Next Article in Journal
Pyrazolo[4,3-e]tetrazolo[1,5-b][1,2,4]triazine Sulfonamides as an Important Scaffold for Anticancer Drug Discovery—In Vitro and In Silico Evaluation
Next Article in Special Issue
The Role of Insulin-like Growth Factor I in Mechanisms of Resilience and Vulnerability to Sporadic Alzheimer’s Disease
Previous Article in Journal
Anti-β2-glycoprotein I/HLA-DR Antibody and Adverse Obstetric Outcomes
Previous Article in Special Issue
Roles of Rac1-Dependent Intrinsic Forgetting in Memory-Related Brain Disorders: Demon or Angel
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Phototherapy of Alzheimer’s Disease: Photostimulation of Brain Lymphatics during Sleep: A Systematic Review

1
Department of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany
2
Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia
3
Interdisziplinäres Schlafmedizinisches Zentrum, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
4
Department of Nervous Diseases, Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya 2, Building 4, 119435 Moscow, Russia
5
Company “Lazma” for Research and Production Enterprise of Laser Medical Equipment, Kuusinena Str. 11, 123308 Moscow, Russia
6
Department of Complexity Science, Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(13), 10946; https://doi.org/10.3390/ijms241310946
Submission received: 20 April 2023 / Revised: 18 June 2023 / Accepted: 26 June 2023 / Published: 30 June 2023
(This article belongs to the Special Issue Recent Advances in Alzheimer’s Disease)

Abstract

:
The global number of people with Alzheimer’s disease (AD) doubles every 5 years. It has been established that unless an effective treatment for AD is found, the incidence of AD will triple by 2060. However, pharmacological therapies for AD have failed to show effectiveness and safety. Therefore, the search for alternative methods for treating AD is an urgent problem in medicine. The lymphatic drainage and removal system of the brain (LDRSB) plays an important role in resistance to the progression of AD. The development of methods for augmentation of the LDRSB functions may contribute to progress in AD therapy. Photobiomodulation (PBM) is considered to be a non-pharmacological and safe approach for AD therapy. Here, we highlight the most recent and relevant studies of PBM for AD. We focus on emerging evidence that indicates the potential benefits of PBM during sleep for modulation of natural activation of the LDRSB at nighttime, providing effective removal of metabolites, including amyloid-β, from the brain, leading to reduced progression of AD. Our review creates a new niche in the therapy of brain diseases during sleep and sheds light on the development of smart sleep technologies for neurodegenerative diseases.

1. Introduction

Alzheimer’s disease (AD) is a brain pathology that is accompanied by progressive memory loss. From 1990 to 2019, the global number of people with AD increased by 117% [1,2,3]. The number of people over 65 years old with AD doubles every 5 years [4]. It is estimated that if an effective therapy of AD is not found, the incidence of AD will triple by 2060 [4]. The large number of clinical studies have failed to show effective pharmacological treatment of AD [5,6,7,8]. Indeed, ENGAGE phase 3 randomized clinical trials of aducanumab obtained in 3285 patients with AD report different consequences associated with therapy, including edema (35.2%), headache (46.6%), confusion (14.6%), dizziness (10.7%), nausea (7.8%), microhemorrhage (19.1%), and superficial siderosis (14.7%) [8].
Photobiomodulation (PBM) is a non-pharmacological therapeutic approach based on the use of red or near-infrared light that has shown very promising results in the treatment of AD in pilot clinical and animal studies [9,10,11,12,13,14,15,16,17,18,19,20]. PBM has been recognized as safe by the U.S. Food and Drug Administration (FDA) because PBM is a non-invasive method without any major side effects. Many publications suggest that the mechanisms of PBM underlie the increase in metabolism and microcirculation of brain tissue, neuroprotection, and reduction in oxidative stress and inflammation [12,17,18,20]. However, it was recently discovered that PBM effectively stimulates lymphatic removal of wastes and toxins, including amyloid-β (Aβ), from the brain [21,22,23,24,25,26,27,28]. Furthermore, PBM affects lymphatic delivery of drugs and nanocarriers to the brain [29,30,31,32].
Recently, the phenomenon of activation of the lymphatic drainage and removal system of the brain (LDRSB) during deep sleep was discovered [21,33,34]. The relationship between the LDRSB and sleep was first described by Xie et al. [33]. This research group compared the brain influx of tracers in mice during sleep, awake, and under anesthesia. The authors clearly demonstrated a 95% reduction in Aβ removal from the brain during wakefulness and the activation of this process during deep sleep due to a 60% increase in the interstitial fluid (ISF) space. Later, Fultz et al. reported that deep sleep is accompanied by high oscillation of cerebral spinal fluid (CSF) in the human brain [34]. There is a hypothesis that the LDRSB is the main driving factor for the removal of metabolic wastes from the sleeping brain [35]. According to this hypothesis, during deep sleep, the size of perivascular spaces and the volume of ISF increase, promoting the removal of metabolites from brain tissue. During wakefulness, the size of the perivascular spaces decreases and the exchange between fluids and brain tissue is suppressed. The similar changes are observed during the development of AD, which is associated with suppression of the LDRSB functions, leading to accumulation of Aβ in the brain tissue.
A few years ago, the idea of using PBM during sleep was proposed as a promising new direction in the treatment of AD [21,35,36,37,38]. Encouraged by this idea, in this review, we discuss how PBM of LDRSB during sleep could be used as a new innovative treatment tool in the therapy of AD. The first session provides information about a strong correlation between the LDRSB and AD pathology. Emerging evidence suggests that the LDRSB is a promising therapeutic target for AD and the development of methods for stimulation of the LDRSB functions could ameliorate cognitive dysfunction in AD and may contribute to progress in the therapy of AD. The second session is devoted to the benefits of PBM for AD as a non-invasive, non-pharmacological, and safe therapeutic approach. Data from animal and clinical studies show that PBM accelerates clearance of Aβ and tau from the brain and could ameliorate cognitive dysfunction in subjects with AD. The third session is focused on PBM during sleep as a new trend in the therapy of AD. The LDRSB and removal of Aβ from the brain is activated at night. Data from animal studies clearly show that nighttime application of PBM can result in a better therapeutic effect in AD mice than PBM during the day. At present, no commercial device for monitoring PBM during sleep is available. We present a concept of such a device and strategy for the development of sleep-PBM technologies to augment the LDRSB and clearance of toxins from the sleeping brain. We suggest that PBM during sleep can be a novel approach to therapy of AD and also for treatment of other brain pathologies associated with impaired LDRSB, including Parkinson’s disease, intracranial hemorrhages, brain trauma, and glioblastoma.

2. Methods

2.1. Literature Search

The research topic of this review was PBM of AD with a focus on PBM-mediated augmentation of the LDRSB functions during sleep as a strategy for targeting AD.
For this research topic, to capture as many relevant citations as possible, the keywords referring to PBM of interest (photobiomodulation and Alzheimer’s diseases) were combined with the following keywords referring to the LDRSB: meningeal lymphatics, sleep, lymphatic drainage, and clearance. Accordingly, the search engine of the U.S. National Institutes of Health (PubMed) and on the clinicaltrials.gov website (ClinicalTrials.gov is a database of privately and publicly funded clinical studies conducted around the world) was used to retrieve the relevant studies with the following search terms: “photobiomodulation + Alzheimer’s diseases”; “photobiomodulation + the meningeal lymphatics”; “photobiomodulation + sleep”; “photobiomodulation + lymphatic drainage and clearance”; “Alzheimer’s diseases and sleep”; “Alzheimer’s diseases + the meningeal lymphatics”; “Alzheimer’s diseases + lymphatic drainage and clearance”; “the meningeal lymphatics + sleep”; “lymphatic drainage and clearance + sleep”. These search terms retrieved a total of 1471 hits (82 + 4 + 40 + 3 + 1250 + 30 + 24 + 25 + 13 studies). Given the limited information about PBM for healthy volunteers and patients with AD, this review includes studies from the period 2008–2023 and focuses on those investigations into the clinical benefit of using PBM to treat AD.

2.2. Inclusion and Exclusion Criteria

Only experimental and clinical trials as well as case reports assessing the effects and mechanisms of PBM for AD were considered. Meta-analyses, conference abstracts, duplicates, or non-English papers were excluded. Furthermore, papers discussing sleep disturbances during the development of AD, PBM used for other brain diseases or for improvement of sleep quality, and other types of therapies for AD were excluded. The total number of papers for further screening was thereby reduced to 133, addressing the LDRSB as a therapeutic target for AD, PBM for AD in basic and clinical studies, and PBM of LDRSB during sleep as a new trend in the therapy of AD.

3. Results

We collected 1471 articles from the PubMed database and the clinicaltrials.gov website, including reference lists from previous relevant reviews. After removing duplicates, 973 items remained. We identified 301 records via title screening, and the abstracts of these records were screened more comprehensively. A total of 173 articles were selected for a full-text review, and 133 records were included in the review. The details of excluded trials are shown in Figure 1.

3.1. LDRSB as a Therapeutic Target for AD

A growing body of evidence suggests that the meningeal lymphatic vessels (MLVs) as a part of the LDRSB are pathways for the clearance of cell debris, toxic molecules, and wastes from the central nervous system (CNS) [39,40,41,42,43,44,45,46]. It has recently been discovered as an important role of MLVs in the removal of amyloid-β (Aβ) from the brain tissue [38,39]. The accumulation in the brain of Aβ species and plaques is considered to be a biomarker of AD, which precedes neuronal dysfunction and clinical manifestations of AD by up to 20–30 years [47,48,49]. In 1988, Joachim et al. identified Aβ depositions in the meninges of patients with AD [50]. Later, in 2018, Da Mesquita et al. clearly demonstrated that MLVs are tunnels for the clearance of Aβ from the brain [39]. This group showed that photo-injury of MLVs in transgenic mice with AD reduces clearance of Aβ from the mouse brain. The mechanisms underlying Aβ deposition in the brain and the meninges of subjects with AD are still not fully understood. The age-related decrease in recirculation of brain fluids, including the cerebral spinal fluid (CSF) and the interstitial fluid (ISF), is thought to be partly responsible for the accumulation of Aβ in the brain tissue [51,52]. The decline in functions of MLVs with age is associated with reduced excretion of Aβ from brain parenchyma that also contributes to neuronal loss and behavioral changes [52,53,54,55,56]. Therefore, augmentation of functions of MLVs is considered to be a promising therapeutic target for preventing or delaying AD [39,40,57].
The importance of MLVs’ function in the treatment of AD has been demonstrated with immunotherapy [58,59]. This method involves the use of anti-Aβ monoclonal antibodies to reduce Aβ accumulation and to increase Aβ clearance via activation of microglial phagocytosis of this protein. The effects of Aβ immunotherapy are variable in different clinical studies [5,6]. The anti-Aβ monoclonal drug aducanumab was recently approved by the FDA for therapy of patients with AD. However, the approval has sparked a contentious debate over whether the drug is effective [7]. Furthermore, the EMERGE and ENGAGE phase 3 randomized clinical trials of aducanumab obtained in 3285 participants report different symptoms associated with therapy, including edema (35.2%), headache (46.6%), confusion (14.6%), dizziness (10.7%), nausea (7.8%), and microhemorrhage and superficial siderosis in 197 participants (19.1%) and 151 participants (14.7%), respectively [8]. Thus, the use of aducanumab has not been reported as safe.
The latest research has provided a new insight into Aβ immunotherapy of AD via our better understanding of MLVs in regulation of the brain’s immune system [58]. Da Mesquita et al. studied how photo-injury of MLVs and Aβ immunotherapy (m-aducanumab and mAb158, anti-Aβ protofibril monoclonal antibody) can influence AD progression in 5xFAD mice. They demonstrated that Aβ immunotherapy was not effective in reducing Aβ deposits in the brains of mice with the injured MLVs compared with the intact MLVs [58]. Conversely, stimulation of MLVs by intracisternal injection of vascular endothelial growth factor C (VEFG-C), causing lymphangiogenesis, provides for Aβ clearance and improves Aβ immunotherapy. Wen et al., using the APP/PS1 mouse AD model, also show that VEFG-C injection reduces the accumulation of Aβ and alleviates the cognitive deficit [60]. The number of CD8+ “killer” (cytotoxic) and CD4+ “helper” T cells in the meninges significantly increases in old 5xFAD mice with a decrease in MLV coverage that can be improved by VEGF-C via restoring the MLVs’ function [58,59].
These data highlight the potential of VEGF-mediated restoration of the MLVs’ function of improving neuroinflammation and cognitive status in AD subjects. Thus, the effectiveness of Aβ immunotherapy depends on the functional status of MLVs, which is significantly reduced in AD subjects and with age. This can partly explain differences in the reported efficacy of Aβ immunotherapy in humans. These results also provide a new opportunity to further study the effectiveness of Aβ immunotherapy combined with VEGF-C for AD therapy using the different AD models.
In addition, DSCR1, Down syndrome critical region gene 1, regulates the MLV functions in AD [61]. The over-expression of DCSR1 is associated with an increase in clearance of Aβ via MLVs that contribute to an improvement of cognitive function in AD [29]. Wu et al. reveal that borneol, a bicyclic monoterpene belonging to the class of camphene, which is sourced from Blumea balsamifera, improves lymphatic clearance of Aβ as well as other macromolecular polymers in the weight range of 2–45 KD [62].
In summary, current evidence shows a strong relationship between LDRSB and AD pathology. These observations suggest that targeting LDRSB may represent a promising therapeutic strategy for AD. The enhancement of MLV drainage by VEGF-C, DSCR1, and Aβ immunotherapy to accelerate Aβ clearance from the brain could ameliorate cognitive dysfunction in AD [58,60,61]. However, these lymphatic drainage improvement strategies are too far off being introduced in routine clinical practice due to the difficulties of intra-cisterna magna delivery of VEGF-C or high off-targets and low controllability. Therefore, new ideas for stimulation of transport capacity of LDRSB, including clearance of Aβ and tau protein from the brain, are strongly required.

