, Oron U. Reduced axonal transport in Parkinson’s disease cybrid neurites is restored by light therapy. Mol Neurodegener. 2009;4:26. doi: 10.1186/1750-1326-4-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Tucker LD, Lu Y, Dong Y, Yang L, Li Y, Zhao N, Zhang Q. Photobiomodulation therapy attenuates hypoxic-ischemic injury in a neonatal rat model. J Mol Neurosci. 2018;65:514–526. doi: 10.1007/s12031-018-1121-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Urrutia PJ, Mena NP, Núñez MT. The interplay between iron accumulation, mitochondrial, dysfunction, and inflammation during the execution step of neurodegenerative disorders. Front Pharmacol. 2014;5:38. doi: 10.3389/fphar.2014.00038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Vos M, Lovisa B, Geens A, Morais VA, Wagnières G, van den Bergh H, Ginggen A, De Strooper B, Tardy Y, Verstreken P. Near-infrared 808 nm light boosts complex IV-dependent respiration and rescues a Parkinson-related pink1 model. PLoS One. 2013;8:e78562. doi: 10.1371/journal.pone.0078562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Wang R, Dong Y, Lu Y, Zhang W, Brann DW, Zhang Q. Photobiomodulation for global cerebral ischemia: targeting mitochondrial dynamics and functions. Mol Neurobiol. 2019;56:1852–1869. doi: 10.1007/s12035-018-1191-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wu Q, Xuan W, Ando T, Xu T, Huang L, Huang YY, Dai T, Dhital S, Sharma SK, Whalen MJ, Hamblin MR. Low-level laser therapy for closed-head traumatic brain injury in mice: effect of different wavelengths. Lasers Surg Med. 2012;44:218–226. doi: 10.1002/lsm.22003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Xu Z, Guo X, Yang Y, Tucker D, Lu Y, Xin N, Zhang G, Yang L, Li J, Du X, Zhang Q, Xu X. Low-level laser irradiation improves depression-like behaviors in mice. Mol Neurobiol. 2017;54:4551–4559. doi: 10.1007/s12035-016-9983-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Xuan W, Agrawal T, Huang L, Gupta GK, Hamblin MR. Low-level laser therapy for traumatic brain injury in mice increases brain derived neurotrophic factor (BDNF) and synaptogenesis. J Biophotonics. 2015;8:502–511. doi: 10.1002/jbio.201400069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Xuan W, Huang L, Hamblin MR. Repeated transcranial low-level laser therapy for traumatic brain injury in mice: Biphasic dose response and long-term treatment outcome. J Biophotonics. 2016;9:1263–1272. doi: 10.1002/jbio.201500336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Xuan W, Vatansever F, Huang L, Hamblin MR. Transcranial low-level laser therapy enhances, memory, and neuroprogenitor cells after traumatic brain injury in mice. J Biomed Opt. 2014;19:108003. doi: 10.1117/1.JBO.19.10.108003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Xuan W, Vatansever F, Huang L, Wu Q, Xuan Y, Dai T, Ando T, Xu T, Huang YY, Hambin MR. Transcranial low-level laser therapy improves neurological performance in traumatic brain injury in mice: effect of treatment repetition regimen. PLoS One. 2013;8:e53454. doi: 10.1371/journal.pone.0053454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Yang L, Dong Y, Wu C, Li Y, Guo Y, Yang B, Zong X, Hamblin MR, Liu TC, Zhang Q. Photobiomodulation preconditioning prevents cognitive impairment in a neonatal rat model of hypoxia-ischemia. J Biophotonics. 2019;12:e201800359. doi: 10.1002/jbio.201800359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Yang L, Tucker D, Dong Y, Wu C, Lu Y, Li Y, Zhang J, Liu TC, Zhang Q. Photobiomodulation therapy promotes neurogenesis by improving post-stroke local microenvironment and stimulating neuroprogenitor cells. Exp Neurol. 2018;299(pt A):86–96. doi: 10.1016/j.expneurol.2017.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Ying R, Liang HL, Whelan HT, Eells JT, Wong-Riley MT. Pretreatment with near-infrared light via light-emitting diode provides added benefit against rotenone-and MPP+-induced neurotoxicity. Brain Res. 2008;1243:167–173. doi: 10.1016/j.brainres.2008.09.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Yip K, Lo S, Leung M, So K, Tang C, Poon D. The effect of low-energy laser irradiation on apoptotic factors following experimentally induced transient cerebral ischemia. Neuroscience. 2011;190:301–306. doi: 10.1016/j.neuroscience.2011.06.022. [DOI] [PubMed] [Google Scholar]
- 109.Yun YC, Jang D, Yoon SB, Kim D, Choi DH, Kwon O, Lee YM, Youn D. Laser acupuncture exerts neuroprotective effects via regulation of Creb, Bdnf, Bcl-2, and Bax gene expressions in the hippocampus. Evid Based Complement Alternat Med. 2017;2017:7181637. doi: 10.1155/2017/7181637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Zhang Q, Zhou C, Hamblin MR, Wu MX. Low-level laser therapy effectively prevents secondary brain injury induced by immediate early responsive gene X-1 deficiency. J Cereb Blood Flow Metab. 2014;34:1391–1401. doi: 10.1038/jcbfm.2014.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Zivin JA, Albers GW, Bornstein N, Chippendale T, Dahlof B, Devlin T, Fisher M, Hacke W, Holt W, Ilic S, Kasner S, Lew R, Nash M, Perez J, Rymer M, Schellinger P, Schneider D, Schwab S, Veltkamp R, Walker M, Streeter J, NeuroThera E, Safety Trial I. Effectiveness and safety of transcranial laser therapy for acute ischemic stroke. Stroke. 2009;40:1359–1364. doi: 10.1161/STROKEAHA.109.547547. [DOI] [PubMed] [Google Scholar]
- 112.Zivin JA, Sehra R, Shoshoo A, Albers GW, Bornstein NM, Dahlof B, Kasner SE, Howard G, Shuaib A, Streeter J, Richieri SP, Hacke W, investigators N, NeuroThera(R) Efficacy and Safety Trial-3 (NEST-3): a double-blind, randomized, sham-controlled, parallel group, multicenter, pivotal study to assess the safety and efficacy of transcranial laser therapy with the NeuroThera(R) Laser System for the treatment of acute ischemic stroke within 24 h of stroke onset. Int J Stroke. 2014;9:950–955. doi: 10.1111/j.1747-4949.2012.00896.x. [DOI] [PubMed] [Google Scholar]