3.2. PBM for AD in Basic and Clinical Studies

3.2.1. PBM for AD In Vitro Studies

There are a number of studies showing PDT-mediated reduction in Aβ(42) aggregation in different cell lines. Sommer et al. demonstrated that irradiation with moderately intense 670 nm laser light reduces Aβ aggregates in human neuroblastoma cells [63]. Yang et al. reported that a 632.8 nm laser was capable of suppressing cellular pathways of Aβ-induced oxidative stress and inflammatory responses in primary rat astrocytes [64]. Liang et al. found that a 623.8 nm laser attenuates Aβ-induced cell apoptosis through the Akt/GSK3β/β-catenin pathway [65]. They showed that PBM activates Akt, which interacts with GSK3β and phosphorylates it on Ser9 in the presence of Aβ(25–35), leading to the inhibition of GSK3β and promoting cell survival upon treatment with Aβ(25–35). Zhang et al. discovered reduction in Aβ(25–35)-induced cell apoptosis by a 632.8 nm laser through promoting Akt-dependent Yes-associated protein cytoplasmic translocation [66]. The downregulation of brain-derived neurotrophic factor (BDNF) in the hippocampus is a consequence of the progression of AD and Aβ-induced neurotoxicity. There is a hypothesis that BDNF plays an important role in neuronal survival and contributes to rescuing dendrite atrophy and cell loss in AD. Meng et al. clearly showed that a 632.8 nm laser protects Aβ-treated hippocampal neurons and cultured APP/PS1 mouse hippocampal neurons via upregulation of BDNF in both [67]. Zinchenko et al. investigated 1267 nm laser-induced extravasation of Aβ (1–42) through the model of the blood–brain barrier as a mechanism of PBM for AD [25].

3.2.2. PBM for AD in Animal Studies

In 2011, De Taboada et al. presented promising results demonstrating the effective transcranial PBM therapy of Aβ-precursor transgenic mice by using a 810 nm laser (3 sessions per week for 6 months) [68]. They found that Aβ plaques and inflammatory markers were reduced significantly in the brain tissue in a laser dose-related manner (4.8 J/cm2−48 J/cm2 on the skull and 1.2 J/cm2−12 J/cm2 on the cortex). These therapeutic effects attenuated the progression of AD, leading to an improvement of cognitive functions in mice. The dose-dependent reduction in Aβ plaques in the brain of TASTPM transgenic mice was also shown by Grillo [69]. Purushothuman et al. demonstrated the beneficial effect of transcranial PBM (670 nm LEDs, 4 J/cm2, 20 min per session for 4 weeks) in two mouse models of AD, such as the K369I tau transgenic model and the APPswe/PSEN1dE9 transgenic model [70,71]. In the tau model, PBM restored the expression of mitochondrial cytochrome c oxidase in neurons as well as decreased markers of neurofibrillary tangles and oxidative stress, including 4-hydroxynonenal and 8-hydroxy-2-deoxyguanosine, to the normal values in the neocortex and the hippocampus. In both tau and Aβ models, PBM reduced the level of tau protein and Aβ plaques in the brain tissue. Using the 5XFAD model of AD, Farfara et al. treated mice by PBM (810 nm, 1 J/cm2 applied to the tibia via a small incision of the skin) for 4 weeks with 10-day intervals starting at the age of 4 months. The results revealed a 68% decrease in Aβ plaques in the brain, which was associated with better performance in the object recognition and fear conditioning tests in the PBM group compared with the sham group [72]. Oron et al. discussed these results in their review as the new strategy of the PBM therapy of AD via the secondary PBM effects through photostimulation of proliferation of mesenchymal stem cells [73]. Lu et al. showed the therapeutic effects of PBM (810 nm, 2 min per session for 5 days) in rats with the injected model of AD [74]. They showed that PBM inhibited neurodegeneration in the hippocampus, leading to reduction in Aβ accumulation in the brain and improvement of recognition memory. They also found positive PBM effects on the mitochondrial functions, including enhancement of mitochondrial antioxidant expression, the level of cytochrome c oxidase activity and ATP, and suppression of apoptosis, reactive gliosis, and inflammation. De Luz Eltchechem et al. also demonstrated a significant reduction in Aβ plaques in the hippocampus and an improvement of cognitive, behavioral, and motor skills in rats with the injected model of AD after PBM therapy (627 nm, 7 J/cm2, 1.5 min for session for 3 weeks) [75]. Brivelet et al. reported similar therapeutic effects of PBM in Swiss mice with the injected model of AD after PBM (LED, 625 nm, 10 Hz, 8.4 J/cm2, 10 min for 7 days) [76]. Cho et al. used the transcranial PBM treatment (610 nm, 1.7 mW/cm2, 2.0 J/cm2, 20 min per session three times per week for 14 weeks) of 5XFAD mice and found that PBM in early stages of AD reduced Aβ accumulation in the brain and neuronal loss and alleviated cognitive dysfunction [77]. Zhang et al. presented the results of transcranial PBM (632.8 nm, 2 J/cm2, 10 min per session for 30 days) in double transgenic mice (APPswe/PSENdE9) and showed that PBM reduced Aβ levels in the brain and improved spatial learning and memory deficits [78]. Zhinchenko et al. reported the optimal 1267 nm laser dose (32 J/cm2) for reduction in Aβ accumulation in the brain and restoration of recognition memory in mice with the injected model of AD after 10 days of PBM therapy [27].

3.2.3. PBM for AD in Clinical Studies

Several studies demonstrate that PBM improves memory, attention, sleep quality, and emotional state in healthy humans. Table 1 presents data on the parameters of PBM used in healthy volunteers involved in studies investigating the PBM effects on mental functions and memory. There is a hypothesis that the mechanisms of PBM effects on a healthy brain can be an increase in cerebral perfusion, brain energy, and oxygen metabolism, which are necessary to maintain normal cognitive function [12,17,20,79]. Barret and Gonzalez-Lima used a 1064 nm laser in a placebo-controlled study and reported the beneficial PBM effects on executive functions, attention, and memory in healthy volunteers 2 weeks after PBM [80]. Blanco et al. demonstrated a superior Wisconsin Card Sorting Test performance after PBM (1064 nm) compared with the control group [81]. Chan et al. observed an improvement of reaction time evaluated by the Eriksen flanker test and mental flexibility assessed by the category fluency test in older (over 60 years) healthy volunteers after a single session of PBM (a mix of 633 nm and 870 nm) [82]. Wu et al. investigated the PBM effects (830 nm) on the pattern of electrical brain activity using electroencephalogram (EEG) in forty normal healthy male subjects [83]. They observed for 10 min after PBM an increase in the power of alpha rhythms and theta activities in the occipital, parietal, and temporal areas (mainly in the posterior head regions). The authors suggested that the PBM effects on the EEG changes are comparable to those in meditation. Jahan at el. suggested that transcranial PBM (850 nm) on the right prefrontal cortex changed the brainwaves (decreases a delta power) and had a beneficial effect on cognitive performance as well as improving attention and alertness in 30 healthy young adult participants [84].
The clinical studies of the PBM effects for AD are still limited [9,10,11,19,85]. Table 2 summarizes the results of recent clinical studies on the therapeutic effects of PBM in patients with AD. Salmarche et al. investigated five patients with mild to moderately severe dementia or possible AD and demonstrated that home transcranial–intranasal PBM (810 nm) therapy for 12 weeks can improve cognition and functional abilities for daily living [86]. Chao et al. also reported that home transcranial PBM (801 nm) for 12 weeks significantly improved cognitive and behavioral functions and increased cerebral microcirculation in four patients with mild-to-moderate dementia [87]. Later, the same group demonstrated a decrease in the CSF levels of Aβ42, tau, and neurofilament light chain in seven patients with AD 16 weeks after home transcranial–intranasal PBM (810 nm) therapy [88]. The reduction in the CSF levels of Aβ42 was also observed after therapy of patients with AD, using flickering lights at gamma frequency for 8 weeks [89]. Zomorrodi et al. presented similar results using home PBM (810 nm) treatment of one patient with moderate AD [90]. Salehpour at el. reported positive effects of a transcranial–intranasal PBM treatment (a mix of 635 nm and 810 nm) of a single case on cognitive enhancement and reversal of olfactory dysfunction [91]. Maksimovich used low-energy lasers in the visible region of the spectrum for trans-catheter treatment of AD and showed that PBM reduces the effects of dyscirculatory angiopathy of AD and improves cerebral microcirculation and metabolism, leading to dementia regression and cognitive impairment reduction [15]. However, in another study on 11 patients with dementia, no significant differences were found between the PBM (1060–1080 nm and the sham groups, probably due to the small number of patients in the PBM group (n = 6) and in the sham group (n = 5) [92]. However, the authors reported a trend of improvement in executive functioning, praxis memory, visual attention, and task switching in participants receiving PBM for 4 weeks. Later, the same group repeated the clinical studies of the PBM therapy for AD on a larger group of 47 patients and showed that PBM has positive cognitive, executive, and mood changes in participants with dementia that improve their quality of life [93]. Further analysis of these human data revealed no gender difference in the effectiveness of the PBM therapy of dementia [93]. Horner et al. analyzed the effectiveness of the transcranial PBM (810 nm) therapy of one patient with AD and type 2 diabetes and showed an improvement of cognitive function and restoration of mitochondrial function after 10 weeks of treatment [94].

3.2.4. PBM for Other Brain Diseases

Parkinson’s disease (PD) is a progressive brain disease that leads to degeneration of neurons and affects the control of body movements. PBM for PD has received much attention as one of the most promising methods to treat this neurodegeneration in the brain [10,18,79,95]. Several clinical in vitro (the blood samples of 10 PD patients) and in vivo (n = 70 PD patients) studies reported the therapeutic effects of PBM (632.8 nm; 500 mW/cm2) on enzyme activity of monoamine oxidase B (MAO B), Cu/Zn-superoxide dismutase (Cu/Zn-SOD), Mn-SOD, and catalase in blood from PD patients [96,97]. PBM consisted of five daily 20 min sessions of blood irradiation (intravenous irradiation in vivo), and the blood was analyzed on day 3 after the last irradiation session. PBM significantly improved neurological status of PD patients assessed by Unified Parkinson’s Disease Rating Scale. The improvement was accompanied by normalization of MAO B and Cu/Zn-SOD activities. The effects of PBM on blood enzymes were much stronger after in vivo application of PBM than after in vitro blood irradiation.
Reduced axonal transport is one of the mechanisms underlying the progressive loss of dopaminergic nerve terminals in PD. The ATP synthesis in mitochondria supports axonal transport and contributes to the survival of neuronal cells. In PD subjects, mitochondria in brain tissue are metabolically and functionally suppressed. Trimmer et al. demonstrated that PBM (810 nm, 50 mW/cm2 for 40 s) improves axonal transport in model human dopaminergic neuronal cells [98]. They measured the velocity of movement of labeled mitochondria in human transmitochondrial cybrid neuronal cells bearing mitochondrial DNA from patients with PD and healthy volunteers. They found that the velocity of mitochondrial movement in neuronal cells was significantly reduced in the PD group vs. the control group. However, 2 h after PBM, the velocity of mitochondrial movement in PD returned to normal values, while it was unaltered by PBM in the control groups. The authors suggest that PBM can be seen as a novel strategy in the therapy of PD.
Amyotrophic lateral sclerosis (ALS) is another neurodegenerative disorder that leads to gradual muscle paralysis, resulting in respiratory failure and death within 1–5 years after onset of clinical signs. The mitochondrial dysfunction is involved in the pathogenesis of ALS. Moreover, oxidative stress is also thought to play an important role in ALS.
Moges et al. examined the effects of PBM (810 nm, 140 mW, 1.4 cm2 for 120 s, for three consecutive days every week for 51 days) on the survival of motor neurons in the G93A SOD1 transgenic mice (ALS model) [99]. The PBM was applied in three places: the primary motor cortex, the cervical enlargement of the spinal cord, and the lumbar enlargement of the spinal cord. Despite the fact that motor function in G93A SOD1 mice was improved after PBM, however, PBM was ineffective in altering progression of ALS. Nevertheless, these findings have potential implications for the conceptual use of PBM to treat other neurodegenerative diseases associated with mitochondrial dysfunction.
A PBM has been shown to be effective in therapy for other brain diseases as well. Muili et al. found that PBM (670 nm, 28 mW/cm2 for 3 min, once daily irradiation) ameliorates symptoms in a mouse model of multiple sclerosis [100]. Leisman et al. reported improvement of autism spectrum disorder in children and adolescents of 5–17 years of age after PBM treatment (635 nm, 15 mW for 5 min, a 4-week course of PBM) [101]. PBM can also prevent hypoxic–ischemic brain injuries by maintaining mitochondrial function, decreasing oxidative stress and inhibiting neuronal apoptosis. Indeed, Tucker et al. found that PBM (808 nm, 25 mW/cm2 for 2 min, 7 day treatment) prevents ischemic injury to neurons after cerebral ischemia caused by cardiac arrest [102]. Yang et al. using a model of neonatal hypoxic–ischemic encephalopathy in rats show that PBM (808 nm, 100 mW/cm2 for 2 min) improves ischemic-induced brain injury [103]. Thus, the results discussed above clearly demonstrate that PBM can significantly attenuate cognitive impairment, neuron loss, and dendritic and synaptic injury, and restore ischemic-induced mitochondrial dynamics in various brain diseases.

3.3. PBM of the LDRSB during Sleep as a New Trend in the Therapy of AD

Sleep is considered to be a novel biomarker and promising therapeutic target for AD and dementia [35,36]. Emerging evidence suggests that sleep disorder is an independent risk factor of AD and the analysis of sleep quality can be an informative approach to screen for AD [104,105,106,107,108,109,110,111,112]. It is well known that AD has two interrelated characteristics, such as poor sleep and increased Aβ deposition in the brain [107,109]. In a recent long-term study on a large group of 8000 volunteers aged 50–60 years, it has been discovered that chronic sleep deficit (less than 6 h) for 25 years can lead to the development of dementia [113]. However, even one night without sleep is associated with a rise in Aβ level in the brain of young and healthy people [114,115].
There is a growing body of evidence suggesting that sleep is accompanied by activation of the LDRSB, leading to Aβ clearance from the brain [21,33,34,35,36,116,117,118]. Indeed, the CSF level of Aβ42 is increased at night before sleep and is decreased in the early morning after sleep in both humans and animals [114]. However, not all sleep is equivalent to activation of the LDRSB. Recent studies clearly demonstrate that only slow-wave activity (SWA, 0–4 Hz) during non-rapid eye movement (NREM) sleep is optimal for the LDRSB [119]. In 2013, Xie et al. reported that the LDRSB function is highest during NREM sleep and lowest during wakefulness [33]. This group found a 95% reduction in Aβ removal from the brain in awake mice but a significant activation of Aβ clearance during NREM sleep associated with a 60% increase in the interstitial space [33]. Later, Benveniste et al. confirmed that sleep is accompanied by the expansion of the interstitial space, probably due to an increase in the flow of ISF for optimization of removal of metabolites [120]. Figure 2a illustrates our experimental results, demonstrating an activation of drainage of the mouse brain along the perivascular space during deep sleep. The SWA disturbances reflect the AD pathology in both humans and animals [109,121,122,123,124]. In humans, impaired memory consolidation associated with AD progression is strongly correlated with decreased SWA time [125,126,127]. The mechanisms underlying the Aβ-mediated reduction in the SWA dynamic and a decrease in the LDRSB functions remain unknown. The Aβ accumulation in the brain leads to hyperactivity of the cortical neurons and synaptic abnormalities that can be a possible mechanism underlying the SWA deficit in AD subjects [128,129,130,131,132,133]. The insufficient sleep is related to an increase in the interstitial noradrenaline level, leading to a reduction in the astrocytic volume and the cerebral vasoconstriction that can be another mechanism of a suppression of the LDRSB function and removal of wastes from the brain [33,134,135]. The horizontal body posture during sleep plays an important role in the LDRSB function, contributing to a better Aβ clearance from the brain [136].

4. Discussion

The global number of elderly people with AD is progressively increasing worldwide. However, pharmacological therapies for AD have failed to show effectiveness and safety [5,6,7,8]. Therefore, the development of alternative non-pharmacological therapeutic methods for AD is highly relevant for medicine. This review is focused on PBM as a non-invasive, non-pharmacological, safe, and promising approach for treating AD and other brain pathologies. Since the treatment of AD is incurable, therapy aimed at reducing the progression of the disease should be applied throughout the life of patients. In this respect, PBM technologies are the most promising, since they are already widely used in the clinical field for the treatment of various brain diseases [137,138,139,140].
The rapidly increasing number of studies demonstrates the importance of the LDRSB in resistance to AD pathology and suggests that the development of methods for augmentation of the LDRSB functions may contribute to strong progress in the therapy of AD [21,27,35,36,39]. Traditionally, it was thought that the mechanisms of therapeutic effects of PBM underlie the increase in metabolism and microcirculation of brain tissue, neuroprotection, and reduction in oxidative stress and inflammation [12,17,18,20]. However, it was recently discovered that PBM also can augment the LDRSB functions, providing effective removal of metabolites, including Aβ, from the brain, leading to reduced AD progression [21,22,23,24,25,26,27,28,29,30,31,32,33,34,35].
Since the LDRSB and removal of Aβ from the brain are activated at night, it is logical to expect that PBM for AD during sleep will be more effective than PBM during day. Indeed, the pilot animal data clearly demonstrate that the night-course of PBM has better therapeutic effects in AD mice compared with the day-course of PBM [21] (Figure 2b–d). These pioneering results open up new perspectives for PBM therapy of AD during sleep, which is of growing interest to researchers [35,36,37,38].
Typically, in experimental and clinical studies, PBM is performed in awake subjects [9,10,11,12,13,14,15,16,17,18,19,20,68,69,70,71,72,73,74,75,76,77,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94]. In our recent review, we suggested that there are no commercial devices for simultaneous PBM and sleep monitoring, which significantly limits the clinical investigations [36]. The development of sleep-PBM technologies for stimulation of the LDRSB during deep sleep is urgently needed and could become a breakthrough step in the progress of AD therapy in humans (Figure 3).
The mechanism by which PBM affects the lymphatics has not been sufficiently explored. Data from animals demonstrate that PBM stimulates the drainage and cleaning functions of the lymphatics via PBM-mediated activation of lymphatic contractility [29]. Li et al. also report that PBM increases lymphangion contraction [45]. Nitric oxide (NO) plays an important role in regulation of relaxation of the lymphatic vessels after their contraction [141,142]. The contraction cycle of the lymphatic vessels is a combination of mechanical and conducted electrophysiological events. The relaxation phase is a component of locally generated NO in the lymphatic endothelial cells (LECs) in response to transiently elevated shear forces. The NO is a vasodilator that acts via stimulation of soluble guanylate cyclase to form cyclic-GMP, which activates protein kinase G, causing the opening of calcium-activated potassium channels and reuptake of Ca2+. The decrease in the concentration of Ca2+ prevents myosin light-chain kinase from phosphorylating the myosin molecule, leading to relaxation of the lymphatic vessels after their contraction. Based on these facts, Li et al. studied the NO production in in vitro experiments on the mesenteric LECs without and after PBM. These results revealed a significant increase in the 24 h accumulation of NO in the cell culture medium after PBM compared with the accumulation of NO produced by LECs without PBM [45]. These results suggest that PBM stimulates the LDRSB functions by influencing the mechanisms of regulation of lymphatic pumping via regulation of the contraction cycle and phase of relaxation of the lymphatic vessels by activation of the intracellular NO production. NO-induced relaxation allows for diastolic filling of the lymphatic vessels and thus prepares them for their next contraction [143,144,145].
How PBM can improve cognitive function in subjects with AD remains poorly understood. Emerging evidence indicates the important homeostatic and pathophysiological roles of brain lymphatics in the progression of AD [21,39,40,146,147]. Indeed, genetic and pharmacological disruption of MLVs results in less drainage of CSF and ISF to the cervical lymph nodes [31,39]. Such disruption also results in cognitive impairment and behavioral alterations [39]. Increasing lymphangiogenesis of MLVs via administration of the vascular endothelial growth factor improves the drainage of macromolecules to the cervical lymph nodes of elderly mice [58,148,149,150,151]. Finally, disruption of MLVs worsens mouse models of AD. Collectively, these findings suggest that dysfunction of the LDRSB might provide an important contribution to age-related cognitive decline and neurodegenerative disease [146,147].
We assume that the mechanisms of the PBM therapy of AD during sleep might be different from those during wakefulness, which requires careful and detailed research [35,36,37,38]. Further animal and clinical studies of night PBM of AD will shed light on optimal parameters of PBM, including light wavelength, power density and intensity, treatment duration, and comfort light application on the head in sleeping subjects that are critical for the successful PBM therapy of AD.
Since there are few studies on the use of PBM for healthy volunteers and patients with AD, this limits information about reproducibility of reported protocols, parameters, intensity, duration, and intervals of PBM in humans as well as about an optimal optical window and mechanisms responsible for PBM-mediated therapy of AD. There is a wide range of parameters for the transcranial–intranasal PBM in healthy subjects and AD patients (Table 1 and Table 2). Devices with LEDs both in red light (633–635 nm) and in near-infrared wavelength (800–900 nm and 1064–1080 nm) are most popular in PBM for AD. The benefit of an optical window is that the light wavelengths (600–1200 nm) can effectively penetrate the skin and the skull. Tedford et al. estimated that the wavelength of 808 nm has a maximal penetration depth [152]. Wang and Li proposed that the wavelengths 660 nm and 810 nm are optimal for transcranial PBM [153]. In our recent review, we discussed in detail the depth of penetration of different light wavelengths into the head and brain tissue in humans [154]. The energy density ranges from 10 to 60 J/cm2 and pulsing of light is at 10–40 Hz. The duration of PBM is rather small in healthy subjects (typically, single irradiation lasting 2.5–10 min) but is long enough in AD patients (usually 20–25 min per session and for 4–16 weeks but can be from 1 year to 12 years).
A PBM has been recognized as safe by the U.S. Food and Drug Administration (FDA). Red-light therapy is generally considered safe, even though researchers are not exactly sure how and why it works. Maximal permissible exposures of skin surface as well as other conditions for safe use of light radiation are regulated with relevant standards, e.g., ANSI Z136 [155,156]. Even though this type of treatment is generally very safe, some negative effects may occur [157,158,159,160]. Mild visual side effects are not unusual but disappear promptly. Therefore, determining the appropriate dose and timing of light is essential in order to diminish the occurrence of such side effects. However, there are no set rules on how much light to use for the therapy of AD. Nonetheless, the use of PBM for people with drug-resistant non-seasonal depression can result in a hyperactive state and an increase in blood pressure [161]. In these rare cases, light therapy must be reduced or stopped and the condition adequately treated.
We want to emphasize that given the complex pathophysiologic nature of AD, a single therapeutic intervention is unlikely to be a satisfactory response, and that a combination of various interventions is probably critical.
Noninvasive brain stimulation (NIBS) is another strategy for AD treatment [162,163,164,165,166,167,168,169,170,171]. Transcranial magnetic stimulation (TMS) is the most widely used technique for NIBS. TMS consists of delivering short (up to 300 µs) magnetic pulses of high intensity (up to 2.5 Teslas) by a copper-wired coil applied to the scalp. The induced electric field in the brain triggers action potentials, and alters neural activity.
In this respect, TMS has been successfully used in investigating molecular and neurotransmitter dysfunctions, characterizing AD pathology and highlighting biomarkers for the differential diagnosis between AD and other forms of dementia [172,173]. In a transgenic animal model, 14 days of TMS reduces Aβ deposition in the hippocampus and prevents spatial memory loss [161]. Remarkably, such neuroprotective effects are associated with increased CSF-ISF exchange dynamics, as observed by increased CSF glymphatic influx and meningeal lymphatic outflow into the deep cervical lymph nodes, resulting in reduced gliosis and increased neuronal activation in the hippocampus [174]. It is worth noting that a single session of TMS using continuous theta burst stimulation can restore the sleep deprivation-induced reduction in the expression of polarized AQP4 [175] and increase meningeal lymphatic dilation [176], suggesting that TMS increases clearance function by acting on both the glymphatic and meningeal lymphatic pathways. However, the major limitation across TMS lies in the difficulty of comparing its efficacy due to the high variability observed across study protocols [163,165]. Thus, current results on the use of NIBS in AD are encouraging, but there is still a need to better characterize the long-term benefits of stimulation [165].

5. Conclusions

In this review, we highlighted the most recent and relevant experimental and clinical studies of the effectiveness of PBM for the therapy of AD and other brain diseases. Increasing evidence for the important role of the LDRSB in resistance to AD has provided new insights into AD progression and laid the foundation for the development of innovative approaches for stimulation of the LDRSB functions. We suggested that PBM might be a promising technology to target the LDRSB. Since the LDRSB and removal of Aβ from the brain are activated at night, we proposed using PBM during sleep as a new trend in AD therapy. We also presented a concept for a device and strategy for the development of sleep-PBM technologies to augment the LDRSB and clearance of toxins from the sleeping brain. Many brain diseases are associated with impaired LDRSB and suppression of removal of toxins from the brain, including PD, intracranial hemorrhages, brain trauma, and glioblastoma [41,42,43,44,45,177]. There are experimental and clinical data suggesting the effective therapy of these brain diseases by PBM [29,31,45,137,138,139,140,178,179]. Therefore, nighttime therapy of brain diseases using PBM and other approaches during deep sleep might herald a new era in neurorehabilitation medicine.

Author Contributions

O.S.-G., J.K., T.P., M.P. and V.S. initiated and supervised this review article; O.S.-G. wrote the Abstract, Introduction, and Conclusions; O.S.-G., A.T., I.B. and M.T. prepared the section “LDRSB as a therapeutic target for AD”; I.F. prepared the section “PBM of LDRSB during sleep as a new trend in the therapy of AD”; O.S.-G., M.P., I.F., A.T., I.B. and M.T. prepared the section “PBM for AD in basic and clinical studies”. All authors were also involved in the editing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

O.S.-G., T.P., M.P., I.F., I.B., A.T. and J.K. were supported by an RSF grant (23-75-30001) and RF Governmental grant no. 075-15-2022-1094; J.K. was supported by the Academic leadership program Priority 2030 proposed by the First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank a designer, Elena Saranseva, for preparing the figures for our manuscript.

Conflicts of Interest

The authors declare that they have no competing interests.

References

  1. Li, X.; Feng, X.; Sun, X.; Hou, N.; Han, F.; Liu, Y. Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990–2019. Front. Aging Neurosci. 2022, 14, 937486. [Google Scholar] [CrossRef] [PubMed]
  2. GBD 2019 Risk Factors Collaborators. Global burden of 87 risk factors in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2020, 396, 1223–1249. [Google Scholar] [CrossRef] [PubMed]
  3. GBD 2019 Dementia Forecasting Collaborators. Estimation of the global prevalence of dementia in 2019 and forecasted prevalence in 2050: An analysis for the Global Burden of Disease Study 2019. Lancet Public Health 2022, 7, e105–e125. [Google Scholar] [CrossRef]
  4. Matthews, K.A.; Xu, W.; Gaglioti, A.H.; Holt, J.B.; Croft, J.B.; Mack, D.; McGuire, L.C. Racial and ethnic estimates of Alzheimer’s disease and related dementias in the United States (2015–2060) in adults aged ≥65 years. Alzheimers Dement. 2018, 15, 17–24. [Google Scholar] [CrossRef]
  5. Howard, R.; Liu, K.Y. Questions EMERGE as Biogen claims aducanumab turnaround. Nat. Rev. Neurol. 2020, 16, 63–64. [Google Scholar] [CrossRef]
  6. Panza, F.; Lozupone, M.; Seripa, D.; Imbimbo, B. Amyloid-β immunotherapy for Alzheimer disease: Is it now a long shot? Ann. Neurol. 2019, 85, 303–315. [Google Scholar] [CrossRef] [Green Version]
  7. Mullard, A. Landmark Alzheimer’s drug approval confounds research community. Nature 2021, 594, 309–310. [Google Scholar] [CrossRef] [PubMed]
  8. Salloway, S.; Chalkias, S.; Barkhof, F.; Burkett, P.; Barakos, J.; Purcell, D.; Suhy, J.; Forrestal, F.; Tian, Y.; Umans, K.; et al. Amyloid-Related Imaging Abnormalities in 2 Phase 3 Studies Evaluating Aducanumab in Patients with Early Alzheimer Disease. JAMA Neurol. 2022, 79, 13–21. [Google Scholar] [CrossRef]
  9. Lim, L. The Growing Evidence for Photobiomodulation as a Promising Treatment for Alzheimer’s Disease. J. Biosci. Med. 2018, 6, 100–110. [Google Scholar] [CrossRef] [Green Version]
  10. Berman, M.H.; Nichols, T.W. Treatment of Neurodegeneration: Integrating Photobiomodulation and Neurofeedback in Alzheimer’s Dementia and Parkinson’s: A Review. Photobiomodul. Photomed. Laser Surg. 2019, 37, 623–634. [Google Scholar] [CrossRef]
  11. Caldieraro, M.A.; Laufer-Silva, T.; Cassano, P. Dosimetry and Clinical Efficacy of Transcranial Photobiomodulation for Major Depression Disorder: Could they Guide Dosimetry for Alzheimer’s Disease? J. Alzheimers Dis. 2021, 83, 1453–1469. [Google Scholar] [CrossRef]
  12. Pan, W.T.; Liu, P.M.; Ma, D.; Yang, J.J. Advances in photobiomodulation for cognitive improvement by near-infrared derived multiple strategies. J. Transl. Med. 2023, 21, 135. [Google Scholar] [CrossRef] [PubMed]
  13. Saltmarche, A.E.; Naeser, M.A.; Ho, K.F.; Hamblin, M.R.; 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. [Google Scholar] [CrossRef] [PubMed]
  14. Chao, L.L. Effects of Home Photobiomodulation Treatments on Cognitive and Behavioral Function, Cerebral Perfusion, and Resting-State Functional Connectivity in Patients with Dementia: A Pilot Trial. Photobiomodul. Photomed. Laser Surg. 2019, 37, 133–141. [Google Scholar] [CrossRef] [PubMed]
  15. Maksimovich, I.V. Dementia and cognitive impairment reduction after laser transcatheter treatment of Alzheimer’s disease. World J. Neurosci. 2015, 5, 189–203. [Google Scholar] [CrossRef] [Green Version]
  16. Nizamutdinov, D.; Qi, X.; Berman, M.; Dougal, G.; Dayawansa, S.; Wu, E.; Yi, S.; Stevens, A.; Huang, J. Transcranial near infrared light stimulations improve cognition in patients with dementia. Aging Dis. 2021, 12, 954–963. [Google Scholar] [CrossRef] [PubMed]
  17. Hamblin, M.R. Photobiomodulation for Alzheimer’s Disease: Has the Light Dawned? Photonics 2019, 6, 77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Yang, M.; Yang, Z.; Wang, P.; Sun, Z. Current application and future directions of photobiomodulation in central nervous diseases. Neural Regen Res. 2021, 16, 1177–1185. [Google Scholar] [CrossRef]
  19. Stephan, W.; Banas, L.; Hamblin, M. Treatment Efficacy of Photobiomodulation for Moderate and Advanced Dementia or Alzheimer’s Disease: Case Studies. Adv. Alzheimer’s Dis. 2022, 11, 39–47. [Google Scholar] [CrossRef]
  20. Enengl, J.; Hamblin, M.; Dungel, P. Photobiomodulation for Alzheimer’s Disease: Translating Basic Research to Clinical Application. J. Alzheimers Dis. 2020, 75, 1073–1082. [Google Scholar] [CrossRef]
  21. Semyachkina-Glushkovskaya, O.; Penzel, T.; Blokhina, I.; Khorovodov, A.; Fedosov, I.; Yu, T.; Karandin, G.; Evsukova, A.; Elovenko, D.; Adushkina, V.; et al. Night Photostimulation of Clearance of Beta-Amyloid from Mouse Brain: New Strategies in Preventing Alzheimer’s Disease. Cells 2021, 10, 3289. [Google Scholar] [CrossRef] [PubMed]
  22. Semyachkina-Glushkovskaya, O.; Postnov, D.; Lavrova, A.; Fedosov, I.; Borisova, E.; Nikolenko, V.; Penzel, T.; Kurths, J.; Tuchin, V. Biophotonic Strategies of Measurement and Stimulation of the Cranial and the Extracranial Lymphatic Drainage Function. IEEE J. Sel. Top. 2021, 27, 1–13. [Google Scholar] [CrossRef]
  23. Semyachkina-Glushkovskaya, O.; Klimova, M.; Iskra, T.; Bragin, D.; Abdurashitov, A.; Dubrovsky, A.; Khorovodov, A.; Terskov, A.; Blokhina, I.; Lezhnev, N.; et al. Transcranial Photobiomodulation of Clearance of Beta-Amyloid from the Mouse Brain: Effects on the Meningeal Lymphatic Drainage and Blood Oxygen Saturation of the Brain. Advances in Experimental Medicine and Biology. Adv. Exp. Med. Biol. 2021, 1269, 57–61. [Google Scholar]
  24. Semyachkina-Glushkovskaya, O.; Abdurashitov, A.; Dubrovsky, A.; Klimova, M.; Agranovich, I.; Terskov, A.; Shirokov, A.; Vinnik, V.; Kuznecova, A.; Lezhnev, N.; et al. Photobiomodulation of lymphatic drainage and clearance: Perspective strategy for augmentation of meningeal lymphatic functions. Biomed Opt. Express 2020, 11, 725–734. [Google Scholar] [CrossRef]
  25. Zhinchenko, E.; Klimova, M.; Mamedova, A.; Agranovich, I.; Blokhina, I.; Antonova, T.; Terskov, A.; Shirokov, A.; Navolkin, N.; Morgun, A.; et al. Photostimulation of extravasation of beta-amyloid through the model of blood-brain barrier. Electronics 2020, 9, 1056. [Google Scholar] [CrossRef]
  26. Semyachkina-Glushkovskaya, O.; Abdurashitov, A.; Klimova, M.; Dubrovsky, A.; Shirokov, A.; Fomin, A.; Terskov, A.; Agranovich, I.; Mamedova, A.; Khorovodov, A.; et al. Photostimulation of cerebral and peripheral lymphatic functions. Transl. Biophotonics 2020, 2, e201900036. [Google Scholar] [CrossRef]
  27. Zhinchenko, E.; Navolokin, N.; Shirokov, A.; Khlebcov, B.; Dubrovsky, A.; Saranceva, E.; Abdurashitov, A.; Khorovodov, A.; Terskov, A.; Mamedova, A.; et al. Pilot study of transcranial photobiomodulation of lymphatic clearance of beta-amyloid from the mouse brain: Breakthrough strategies for nonpharmacologic therapy of Alzheimer’s disease. Biomed Opt. Exp. 2019, 8, 4003–4017. [Google Scholar] [CrossRef] [PubMed]
  28. Salehpour, F.; Khademi, M.; Bragin, D.; DiDuro, J. Photobiomodulation Therapy and the Glymphatic System: Promising Applications for Augmenting the Brain Lymphatic Drainage System. Int. J. Mol. Sci. 2022, 23, 2975. [Google Scholar] [CrossRef]
  29. Semyachkina-Glushkovskaya, O.; Fedosov, I.; Shirokov, A.; Vodovozov, E.; Alekseev, A.; Khorovodov, A.; Blokhina, I.; Terskov, A.; Mamedova, A.; Klimova, M.; et al. Photomodulation of lymphatic delivery of liposomes to the brain bypassing the blood-brain barrier: New perspectives for glioma therapy. Nanophotonics 2021, 12, 3215–3227. [Google Scholar] [CrossRef]
  30. Semyachkina-Glushkovskaya, O.; Diduk, S.; Anna, E.; Elina, D.; Artem, K.; Khorovodov, A.; Shirokov, A.; Fedosov, I.; Dubrovsky, A.; Blokhina, I.; et al. Photomodulation of Lymphatic Delivery of Bevacizumab to the Brain: The Role of Singlet Oxygen. Adv. Exp. Med. Biol. 2022, 1395, 53–57. [Google Scholar]
  31. Semyachkina-Glushkovskaya, O.; Shirokov, A.; Blokhina, I.; Telnova, V.; Vodovozova, E.; Alekseeva, A.; Boldyrev, I.; Fedosov, I.; Dubrovsky, A.; Khorovodov, A.; et al. Intranasal delivery of liposomes to glioblastoma by photostimulation of the lymphatic system. Pharmaceutics 2023, 15, 36. [Google Scholar] [CrossRef]
  32. Semyachkina-Glushkovskaya, O.; Bragin, D.; Bragina, O.; Socolovski, S.; Shirokov, A.; Fedosov, I.; Ageev, V.; Blokhina, I.; Dubrovsky, A.; Telnova, V.; et al. Low-Level Laser Treatment Induces the Blood-Brain Barrier Opening and the Brain Drainage System Activation: Delivery of Liposomes into Mouse Glioblastoma. Pharmaceutics 2023, 15, 567. [Google Scholar] [CrossRef]
  33. Xie, L.; Kang, H.; Xu, Q.; Chen, M.J.; Liao, Y.; Thiyagarajan, M.; O’Donnell, J.; Christensen, D.J.; Nicholson, C.; Iliff, J. Sleep drives metabolite clearance from the adult brain. Science 2013, 342, 373–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Fultz, N.E.; Bonmassar, G.; Setsompop, K.; Stickgold, R.; Rosen, B.; Polimeni, J.; Lewis, L. Coupled electrophysio-logical, hemodynamic, and cerebrospinal fluid oscillations in human sleep. Science 2019, 366, 628–631. [Google Scholar] [CrossRef] [PubMed]
  35. Semyachkina-Glushkovskaya, O.; Postnov, D.; Penzel, T.; Kurths, J. Sleep as a Novel Biomarker and a Promising Therapeutic Target for Cerebral Small Vessel Disease: A Review Focusing on Alzheimer’s Disease and the Blood-Brain Barrier. Int. J. Mol. Sci. 2020, 21, 6293. [Google Scholar] [CrossRef] [PubMed]
  36. Semyachkina-Glushkovskaya, O.; Fedosov, I.; Penzel, T.; Li, D.; Yu, T.; Telnova, V.; Kaybeleva, E.; Saranceva, E.; Terskov, A.; Khorovodov, A.; et al. Brain Waste Removal System and Sleep: Photobiomodulation as an Innovative Strategy for Night Therapy of Brain Diseases. Int. J. Mol. Sci. 2023, 24, 3221. [Google Scholar] [CrossRef]
  37. Moro, C.; Valverde, A.; Dole, M.; Hoh Kam, J.; Hamilton, C.; Liebert, A.; Bicknell, B.; Benabid, A.-L.; Magistretti, P.; Mitrofanis, J. The effect of photobiomodulation on the brain during wakefulness and sleep. Front. Neurosci. 2022, 16, 942536. [Google Scholar] [CrossRef]
  38. Valverde, A.; Hamilton, C.; Moro, C.; Billeres, M.; Magistretti, P.; Mitrofanis, J. Lights at night: Does photobiomodulation improve sleep? Neural Regen. Res. 2023, 18, 474–477. [Google Scholar]
  39. Da Mesquita, S.; Louveau, A.; Vaccari, A. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature 2018, 560, 185–191. [Google Scholar] [CrossRef]
  40. Dupont, G.; Iwanaga, J.; Yilmaz, E.; Tubbs, R.S. Connections Between Amyloid Beta and the Meningeal Lymphatics as a Possible Route for Clearance and Therapeutics. Lymphat. Res. Biol. 2020, 18, 2–6. [Google Scholar] [CrossRef] [Green Version]
  41. Chen, J.; Wang, L.; Xu, H. Meningeal lymphatics clear erythrocytes that arise from subarachnoid hemorrhage. Nat. Commun. 2020, 11, 3159. [Google Scholar] [CrossRef] [PubMed]
  42. Liu, X.; Gao, C.; Yuan, J. Subdural haematomas drain into the extracranial lymphatic system through the meningeal lymphatic vessels. Acta Neuropathol. Commun. 2020, 8, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Bolte, A.C.; Dutta, A.B.; Hurt, M.E. Meningeal lymphatic dysfunction exacerbates traumatic brain injury pathogenesis. Nat. Commun. 2020, 11, 4524. [Google Scholar] [CrossRef]
  44. Hu, X.; Deng, Q.; Ma, L.; Li, Q.; Chen, Y.; Liao, Y.; Zhou, F.; Zhang, C.; Shao, L.; Feng, J.; et al. Meningeal lymphatic vessels regulate brain tumor drainage and immunity. Cell Res. 2020, 30, 229–243. [Google Scholar] [CrossRef] [Green Version]
  45. Li, D.; Liu, S.; Yu, T.; Liu, Z.; Sun, S.; Bragin, D.; Navolokin, N.; Kurths, J.; Glushkovskaya-Semyachkina, O.; Zhu, D. Photostimulation of lymphatic clearance of red blood cells from the mouse brain after intraventricular hemorrhage. bioRxiv 2020, 16, 384149. [Google Scholar]
  46. Louveau, A.; Smirnov, I.; Keyes, T.; Eccles, J.; Rouhani, S.; Peske, J.; Derecki, N.; Castle, D.; Mandell, J.; Lee, K.; et al. Structural and functional features of central nervous system lymphatic vessels. Nature 2015, 523, 337–341. [Google Scholar] [CrossRef] [Green Version]
  47. Hampel, H.; Hardy, J.; Blennow, K.; Chen, C.; Perry, G.; Kim, S.H.; Villemagne, V.L.; Aisen, P.; Vendruscolo, M.; Iwatsubo, T.; et al. The Amyloid-β Pathway in Alzheimer’s Disease. Mol. Psychiatry 2021, 26, 5481–5503. [Google Scholar] [CrossRef]
  48. Veitch, D.; Weiner, M.; Aisen, P.; Beckett, L.; Cairns, N.; Green, R.; Harvey, D.; Jack, C.; Jagust, W.; Morris, J.; et al. Alzheimer’s Disease Neuroimaging Initiative. Understanding disease progression and improving Alzheimer’s disease clinical trials: Recent highlights from the Alzheimer’s Disease Neuroimaging Initiative. Alzheimers Dement. 2019, 15, 106–152. [Google Scholar] [CrossRef]
  49. Baumann, K.; Šneiderienė, G.; Sanguanini, M.; Schneider, M.; Rimon, O.; González Díaz, A.; Greer, H.; Thacker, D.; Linse, S.; Knowles, T.P.J.; et al. Kinetic Map of the Influence of Biomimetic Lipid Model Membranes on Aβ42 Aggregation. ACS Chem. Neurosci. 2023, 14, 323–329. [Google Scholar] [CrossRef]
  50. Joachim, C.; Duffy, L.; Morris, J.; Selkoe, D. Protein chemical and immunocytochemical studies of meningovascular β-amyloid protein in Alzheimer’s disease and normal aging. Brain Res. 1988, 474, 100–111. [Google Scholar] [CrossRef]
  51. Iliff, J.; Wang, M.; Liao, Y.; Plogg, B.; Peng, W.; Gundersen, G.; Benveniste, H.; Vates, G.; Deane, R.; Goldman, S.; et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci. Transl. Med. 2012, 4, 147ra111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Kress, B.; Iliff, J.; Xia, M.; Wang, M.; Wei, H.; Zeppenfeld, D.; Xie, L.; Kang, H.; Xu, Q.; Liew, J.; et al. Impairment of paravascular clearance pathways in the aging brain. Ann. Neurol. 2014, 76, 845–861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ahn, J.H.; Cho, H.; Kim, J.; Kim, S.; Ham, J.; Park, I.; Suh, S.; Hong, S.; Song, J.; Hong, Y.; et al. Meningeal lymphatic vessels at the skull base drain cerebrospinal fluid. Nature 2019, 572, 62–66. [Google Scholar] [CrossRef]
  54. Jiang, H.; Wei, H.; Zhou, Y.; Xiao, X.; Zhou, C.; Ji, X. Overview of the meningeal lymphatic vessels in aging and central nervous system disorders. Cell Biosci. 2022, 12, 202. [Google Scholar] [CrossRef]
  55. Chee, J.; Solito, E. The Impact of Ageing on the CNS Immune Response in Alzheimer’s Disease. Front. Immunol. 2021, 12, 738511. [Google Scholar] [CrossRef]
  56. Wu, K.; Zhang, Y.; Huang, Y.; Dong, Q.; Tan, L.; Yu, J. The role of the immune system in Alzheimer’s disease. Ageing Res. Rev. 2021, 70, 101409. [Google Scholar] [CrossRef]
  57. Xu, J.; Liu, Q.; Huang, S.; Duan, C.; Lu, H.; Cao, Y.; Hu, J. The lymphatic system: A therapeutic target for central nervous system disorders. Neural Regen. Res. 2023, 18, 1249–1256. [Google Scholar]
  58. Da Mesquita, S.; Papadopoulos, Z.; Dykstra, T.; Brase, L.; Farias, F.; Wall, M.; Jiang, H.; Kodira, C.; de Lima, K.; Herz, J.; et al. Meningeal lymphatics affect microglia responses and anti-Aβ immunotherapy. Nature 2021, 593, 255–260. [Google Scholar] [CrossRef]
  59. Laaker, C.; Fabry, Z. The meningeal lymphatics: Regulators of Aβ immunotherapy? Trends Immunol. 2021, 42, 940–942. [Google Scholar] [CrossRef] [PubMed]
  60. Wen, Y.; Yang, J.; Wang, X.; Yao, Z. Induced dural lymphangiogenesis facilities soluble amyloid-beta clearance from brain in a transgenic mouse model of Alzheimer’s disease. Neural Regen. Res. 2018, 13, 709–716. [Google Scholar]
  61. Choi, C.; Park, J.; Kim, H.; Chang, K.T.; Park, J.; Min, K. DSCR1 upregulation enhances dural meningeal lymphatic drainage to attenuate amyloid pathology of Alzheimer’s disease. J. Pathol. 2021, 10, 5767. [Google Scholar]
  62. Wu, Y.; Zhang, T.; Li, X.; Wei, Y.; Li, X.; Wang, S.; Liu, J.; Li, D.; Wang, S.; Ye, T. Borneol-driven meningeal lymphatic drainage clears amyloid-β peptide to attenuate Alzheimer-like phenotype in mice. Theranostics 2023, 13, 106–124. [Google Scholar] [CrossRef] [PubMed]
  63. Sommer, A.; Bieschke, J.; Friedrich, R.; Zhu, D.; Wanker, E.; Fecht, H.; Mereles, D.; Hunstein, W. 670 nm laser light and EGCG complementarily reduce amyloid-β aggregates in human neuroblastoma cells: Basis for treatment of Alzheimer’s disease? Photomed. Laser Surg. 2012, 1, 54–60. [Google Scholar] [CrossRef] [PubMed]
  64. Yang, X.; Askarova, S.; Sheng, W.; Chen, J.K.; Sun, A.; Sun, G.; Yao, G.; Lee, J.C.-M. Low energy laser light (632.8 nm) suppresses amyloid-peptide-induced oxidative and inflammatory responses in astrocytes. Neurosci 2010, 171, 859–868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Liang, J.; Liu, L.; Xing, D. Photobiomodulation by low-power laser irradiation attenuates Aβ-induced cell apoptosis through the Akt/GSK3β/β-catenin pathway. Free Radic. Biol. Med. 2012, 7, 1459–1467. [Google Scholar] [CrossRef]
  66. Zhang, H.; Wu, S.; Xing, D. Inhibition of Aβ (25-35)-induced cell apoptosis by low-power-laser-irradiation (LPLI) through promoting Akt-dependent YAP cytoplasmic translocation. Cell Signal. 2012, 24, 224–232. [Google Scholar] [CrossRef]
  67. Meng, C.; He, Z.; Xing, D. Low-level laser therapy rescues dendrite atrophy via upregulating BDNF expression: Implications for Alzheimer’s disease. J. Neurosci. 2013, 33, 13505–13517. [Google Scholar] [CrossRef] [Green Version]
  68. De Taboada, L.; Yu, J.; El-Amouri, S.; Gattoni-Celli, S.; Richieri, S.; McCarthy, T.; Jackson, S.; Mark, K.S. Transcranial laser therapy attenuates amyloid-beta peptide neuropathology in amyloid-beta protein precursor transgenic mice. J. Alzheimer’s Dis. 2011, 23, 521–535. [Google Scholar] [CrossRef]
  69. 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. [Google Scholar] [CrossRef]
  70. Purushothuman, S.; Johnstone, D.; 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. [Google Scholar] [CrossRef] [Green Version]
  71. Purushothuman, S.; Johnstone, D.; Nandasena, C.; van Eersel, J.; Ittner, L.; Mitrofanis, J.; Stone, J. Near infrared light mitigates cerebellar pathology in transgenic mouse models of dementia. Neurosci. Lett. 2015, 591, 155–159. [Google Scholar] [CrossRef] [PubMed]
  72. Farfara, D.; Tuby, H.; Trudler, D.; Doron-Mandel, E.; Maltz, L.; Vassar, R.J.; 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. [Google Scholar] [CrossRef] [PubMed]
  73. Oron, A.; Oron, U. Low-level laser therapy to the bone marrow ameliorates neurodegenerative disease progression in a mouse model of Alzheimer’s disease: A minireview. Photomed. Laser Surg. 2016, 34, 627–630. [Google Scholar] [CrossRef] [PubMed]
  74. Lu, Y.; Wang, R.; Dong, Y.; Tucker, D.; Zhao, N.; Ahmed, M.; Zhu, L.; Liu, T.; Cohen, R.; Zhang, Q. Low-level laser therapy for beta amyloid toxicity in rat hippocampus. Neurobiol. Aging 2017, 49, 165–182. [Google Scholar] [CrossRef] [Green Version]
  75. Da Luz Eltchechem, C.; Salgado, A.S.I.; Zangaro, R.A.; da Silva Pereira, M.C.; Kerppers, I.I.; da Silva, L.A.; Parreira, R.B. Transcranial LED therapy on amyloid-beta toxin 25–35 in the hippocampal region of rats. Lasers Med. Sci. 2017, 32, 749–756. [Google Scholar] [CrossRef]
  76. Blivet, G.; Meunier, J.; Roman, F.J.; Touchon, J. Neuroprotective effect of a new photobiomodulation technique against Aβ25–35 peptide-induced toxicity in mice: Novel hypothesis for therapeutic approach of Alzheimer’s disease suggested. Alzheimer’s Dement. 2018, 4, 54–63. [Google Scholar] [CrossRef]
  77. Cho, G.; Lee, S.; Park, J.; Kim, M.; Park, M.; Choi, B.; Shin, Y.; Kim, N.; Shin, H. Photobiomodulation using a low-level light-emitting diode improves cognitive dysfunction in the 5XFAD mouse model of Alzheimer’s disease. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 631–639. [Google Scholar] [CrossRef]
  78. Zhang, Z.; Shen, Q.; Wu, X.; Zhang, D.; Xing, D. Activation of PKA/SIRT1 signaling pathway by photobiomodulation therapy reduces Abeta levels in Alzheimer’s disease models. Aging Cell 2020, 19, e13054. [Google Scholar] [CrossRef]
  79. Hamblin, M.; Salehpour, F. Photobiomodulation of the Brain: Shining Light on Alzheimer’s and Other Neuropathological Diseases. Am. J. Alzheimer’s Dis. 2021, 83, 1395–1397. [Google Scholar] [CrossRef]
  80. Barrett, D.; Gonzalez-Lima, F. Transcranial infrared laser stimulation produces beneficial cognitive and emotional effects in humans. Neuroscience 2013, 230, 13–23. [Google Scholar] [CrossRef]
  81. Blanco, N.J.; Maddox, W.T.; Gonzalez-Lima, F. Improving executive function using transcranial infrared laser stimulation. J. Neuropsychol. 2017, 11, 14–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Chan, A.S.; Lee, T.; Yeung, M.; Hamblin, M. Photobiomodulation improves the frontal cognitive function of older adults. Int. J. Geriatr. Psychiatry 2019, 2, 369–377. [Google Scholar] [CrossRef]
  83. Wu, J.; Chang, W.; Hsieh, C.; Jiang, J.; Fang, W.; Shan, Y.; Chang, Y. Effect of low-level laser stimulation on EEG. Evid. Based Complement Alternat. Med. 2012, 2012, 951272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Jahan, A.; Nazari, M.; Mahmoudi, J. Transcranial near-infrared photobiomodulation could modulate brain electrophysiological features and attentional performance in healthy young adults. Lasers Med. Sci. 2019, 34, 1193–1200. [Google Scholar] [CrossRef] [PubMed]
  85. Maksimovich, I.V. Laser Technologies as a New Direction in Transcatheter Interventions. Photobiomodul. Photomed. Laser Surg. 2019, 37, 455–456. [Google Scholar] [CrossRef] [PubMed]
  86. Ailioaie, L.M.; Ailioaie, C.; Litscher, G. Photobiomodulation in Alzheimer’s Disease—A Complementary Method to State-of-the-Art Pharmaceutical Formulations and Nanomedicine? Pharmaceutics 2023, 15, 916. [Google Scholar] [CrossRef]
  87. Huynh, Q.-S.; Elangovan, S.; Holsinger, R.M.D. Non-Pharmacological Therapeutic Options for the Treatment of Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 11037. [Google Scholar] [CrossRef]
  88. Chao, L. Impact of Photobiomodulation (PBM) on Biomarkers of Alzheimer’s Disease (PBMbiomarker). 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT03405662 (accessed on 20 April 2023).
  89. Lah, J. Stimulating Neural Activity to Improve Blood Flow and Reduce Amyloid: Path to Clinical Trials. 2021. Available online: https://clinicaltrials.gov/ct2/show/NCT03543878 (accessed on 20 April 2023).
  90. Zomorrodi, R.; Saltmarche, A.; Loheswaran, G.; Ho, K.; Lim, L. Complementary EEG Evidence for a Significantly Improved Alzheimer’s Disease Case after Photobiomodulation Treatment. Paper Presented at the 26th Annual Scientific Conference, Canadian Academy of Geriatric Psychiatry Toronto. Alzheimer’s Dement. 2017, 13, 621. [Google Scholar] [CrossRef]
  91. Salehpour, F.; Hamblin, M.R.; Di Duro, J. Rapid Reversal of Cognitive Decline, Olfactory Dysfunction, and Quality of Life Using Multi-Modality Photobiomodulation Therapy: Case Report. Photobiomodul. Photomed. Laser Surg. 2019, 37, 159–167. [Google Scholar] [CrossRef]
  92. Berman, M. Photobiomodulation with near infrared light helmet in a pilot, placebo controlled clinical trial in dementia patients testing memory and cognition. J. Neurol. Neurosci. 2017, 2171, 76. [Google Scholar] [CrossRef] [Green Version]
  93. Qi, X.; Nizamutdinov, D.; Berman, M.; Dougal, G.; Chazot, P.; Wu, E.; Stevens, A.; Yi, S.; Huang, J. Gender Differences of Dementia in Response to Intensive Self-Administered Transcranial and Intraocular Near-Infrared Stimulation. Cureus 2021, 13, e16188. [Google Scholar] [CrossRef] [PubMed]
  94. Horner, S.; Berger, L.; Gibas, K. Nutritional Ketosis and photobiomodulation remediate mitochondria warding off Alzheimer’s disease in a diabetic, ApoE4+ patient with mild cognitive impairment: A case report. Photodiagnosis. Photodyn. Ther. 2020, 30, 101777. [Google Scholar] [CrossRef] [PubMed]
  95. Lapchak, P. Transcranial near-infrared laser therapy applied to promote clinical recovery in acute and chronic neurodegenerative diseases. Expert Rev. Med. Devices 2012, 1, 71–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Komel’kova, L.; Vitreshchak, T.; Zhirnova, I. Biochemical and immunological induces of the blood in Parkinson’s disease and their correction with the help of laser therapy. Patol. Fiziol. Eksp. Ter. 2004, 1, 15–18. [Google Scholar]
  97. Vitreshchak, T.; Mikhailov, V.; Piradov, M. Laser modification of the blood in vitro and in vivo in patients with Parkinson’s disease. Bull. Exp. Biol. Med. 2003, 5, 430–432. [Google Scholar] [CrossRef]
  98. Trimmer, P.; Schwartz, K.; Borland, M. Reduced axonal transport in Parkinson’s disease cybrid neurites is restored by light therapy. Mol. Neurodegener. 2009, 4, 26. [Google Scholar] [CrossRef] [Green Version]
  99. Moges, H.; Vasconcelos, O.; Campbell, W. Light therapy and supplementary riboflavin in the SOD1 transgenic mouse model of familial amyotrophic lateral sclerosis (FALS). Lasers Surg. Med. 2009, 1, 52–59. [Google Scholar] [CrossRef]
  100. Muili, K.; Gopalakrishnan, S.; Meyer, S.; Eells, J.; Lyons, J. Amelioration of experimental autoimmune encephalomyelitis in C57BL/6 mice by photobiomodulation induced by 670 nm light. PLoS ONE 2012, 7, e30655. [Google Scholar] [CrossRef] [Green Version]
  101. 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. [Google Scholar]
  102. Tucker, L.; 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. [Google Scholar] [CrossRef]
  103. Yang, L.; Dong, Y.; Wu, C.; Li, Y.; Guo, Y.; Yang, B.; Zong, X.; Hamblin, M.; Liu, T.; Zhang, Q. Photobiomodulation preconditioning prevents cognitive impairment in a neonatal rat model of hypoxia-ischemia. J. Biophotonics 2019, 12, e201800359. [Google Scholar] [CrossRef] [PubMed]
  104. Lucey, B.; McCullough, A.; Landsness, E.; Toedebusch, C.; McLeland, J.; Zaza, A.; Fagan, A.; McCue, L.; Xiong, C.; Morris, J. Reduced non–rapid eye movement sleep is associated with tau pathology in early Alzheimer’s disease. Sci. Transl. Med. 2019, 11, eaau6550. [Google Scholar] [CrossRef] [PubMed]
  105. Lutsey, P.; Misialek, J.; Mosley, T.; Gottesman, R.; Punjabi, N.; Shahar, E.; MacLehose, R.; Ogilvie, R.; Knopman, D.; Alonso, A. Sleep characteristics and risk of dementia and Alzheimer’s disease: The atherosclerosis risk in communities study. Alzheimer Dement. 2018, 14, 157–166. [Google Scholar] [CrossRef] [PubMed]
  106. Mendelsohn, A.; Larrick, J. Sleep facilitates clearance of metabolites from the brain: Glymphatic function in aging and neurodegenerative diseases. Rejuven. Res. 2013, 16, 518–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Carvalho, D.; Knopman, D.; Boeve, D.; Lowe, V.; Roberts, R.; Mielke, M.; Przybelski, S.; Machulda, M.; Petersen, R.; Jack, C.; et al. Association of excessive daytime sleepiness with longitudinal beta-amyloid accumulation in elderly persons without dementia. JAMA Neurol. 2018, 75, 672–680. [Google Scholar] [CrossRef] [Green Version]
  108. Izci-Balserak, B.; Zhu, B.; Wang, H.; Bronas, U.; Gooneratne, N. Independent associations between sleep duration, gamma gap, and cognitive function among older adults: Results from the NHANES 2013–2014. Geriatr. Nurs. 2022, 44, 1–7. [Google Scholar] [CrossRef]
  109. Spira, A.P.; Gamaldo, A.A.; An, Y.; Wu, M.N.; Simonsick, E.M.; Bilgel, M.; Zhou, Y.; Wong, D.F.; Ferrucci, L.; Resnick, S.M. Self-reported sleep and beta-amyloid deposition in community-dwelling older adults. JAMA Neurol. 2013, 70, 1537–1543. [Google Scholar]
  110. Minakawa, E.; Wada, K.; Nagai, Y. Sleep disturbance as a potential modifiable risk factor for Alzheimer’s disease. Int. J. Mol. Sci. 2019, 20, 803. [Google Scholar] [CrossRef] [Green Version]
  111. Song, D.; Zhou, J.; Ma, J.; Chang, J.; Qiu, Y.; Zhuang, Z.; Xiao, H.; Zeng, L. Sleep disturbance mediates the relationship between depressive symptoms and cognitive function in older adults with mild cognitive impairment. Geriatr. Nurs. 2021, 42, 1019–1023. [Google Scholar] [CrossRef]
  112. Wang, Z.; Heizhati, M.; Wang, L.; Li, M.; Yang, Z.; Lin, M.; Abudereyimu, R.; Hong, J.; Yang, W.; Yao, L.; et al. Poor sleep quality is negatively associated with low cognitive performance in general population independent of self-reported sleep disordered breathing. BMC Public Health 2022, 22, 3. [Google Scholar] [CrossRef]
  113. Sabia, S.; Fayosse, A.; Dumurgier, J.; Van Hees, V.; Paquet, C.; Sommerlad, A.; Kivimäki, M.; Dugravot, A.; Singh-Manoux, A. Association of sleep duration in middle and old age with incidence of dementia. Nat. Commun. 2021, 12, 2289. [Google Scholar] [CrossRef] [PubMed]
  114. Ooms, S.; Overeem, S.; Besse, K.; Rikkert, M.; Marcel Verbeek, M.; Claassen, J. Effect of 1 night of total sleep deprivation on cerebrospinal fluid β-amyloid 42 in healthy middle-aged men: A randomized clinical trial. JAMA Neurol. 2014, 71, 971–977. [Google Scholar] [CrossRef] [PubMed]
  115. Shokri-Kojoria, E.; Wanga, G.-J.; Wiersa, C.; Demirala, S.; Guoa, M.; Kima, S.; Lindgrena, E.; Ramireza, V.; Zehraa, A.; Freemana, C.; et al. β-Amyloid accumulation in the human brain after one night of sleep deprivation. Proc. Natl. Acad. Sci. USA 2018, 115, 4483–4488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Reddy, O.; Van der Werf, Y. The Sleeping Brain: Harnessing the Power of the Glymphatic System through Lifestyle Choices. Brain Sci. 2020, 10, 868. [Google Scholar] [CrossRef] [PubMed]
  117. Silva, I.; Silva, J.; Ferreira, R.; Trigo, D. Glymphatic system, AQP4, and their implications in Alzheimer’s disease. Neurol. Res. Pract. 2021, 3, 5. [Google Scholar] [CrossRef]
  118. Nedergaard, M.; Goldman, S. Glymphatic failure as a final common pathway to dementia. Science 2020, 370, 50–56. [Google Scholar] [CrossRef]
  119. Hablitz, L.; Vinitsky, H.; Sun, Q.; Stæger, F.; Sigurdsson, B.; Mortensen, K.; Lilius, T.; Nedergaard, M. Increased glymphatic influx is correlated with high EEG delta power and low heart rate in mice under anesthesia. Sci. Adv. 2019, 5, eaav5447. [Google Scholar] [CrossRef] [Green Version]
  120. Benveniste, H.; Heerdt, P.; Fontes, M.; Rothman, D.; Volkow, N. Glymphatic system function in relation to anesthesia and sleep states. Anesth. Analg. 2019, 128, 747–758. [Google Scholar] [CrossRef]
  121. Lee, Y.; Gerashchenko, D.; Timofeev, I.; Bacskai, B.; Kastanenka, K. Slow wave sleep as a promising intervention target for Alzheimer’s disease. Front. Neurosci. 2020, 14, 705. [Google Scholar] [CrossRef]
  122. Kastanenka, K.; Calvo-Rodriguez, M.; Hou, S.; Zhou, H.; Takeda, S.; Arbel-Ornath, M.; Lariviere, A.; Lee, Y.F.; Kim, A.; Hawkes, J.M.; et al. Frequency-dependent exacerbation of Alzheimer’s disease neuropathophysiology. Sci. Rep. 2019, 9, 8964. [Google Scholar] [CrossRef] [Green Version]
  123. Kent, B.; Strittmatter, S.; Nygaard, H. Sleep and EEG power spectral analysis in three transgenic mouse models of Alzheimer’s disease: APP/PS1, 3xTgAD, and Tg2576. J. Alzheimer Dis. 2018, 64, 1325–1336. [Google Scholar] [CrossRef]
  124. Castano-Prat, P.; Perez-Mendez, L.; Perez-Zabalza, M.; Sanfeliu, C.; Giménez-Llort, L.; Sanchez-Vives, M. Altered slow (<1 Hz) and fast (beta and gamma) neocortical oscillations in the 3xTg-AD mouse model of Alzheimer’s disease under anesthesia. Neurobiol. Ageing 2019, 79, 142–151. [Google Scholar]
  125. Mander, B.; Rao, V.; Lu, B.; Saletin, J.; Lindquist, J.; Ancoli-Israel, S.; Jagust, W.; Walker, M. Prefrontal atrophy, disrupted NREM slow waves and impaired hippocampal-dependent memory in aging. Nat. Neurosci. 2013, 16, 357. [Google Scholar] [CrossRef] [PubMed]
  126. Mander, B.; Marks, S.; Vogel, J.; Rao, V.; Lu, B.; Saletin, J.; Ancoli-Israel, S.; Jagust, W.; Walker, M. b-amyloid disrupts human NREM slow waves and related hippocampus-dependent memory consolidation. Nat. Neurosci. 2015, 18, 1051–1057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Westerberg, C.; Mander, B.; Florczak, S.; Weintraub, S.; Mesulam, M.; Zee, P.; Paller, K. Concurrent impairments in sleep and memory in amnestic mild cognitive impairment. J. Int. Neuropsychol. Soc. JINS 2012, 18, 490. [Google Scholar] [CrossRef] [Green Version]
  128. Kastanenka, K.; Hou, S.; Shakerdge, N.; Logan, R.; Feng, D.; Wegmann, S.; Chopra, V.; Hawkes, J.; Chen, X.; Bacskai, B. Optogenetic restoration of disrupted slow oscillations halts amyloid deposition and restores calcium homeostasis in an animal model of Alzheimer’s disease. PLoS ONE 2017, 12, e0170275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Selkoe, D. Early network dysfunction in Alzheimer’s disease. Science 2019, 365, 540–541. [Google Scholar] [CrossRef] [PubMed]
  130. Cirrito, J.; Kang, J.; Lee, J.; Stewart, F.; Verges, D.; Silverio, L.; Bu, G.; Mennerick, S.; Holtzman, D. Endocytosis is required for synaptic activity-dependent release of amyloid-b in vivo. Neuron 2008, 58, 42–51. [Google Scholar] [CrossRef] [Green Version]
  131. Kuchibhotla, K.; Lattarulo, C.; Hyman, B.; Bacskai, B. Synchronous hyperactivity and intercellular calcium waves in astrocytes in Alzheimer mice. Science 2009, 323, 1211–1215. [Google Scholar] [CrossRef] [Green Version]
  132. Robinson, S. Neuronal expression of glutamine synthetase in Alzheimer’s disease indicates a profound impairment of metabolic interactions with astrocytes. Neurochem. Int. 2000, 36, 471–482. [Google Scholar] [CrossRef]
  133. Poskanzer, K.; Yuste, R. Astrocytes regulate cortical state switching in vivo. Proc. Natl. Acad. Sci. USA 2016, 113, 2675–2684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. O’Donnell, J.; Zeppenfeld, D.; McConnell, E.; Pena, S.; Nedergaard, M. Norepinephrine: A neuromodulator that boosts the function of multiple cell types to optimize CNS performance. Neurochem. Res. 2012, 37, 2496–2512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Cipolla, M.; Li, R.; Vitullo, L. Perivascular innervation of penetrating brain parenchymal arterioles. J. Cardiovasc. Pharmacol. 2004, 44, 1–8. [Google Scholar] [CrossRef] [PubMed]
  136. Lee, H.; Xie, L.; Yu, M.; Kang, H.; Feng, T.; Deane, R.; Logan, J.; Nedergaard, M.; Benveniste, H. The effect of body posture on brain glymphatic transport. J. Neurosci. 2015, 35, 11034–11044. [Google Scholar] [CrossRef] [PubMed]
  137. McGee, C.; Liebert, A.; Herkes, G.; Bicknell, B.; Pang, V.; McLachlan, C.; Kiat, H. Protocol for randomized controlled trial to evaluate the safety and feasibility of a novel helmet to deliver transcranial light emitting diodes photobiomodulation therapy to patients with Parkinson’s disease. Front. Neurosci. 2022, 16, 945796. [Google Scholar] [CrossRef]
  138. Stephan, W.; Banas, L.; Misiak, M.; Brierley, W.; Hamblin, M. Photobiomodulation with Super-Pulsed Laser Shows Efficacy for Stroke and Aphasia: Case Studies. World J. Neurosci. 2023, 13, 12–20. [Google Scholar] [CrossRef]
  139. Hamblin, M. Photobiomodulation for traumatic brain injury and stroke. J. Neurosci. Res. 2018, 96, 731–743. [Google Scholar] [CrossRef] [Green Version]
  140. Rindner, E.; Haroon, J.; Jordan, K.; Mahdavi, K.; Surya, J.; Zielinski, M.; Habelhah, B.; Venkatraman, V.; Becerra, S.; Chan, L.; et al. Transcranial Infrared Laser Stimulation for the Treatment of Traumatic Brain Injury: A Case Series. J. Lasers Med. Sci. 2022, 13, e65. [Google Scholar] [CrossRef]
  141. Bohlen, H.; Gasheva, O.; Zawieja, D. Nitric oxide formation by lymphatic bulb and valves is a major regulatory component of lymphatic pumping. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H1897–H1906. [Google Scholar] [CrossRef] [Green Version]
  142. Kunert, C.; Baish, J.; Liao, S.; Padera, T.; Munn, L. Mechanobiological oscillators control lymph flow. Proc. Natl. Acad. Sci. USA 2015, 112, 10938–10943. [Google Scholar] [CrossRef] [Green Version]
  143. Mizuno, R.; Ono, N.; Ohhashi, T. Involvement of ATP-sensitive K(+) channels in spontaneous activity of isolated lymph microvessels in rats. Am. J. Physiol. 1999, 277, H1453–H1456. [Google Scholar] [CrossRef]
  144. Gasheva, O.; Zawieja, D.; Gashev, A. Contraction-initiated NO-dependent lymphatic relaxation: A self-regulatory mechanism in rat thoracic duct. J. Physiol. 2006, 575, 821–832. [Google Scholar] [CrossRef]
  145. Kesler, C.; Liao, S.; Munn, L.; Padera, T. Lymphatic vessels in health and disease. Wiley Interdiscip. Rev. Syst. Biol. Med. 2013, 1, 111–124. [Google Scholar] [CrossRef] [Green Version]
  146. Fehervari, Z. Brain lymphatic (dys)function. Nat. Immunol. 2018, 19, 901. [Google Scholar] [CrossRef] [PubMed]
  147. Guo, X.; Zhang, G.; Peng, Q.; Huang, L.; Zhang, Z.; Zhang, Z. Emerging Roles of Meningeal Lymphatic Vessels in Alzheimer’s Disease. J. Alzheimers Dis. 2023, 18, 1–12. [Google Scholar] [CrossRef] [PubMed]
  148. Graham, M.; Mellinghoff, I. Meningeal lymphatics prime tumor immunity in glioblastoma. Cancer Cell 2021, 3, 304–306. [Google Scholar] [CrossRef] [PubMed]
  149. Li, X.; Qi, L.; Yang, D.; Hao, S.; Zhang, F.; Zhu, X.; Sun, Y.; Chen, C.; Ye, J.; Yang, J.; et al. Meningeal lymphatic vessels mediate neurotropic viral drainage from the central nervous system. Nat. Neurosci. 2022, 25, 577–587. [Google Scholar] [CrossRef]
  150. Lan, Y.; Wang, H.; Chen, A.; Zhang, J. Update on the current knowledge of lymphatic drainage system and its emerging roles in glioma management. Immunology 2023, 168, 233–247. [Google Scholar] [CrossRef]
  151. Song, E.; Mao, T.; Dong, H.; Boisserand, L.S.B.; Antila, S.; Bosenberg, M.; Alitalo, K.; Thomas, J.L.; Iwasaki, A. VEGF-C-driven lymphatic drainage enables immunosurveillance of brain tumours. Nature 2020, 577, 689–694. [Google Scholar] [CrossRef] [PubMed]
  152. Tedford, C.; DeLapp, S.; Jacques, S.; Anders, J. Quantitative analysis of transcranial and intraparenchymal light penetration in human cadaver brain tissue. Lasers Surg. Med. 2015, 47, 312–322. [Google Scholar] [CrossRef]
  153. Wang, P.; Li, T. Which wavelength is optimal for transcranial low-level laser stimulation? J. Biophotonics 2018, 12, e201800173. [Google Scholar] [CrossRef] [PubMed]
  154. Semyachkina-Glushkovskaya, O.; Terskov, A.; Khorovodov, A.; Telnova, V.; Blokhina, I.; Saranceva, E.; Kurths, J. Photodynamic Opening of the Blood–Brain Barrier and the Meningeal Lymphatic System: The New Niche in Immunotherapy for Brain Tumors. Pharmaceutics 2022, 14, 2612. [Google Scholar] [CrossRef] [PubMed]
  155. Laser Institute of America (Laser Safety Information). Available online: https://www.lia.org/resources/laser-safety-information/laser-safety-standards/ansi-z136-standards/z136-3 (accessed on 29 April 2023).
  156. IEC TR 60825. Available online: https://webstore.iec.ch/publication/63122 (accessed on 25 April 2023).
  157. Hamblin, M.; Huang, Y. Handbook of Photomedicine; CRC Press: Boca Raton, FL, USA, 2013; pp. 55–68. [Google Scholar]
  158. Michael, R.; Hamblin, M. Shining Light on the Head: Photobiomodulation for Brain Disorders. BBA Clin. 2016, 6, 113–124. [Google Scholar]
  159. Shirani, A. Illuminating rationale and uses for light therapy. J. Clin. Sleep Med. 2009, 2, 155–163. [Google Scholar] [CrossRef] [Green Version]
  160. Tafur, J.; Van Wijk, E.; Van Wijk, R.; Mills, P. Biophoton detection and low-intensity light therapy: A potential clinical partnership. Photomed. Laser Surg. 2010, 1, 23–30. [Google Scholar] [CrossRef] [Green Version]
  161. Cassano, P.; Antonio Caldieraro, M.; Norton, R.; Mischoulon, D.; Trinh, N.-H.; Nyer, M.; Dording, C.; Hamblin, M.; Campbell, B.; Iosifescu, D. Reported Side Effects, Weight and Blood Pressure, After Repeated Sessions of Transcranial Photobiomodulation. Photomed. Laser Surg. 2019, 37, 651–656. [Google Scholar] [CrossRef]
  162. Formolo, D.; Yu, J.; Lin, K. Leveraging the glymphatic and meningeal lymphatic systems as therapeutic strategies in Alzheimer’s disease: An updated overview of nonpharmacological therapies. Mol. Neurodegener. 2023, 18, 26. [Google Scholar] [CrossRef]
  163. Buss, S.; Fried, P.; Pascual-Leone, A. Therapeutic noninvasive brain stimulation in Alzheimer’s disease and related dementias. Curr. Opin. Neurol. 2019, 32, 292–304. [Google Scholar] [CrossRef]
  164. Menardi, A.; Rossi, S.; Koch, G.; Hampel, H.; Vergallo, A.; Nitsche, M.; Stern, Y.; Borroni, B.; Cappa, S.; Cotelli, M.; et al. Toward noninvasive brain stimulation 2.0 in Alzheimer’s disease. Ageing Res. Rev. 2022, 75, 101555. [Google Scholar] [CrossRef]
  165. Teselink, J.; Bawa, K.; Koo, G.; Sankhe, K.; Liu, C.; Rapoport, M.; Oh, P.; Marzolini, S.; Gallagher, D.; Swardfager, W.; et al. Efficacy of non-invasive brain stimulation on global cognition and neuropsychiatric symptoms in Alzheimer’s disease and mild cognitive impairment: A meta-analysis and systematic review. Ageing Res. Rev. 2021, 72, 101499. [Google Scholar] [CrossRef]
  166. Rajji, T. Transcranial Magnetic and Electrical Stimulation in Alzheimer’s Disease and Mild Cognitive Impairment: A Review of Randomized Controlled Trials. Clin. Pharmacol. Ther. 2019, 106, 776–780. [Google Scholar] [CrossRef] [PubMed]
  167. Klivényi, P.; Must, A. Non-invasive Brain Stimulation in Alzheimer’s Disease and Mild Cognitive Impairment—A State-of-the-Art Review on Methodological Characteristics and Stimulation Parameters. Front. Hum. Neurosci. 2020, 14, 179. [Google Scholar]
  168. Chu, C.; Li, C.; Brunoni, A.; Yang, F.; Tseng, P.; Tu, Y.; Stubbs, B.; Carvalho, A.; Thompson, T.; Yeh, T.C.; et al. Cognitive effects and acceptability of non-invasive brain stimulation on Alzheimer’s disease and mild cognitive impairment: A component network meta-analysis. J. Neurol. Neurosurg. Psychiatry 2021, 92, 195–203. [Google Scholar] [CrossRef] [PubMed]
  169. Šimko, P.; Kent, J.; Rektorova, I. Is non-invasive brain stimulation effective for cognitive enhancement in Alzheimer’s disease? An updated meta-analysis. Clin. Neurophysiol. 2022, 144, 23–40. [Google Scholar] [CrossRef] [PubMed]
  170. Gu, L.; Xu, H.; Qian, F. Effects of Non-Invasive Brain Stimulation on Alzheimer’s Disease. J. Prev. Alzheimers Dis. 2022, 9, 410–424. [Google Scholar] [CrossRef] [PubMed]
  171. Finisguerra, A.; Borgatti, R.; Urgesi, C. Non-invasive Brain Stimulation for the Rehabilitation of Children and Adolescents with Neurodevelopmental Disorders: A Systematic Review. Front. Psychol. 2019, 10, 135. [Google Scholar] [CrossRef] [Green Version]
  172. Mimura, Y.; Nishida, H.; Nakajima, S.; Tsugawa, S.; Morita, S.; Yoshida, K.; Tarumi, R.; Ogyu, K.; Wada, M.; Kurose, S.; et al. Neurophysiological biomarkers using transcranial magnetic stimulation in Alzheimer’s disease and mild cognitive impairment: A systematic review and meta-analysis. Neurosci. Biobehav. Rev. 2021, 121, 47–59. [Google Scholar] [CrossRef]
  173. Koch, G.; Martorana, A.; Caltagirone, C. Transcranial magnetic stimulation: Emerging biomarkers and novel therapeutics in Alzheimer’s disease. Neurosci. Lett. 2020, 719, 134355. [Google Scholar] [CrossRef]
  174. Lin, Y.; Jin, J.; Lv, R.; Luo, Y.; Dai, W.; Li, W. Repetitive transcranial magnetic stimulation increases the brain’s drainage efficiency in a mouse model of Alzheimer’s disease. Acta Neuropathol. Commun. 2021, 9, 102. [Google Scholar] [CrossRef]
  175. Liu, D.; He, X.; Wu, D.; Zhang, Q.; Yang, C.; Liang, F. Continuous theta burst stimulation facilitates the clearance efficiency of the glymphatic pathway in a mouse model of sleep deprivation. Neurosci. Lett. 2017, 653, 189–194. [Google Scholar] [CrossRef]
  176. Li, M.; Jing, Y.; Wu, C.; Li, X.; Liang, F.; Li, G. Continuous theta burst stimulation dilates meningeal lymphatic vessels by in meninges. Neurosci. Lett. 2020, 735, 135197. [Google Scholar] [CrossRef] [PubMed]
  177. Ding, X.; Wang, X.; Xia, D.; Liu, H.; Tian, H.; Fu, Y.; Chen, Y.; Qin, C.; Wang, J.; Xiang, Z.; et al. Impaired meningeal lymphatic drainage in patients with idiopathic Parkinson’s disease. Nat. Med. 2021, 27, 411–418. [Google Scholar] [CrossRef] [PubMed]
  178. Purushothuman, S.; Nandasena, C.; Johnstone, D.; 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. [Google Scholar] [CrossRef] [PubMed]
  179. Xuan, W.; Huang, L.; Hamblin, M. 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. [Google Scholar] [CrossRef] [Green Version]
Figure 1. PRISMA flow diagram depicting the study process. n, the number of records; PRISMA, Preferred Reporting Items for Systematic Reviews and Meta-Analyses.
Figure 1. PRISMA flow diagram depicting the study process. n, the number of records; PRISMA, Preferred Reporting Items for Systematic Reviews and Meta-Analyses.
Ijms 24 10946 g001
Figure 2. PBM of the LDRSB during deep sleep: (a) activation of drainage of the mouse brain during deep sleep along the perivascular space (green, fluorescein isothiocyanate-dextran 70 kDa filled the perivascular spaces); (b) schematic illustration of hat-shaped device for simultaneous wireless EEG monitoring and PBM of the LDRSB; (c,d) animal data demonstrating that PBM of the LDRSB during deep sleep more effectively reduces Aβ accumulation in the mouse brain and restores recognition memory assessed by performance of the novel object recognition test [21].
Figure 2. PBM of the LDRSB during deep sleep: (a) activation of drainage of the mouse brain during deep sleep along the perivascular space (green, fluorescein isothiocyanate-dextran 70 kDa filled the perivascular spaces); (b) schematic illustration of hat-shaped device for simultaneous wireless EEG monitoring and PBM of the LDRSB; (c,d) animal data demonstrating that PBM of the LDRSB during deep sleep more effectively reduces Aβ accumulation in the mouse brain and restores recognition memory assessed by performance of the novel object recognition test [21].
Ijms 24 10946 g002
Figure 3. Schematic illustration of promising strategy of the therapy of AD during deep sleep, including simultaneous EEG monitoring of sleep stages (a) and PBM of LDRSB (b). This innovative technology can be implemented through the creation of software for sending a signal from a wireless sleep-tracking gadget (bracelet, ring, smart watch, etc.) to a PBM device (helmet, cap, bandanas, etc.) [36]. The PBM during deep sleep corresponds to the time of natural night activation of lymphatic clearance of metabolites, wastes, and cellular debris from the brain tissue [21,33,34,100,112,113,114,115] that provides a better therapeutic effect for AD pathology than PBM during the day [21].
Figure 3. Schematic illustration of promising strategy of the therapy of AD during deep sleep, including simultaneous EEG monitoring of sleep stages (a) and PBM of LDRSB (b). This innovative technology can be implemented through the creation of software for sending a signal from a wireless sleep-tracking gadget (bracelet, ring, smart watch, etc.) to a PBM device (helmet, cap, bandanas, etc.) [36]. The PBM during deep sleep corresponds to the time of natural night activation of lymphatic clearance of metabolites, wastes, and cellular debris from the brain tissue [21,33,34,100,112,113,114,115] that provides a better therapeutic effect for AD pathology than PBM during the day [21].
Ijms 24 10946 g003
Table 1. PBM in healthy people.
Table 1. PBM in healthy people.
First Author (Year of Publication) and
References
Wavelength (nm) and Irradiation ParameterTime and Duration of PBMGroups and Number of VolunteersTherapeutic Effects
Barret and Gonzalez-Lima (2013) [80]1064 nm
250 mW/cm2, 60 J/cm2
Transcranial PBM
8 min,
single irradiation
Total 40 healthy volunteers
(n = 20 in the PBM group n = 20 in the sham group)
Improvement of executive functions, attention, and memory 2 weeks after PBM
Blanco et al. (2017) [81]1064 nm
250 mW/cm2, 60 J/cm2
Transcranial PBM
8 min,
single irradiation
Total 30 healthy volunteers
(n = 15 in the PBM group; n = 15 in the sham group)
Improvement learning assessed by the use of the Wisconsin Card Sorting Test
Chan et al. (2019) [82]A mix of 633 nm and 870 nm LEDs
44.4 mW/cm2, 20 J/cm2
Transcranial PBM
7.5 min,
single irradiation
Total 30 healthy volunteers
(n = 15 in the PBM group; n = 15 in the sham group)
Improvement of reaction time and mental flexibility during performance of the Eriksen flanker and category fluency tests
Wu et al. (2012) [83]830 nm
7 mW per diode,
20 J/cm2
Transcranial PBM
10 min,
single irradiation
Total 40 healthy volunteers
(n = 20 in the PBM group; n = 20 in the sham group)
Increase in alpha rhythms and theta activities in the occipital, parietal, and temporal regions
Jahan et al. (2019) [84]850 nm
285 mW/cm2, 60 J/cm2.
The total power was 400 mW with a 1.4 cm2 irradiation area.
Transcranial PBM
2.5 min,
single irradiation
Total 30 healthy volunteers
(n = 15 in the PBM group; n = 15 in the sham group)
Beneficial effect on cognitive performance Improvement in attention and alertness
Table 2. PBM for AD in clinical studies.
Table 2. PBM for AD in clinical studies.
First Author (Year of Publication) and ReferencesWavelength (nm) and Irradiation ParameterTime and Duration of PhototherapyGroups and Number of PatientsTherapeutic Effects
Saltmarche et al.
(2017) [86]
810 nm
14.2 mW/cm2 (transcranial),
10.65 J/cm2 (intranasal)
Transcranial–intranasal PBM.
25 min every day for 12 weeks.
Five participants with
dementia or AD
Improvement
of cognition, functional ability in everyday life
Chao (2019) [87]810 nm
75 mW/cm2,
45 J/cm2
Transcranial PBM.
Once every other day for 20 min for 12 weeks.
Total 8 patients with mild-to-moderate dementia
(n = 4 in the PBM group; n = 4 in the sham group
Improvement cognitive and behavior functions, increase in cerebral perfusion and connectivity between the posterior cingulate cortex and lateral parietal nodes within the default-mode network
Maksimovich (2015 and 2019) [15]Low-energy lasers
in the visible region of the spectrum, 20 mw power
Under local anesthetic, the common femoral artery was catheterized and a thin, flexible fiber-optic (diameter 0.25 to 100 μm) was advanced to the distal sections of the anterior and middle cerebral arteries where PBM was performed, taking 20–40 min in the period from 1 year to 12 years after the first symptoms of ADTotal number 89 with AD
(n = 46 in the PBM group and n = 43 in the sham group)
Improvement in cerebral microcirculation, reduction in dementia and restoration of cognitive functions
Nizamutdinov (2021) [16]1060–1080 nm, 15,000 mW, 23.1 mW/cm2, ~650 cm2 per treatment areaTranscranial PBM,
two 6 min sessions daily for 8 consecutive weeks
Total 60 patients with dementia
n = 47 in the PBM group and n = 13 in the sham group
Positive cognitive, executive, and mood changes. Improvement in quality of life.
Zomorrodi (2017)
[90]
810 nm wavelength,
40 Hz
Transcranial PBM,
every 20 min once a night, 6 nights a week for 17 weeks
One patient with moderate ADSignificant improvement in cognition.
Outcomes were rapid and significant, noticeable within days, continuous, and
sustained over 3 weeks.
Berman (2017) [92]1100 LEDs set in 15 arrays of 70 LEDs/array with all matched to 1060–1080 nm, 10 Hz with a 50% duty cycleTranscranial PBM,
6 min daily over 4 weeks
Total 11 patients with dementia
(n = 6 in the PBM group; n = 5 in the sham group)
No significant differences between the PBM group and the control group.
A trend of improvement in executive functioning; praxis memory, visual attention, and task switching
Salehpour et al. (2019) [91]A mix of 635 nm and 810 nm LEDs, 10 HzTranscranial–intranasal PBM,
25 min per session twice daily (morning and evening) for 4 weeks
One patient with cognitive impairment and olfactory dysfunctionCognitive enhancement and reversal of olfactory dysfunction
Horner et al. (2020) [94]810 nm, 40 Hz; 50% duty cycle
25 mW/cm2 (the nasal applicator)
100 mW/cm2 (three posterior LEDs) and 75 mW/cm2 (the anterior LED)
Transcranial–intranasal PBM, 25 min per session for 10 weeksOne patient with mild AD and type 2 diabetesImprovement in cognition and restoration of mitochondrial function
The following clinial studies of PBM for AD have been registered on the clinicaltrials.gov website
Chao (2022) [88]810 nm at 40 Hz
40–150 mW/cm2
Transcranial and intranasal
Once every other day for 20 min for 16 weeks
Total 14 patients with mild-to-moderate AD
(n = 7 in the PBM group; n = 7 in the sham group)
Reduction in the CSF levels of Aβ42, tau, and neurofilament light chain
Lah (2020) [89]Flickering lights at gamma frequencyOne hour per day for 8 weeksTotal 10 patients with mild-to-moderate AD
(n = 5 in the Friker group; n = 7 in the sham group)
Improvement in blood flow and reduction in the CSF levels of Aβ42
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Semyachkina-Glushkovskaya, O.; Penzel, T.; Poluektov, M.; Fedosov, I.; Tzoy, M.; Terskov, A.; Blokhina, I.; Sidorov, V.; Kurths, J. Phototherapy of Alzheimer’s Disease: Photostimulation of Brain Lymphatics during Sleep: A Systematic Review. Int. J. Mol. Sci. 2023, 24, 10946. https://doi.org/10.3390/ijms241310946

AMA Style

Semyachkina-Glushkovskaya O, Penzel T, Poluektov M, Fedosov I, Tzoy M, Terskov A, Blokhina I, Sidorov V, Kurths J. Phototherapy of Alzheimer’s Disease: Photostimulation of Brain Lymphatics during Sleep: A Systematic Review. International Journal of Molecular Sciences. 2023; 24(13):10946. https://doi.org/10.3390/ijms241310946

Chicago/Turabian Style

Semyachkina-Glushkovskaya, Oxana, Thomas Penzel, Mikhail Poluektov, Ivan Fedosov, Maria Tzoy, Andrey Terskov, Inna Blokhina, Viktor Sidorov, and Jürgen Kurths. 2023. "Phototherapy of Alzheimer’s Disease: Photostimulation of Brain Lymphatics during Sleep: A Systematic Review" International Journal of Molecular Sciences 24, no. 13: 10946. https://doi.org/10.3390/ijms241310946

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop