Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Front Neurosci ; 16: 1006031, 2022.
Article in English | MEDLINE | ID: mdl-36203812

ABSTRACT

Background: Neuroinflammation is a response that involves different cell lineages of the central nervous system, such as neurons and glial cells. Among the non-pharmacological interventions for neuroinflammation, photobiomodulation (PBM) is gaining prominence because of its beneficial effects found in experimental brain research. We systematically reviewed the effects of PBM on laboratory animal models, specially to investigate potential benefits of PBM as an efficient anti-inflammatory therapy. Methods: We conducted a systematic search on the bibliographic databases (PubMed and ScienceDirect) with the keywords based on MeSH terms: photobiomodulation, low-level laser therapy, brain, neuroinflammation, inflammation, cytokine, and microglia. Data search was limited from 2009 to June 2022. We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. The initial systematic search identified 140 articles. Among them, 54 articles were removed for duplication and 59 articles by screening. Therefore, 27 studies met the inclusion criteria. Results: The studies showed that PBM has anti-inflammatory properties in several conditions, such as traumatic brain injury, edema formation and hyperalgesia, ischemia, neurodegenerative conditions, aging, epilepsy, depression, and spinal cord injury. Conclusion: Taken together, these results indicate that transcranial PBM therapy is a promising strategy to treat brain pathological conditions induced by neuroinflammation.

2.
Int J Mol Sci ; 23(6)2022 Mar 10.
Article in English | MEDLINE | ID: mdl-35328396

ABSTRACT

The glymphatic system is a glial-dependent waste clearance pathway in the central nervous system, devoted to drain away waste metabolic products and soluble proteins such as amyloid-beta. An impaired brain glymphatic system can increase the incidence of neurovascular, neuroinflammatory, and neurodegenerative diseases. Photobiomodulation (PBM) therapy can serve as a non-invasive neuroprotective strategy for maintaining and optimizing effective brain waste clearance. In this review, we discuss the crucial role of the glymphatic drainage system in removing toxins and waste metabolites from the brain. We review recent animal research on the neurotherapeutic benefits of PBM therapy on glymphatic drainage and clearance. We also highlight cellular mechanisms of PBM on the cerebral glymphatic system. Animal research has shed light on the beneficial effects of PBM on the cerebral drainage system through the clearance of amyloid-beta via meningeal lymphatic vessels. Finally, PBM-mediated increase in the blood-brain barrier permeability with a subsequent rise in Aß clearance from PBM-induced relaxation of lymphatic vessels via a vasodilation process will be discussed. We conclude that PBM promotion of cranial and extracranial lymphatic system function might be a promising strategy for the treatment of brain diseases associated with cerebrospinal fluid outflow abnormality.


Subject(s)
Glymphatic System , Low-Level Light Therapy , Neurodegenerative Diseases , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Glymphatic System/metabolism , Lymphatic System/metabolism , Neurodegenerative Diseases/metabolism
4.
J Alzheimers Dis ; 83(4): 1431-1452, 2021.
Article in English | MEDLINE | ID: mdl-33935090

ABSTRACT

BACKGROUND: Photobiomodulation (PBM) involves the use of red and/or near-infrared light from lasers or LEDs to improve a wide range of medical disorders. Transcranial PBM, sometimes accompanied by intranasal PBM, has been tested to improve many brain disorders, including dementia. OBJECTIVE: To conduct a systematic review according to PRISMA guidelines of pre-clinical and clinical studies reporting the use of PBM, which were considered relevant to dementia. METHODS: Literature was searched between 1967 and 2020 using a range of keywords relevant to PBM and dementia. The light source and wavelength(s), output power, irradiance, irradiation time, fluence or total energy (dose), operation mode (continuous or pulsed) irradiation, approach and site, number of treatment sessions, as well as study outcome(s) were extracted. RESULTS: Out of 10,473 initial articles, 36 studies met the inclusion criteria. Nine articles reported in vitro studies, 17 articles reported studies in animal models of dementia, and 10 studies were conducted in dementia patients. All of the included studies reported positive results. The clinical studies were limited by the small number of patients, lack of placebo controls in some instances, and only a few used objective neuroimaging methods. CONCLUSION: The preliminary evidence of clinical benefit, the lack of any adverse effects, and the remarkable ease of use, suggest larger clinical trials should be conducted as soon as possible.


Subject(s)
Clinical Studies as Topic , Dementia/therapy , Low-Level Light Therapy , Models, Animal , Animals , Humans
5.
Biomolecules ; 10(4)2020 04 15.
Article in English | MEDLINE | ID: mdl-32326425

ABSTRACT

Photobiomodulation (PBM) might be an effective treatment for Parkinson's disease (PD) in human patients. PBM of the brain uses red or near infrared light delivered from a laser or an LED at relatively low power densities, onto the head (or other body parts) to stimulate the brain and prevent degeneration of neurons. PD is a progressive neurodegenerative disease involving the loss of dopamine-producing neurons in the substantia nigra deep within the brain. PD is a movement disorder that also shows various other symptoms affecting the brain and other organs. Treatment involves dopamine replacement therapy or electrical deep brain stimulation. The present systematic review covers reports describing the use of PBM to treat laboratory animal models of PD, in an attempt to draw conclusions about the best choice of parameters and irradiation techniques. There have already been clinical trials of PBM reported in patients, and more are expected in the coming years. PBM is particularly attractive as it is a non-pharmacological treatment, without any major adverse effects (and very few minor ones).


Subject(s)
Disease Models, Animal , Low-Level Light Therapy , Parkinson Disease/radiotherapy , Animals , Humans , Publications
6.
Lasers Med Sci ; 35(3): 573-584, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31372913

ABSTRACT

The effectiveness of transcranial photobiomodulation (tPBM) and methylene Blue (MB) in treating learning and memory impairments is previously reported. In this study, we investigated the effect of tPBM and MB in combination or alone on unpredictable chronic mild stress (UCMS)-induced learning and memory impairments in mice. Fifty-five male BALB/c mice were randomly allocated to five groups: control, laser sham + normal saline (NS), tPBM + NS, laser sham + MB, and tPBM + MB. All groups except the control underwent UCMS and were treated simultaneously for 4 weeks. Elevated plus maze (EPM) was used to evaluate anxiety-like behaviors. Novel object recognition (NOR) test and Barnes maze tests were used to evaluate learning and memory function. The serum cortisol and brain nitric oxide (NO), reactive oxygen species (ROS), total antioxidant capacity (TAC), glutathione peroxidase (GPx), and superoxide dismutase (SOD) levels were measured by spectrophotometric methods. Behavioral tests revealed that UCMS impaired learning and memory, and treatment with PBM, MB, and their combination reversed these impairments. Levels of NO, ROS, SOD activity in brain, and serum cortisol levels significantly increased while brain GPx activity and total antioxidant capacity significantly decreased in the sham + NS animals when compared with the controls. A significant improvement was observed in treatment groups due to reversion of the aforementioned molecular analysis caused by UCMS when it was compared with control levels. Both tPBM and MB in combination or alone have significant therapeutic effects on learning and memory impairments in UCMS-received animals.


Subject(s)
Behavior, Animal/drug effects , Behavior, Animal/radiation effects , Low-Level Light Therapy , Methylene Blue/pharmacology , Skull , Animals , Antioxidants/metabolism , Brain/drug effects , Brain/metabolism , Brain/physiology , Brain/radiation effects , Disease Models, Animal , Glutathione Peroxidase/metabolism , Male , Maze Learning/drug effects , Maze Learning/radiation effects , Memory/drug effects , Memory/radiation effects , Mice , Mice, Inbred BALB C , Reactive Oxygen Species/metabolism , Recognition, Psychology/drug effects , Recognition, Psychology/radiation effects , Superoxide Dismutase/metabolism
7.
Rev Neurosci ; 31(3): 269-286, 2020 Apr 28.
Article in English | MEDLINE | ID: mdl-31812948

ABSTRACT

The application of photobiomodulation therapy (PBMT) for neuronal stimulation is studied in different animal models and in humans, and has shown to improve cerebral metabolic activity and blood flow, and provide neuroprotection via anti-inflammatory and antioxidant pathways. Recently, intranasal PBMT (i-PBMT) has become an attractive and potential method for the treatment of brain conditions. Herein, we provide a summary of different intranasal light delivery approaches including a nostril-based portable method and implanted deep-nasal methods for the effective systemic or direct irradiation of the brain. Nostril-based i-PBMT devices are available, using either lasers or light emitting diodes (LEDs), and can be applied either alone or in combination to transcranial devices (the latter applied directly to the scalp) to treat a wide range of brain conditions such as mild cognitive impairment, Alzheimer's disease, Parkinson's disease, cerebrovascular diseases, depression and anxiety as well as insomnia. Evidence shows that nostril-based i-PBMT improves blood rheology and cerebral blood flow, so that, without needing to puncture blood vessels, i-PBMT may have equivalent results to a peripheral intravenous laser irradiation procedure. Up to now, no studies were conducted to implant PBMT light sources deep within the nose in a clinical setting, but simulation studies suggest that deep-nasal PBMT via cribriform plate and sphenoid sinus might be an effective method to deliver light to the ventromedial part of the prefrontal and orbitofrontal cortex. Home-based i-PBMT, using inexpensive LED applicators, has potential as a novel approach for neurorehabilitation; comparative studies also testing sham, and transcranial PBMT are warranted.


Subject(s)
Low-Level Light Therapy/methods , Mental Disorders/therapy , Neurodegenerative Diseases/therapy , Cerebrovascular Circulation , Humans , Low-Level Light Therapy/instrumentation , Nose
8.
Photobiomodul Photomed Laser Surg ; 37(10): 635-643, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31549906

ABSTRACT

Background: Transcranial photobiomodulation (t-PBM) is a noninvasive modality that may improve cognitive function in both healthy and diseased subjects. Objective: This systematic review and meta-analysis addresses the question of whether t-PBM improves cognitive function in healthy adults. Methods: We searched MEDLINE using PubMed, EMBASE, SCOPUS, Web of Science, and Cochrane Library up to March 2019. We also searched ProQuest and Google Scholar databases for unpublished material. The search was limited to articles on the procognitive effects of t-PBM in healthy adults. The initial search resulted in 871 studies, of which nine publications met our criteria for inclusion and exclusion. Seven studies were performed on young, healthy subjects (17-35 years), and two studies were conducted on older (≥49 years), normal subjects. A meta-analysis was performed on six full-text publications whose subjects were young adults. Results: t-PBM administration improved cognition-related outcomes by an 0.833 standardized mean difference (SMD; 95% confidence interval (CI): 0.458-1.209, 14 comparisons) in young, healthy participants. Funnel plotting revealed asymmetry, which was validated using Egger's (p = 0.030) and Begg's regression (p = 0.006) tests. However after reanalysis, this asymmetry disappeared in the attention subgroup, but not in the memory subgroup. The trim-and-fill analysis indicated two studies were lacking required data. Thus, the effect size was adjusted from an SMD of 0.761 (95% CI: 0.573-0.949) to 0.949 (0.779-1.120). The overall quality score of the studies was modest. Conclusions: We demonstrated a significant, beneficial effect of t-PBM on cognitive performance of young, healthy individuals; however, the heterogeneity of the data was high. This could be due to the modest quality or to the low number of included studies, or to the differences between the various subdomains assessed. These shortcomings should be meticulously addressed before concluding that t-PBM is a cognitive-enhancing intervention in healthy individuals.


Subject(s)
Brain/radiation effects , Cognition/radiation effects , Low-Level Light Therapy/methods , Skull/radiation effects , Cognition/physiology , Cohort Studies , Female , Healthy Volunteers , Humans , Male , Quality Improvement , Reference Values , Retrospective Studies , Young Adult
9.
Photobiomodul Photomed Laser Surg ; 37(10): 581-595, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31553265

ABSTRACT

Background and objective: Photobiomodulation (PBM) therapy is a promising and noninvasive approach to stimulate neuronal function and improve brain repair. The optimization of PBM parameters is important to maximize effectiveness and tolerability. Several studies have reported on the penetration of visible-to-near-infrared (NIR) light through various animal and human tissues. Scientific findings on the penetration of PBM light vary, likely due to use of different irradiation parameters and to different characteristics of the subject such as species, age, and gender. Materials and methods: In this article, we review published data on PBM penetration through the tissues of the head in both animal and human species. The patterns of visible-to-NIR light penetration are summarized based on the following study specifications: wavelength, coherence, operation mode, beam type and size, irradiation site, species, age, and gender. Results: The average penetration of transcranial red/NIR (630-810 nm) light ranged 60-70% in C57BL/6 mouse (skull), 1-10% in BALB/c mouse (skull), 10-40% in Sprague-Dawley rats (scalp plus skull), 20% in Oryctolagus cuniculus rabbit (skull), 0.11% in pig (scalp plus skull), and 0.2-10% in humans (scalp plus skull). The observed variation in the reported values is due to the difference in factors (e.g., wavelengths, light coherence, tissue thickness, and anatomic irradiation site) used by researchers. It seems that these data challenge the applicability of the animal model data on transcranial PBM to humans. Nevertheless, two animal models seem particularly promising, as they approximate penetration in humans: (I) Penetration of 808 nm laser through the scalp plus skull was 0.11% in the pig head; (II) Penetration of 810 nm laser through intact skull was 1.75% in BALB/c mouse. Conclusions: In conclusion, it is worthwhile mentioning that since the effectiveness of brain PBM is closely dependent on the amount of light energy reaching the target neurons, further quantitative estimation of light penetration depth should be performed to validate the current findings.


Subject(s)
Brain/radiation effects , Lasers, Semiconductor/therapeutic use , Lasers, Solid-State/therapeutic use , Low-Level Light Therapy/methods , Skull/radiation effects , Animals , Cohort Studies , Dose-Response Relationship, Radiation , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Rabbits , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity
10.
Photobiomodul Photomed Laser Surg ; 37(10): 596-605, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31335302

ABSTRACT

Brain photobiomodulation (PBM) describes the use of visible to near-infrared light for modulation or stimulation of the central nervous system in both healthy individuals and diseased conditions. Although the transcranial approach to delivering light to the head is the most common technique to stimulate the brain, delivery of light to deeper structures in the brain is still a challenge. The science of nanoparticle engineering in combination with biophotonic excitation could provide a way to overcome this problem. Upconversion is an anti-Stokes process that is capable of transforming low energy photons that penetrate tissue well to higher energy photons with a greater biological effect, but poor tissue penetration. Wavelengths in the third optical window are optimal for light penetration into brain tissue, followed by windows II, IV, and I. The combination of trivalent lanthanide ions within a crystalline host provides a nanostructure that exhibits the upconversion phenomenon. Upconverting nanoparticles (UCNPs) have been successfully used in various medical fields. Their ability to cross the brain-blood barrier and their low toxicity make them a good candidate for application in brain disorders. It is possible that delivery of UCNPs to the brainstem or deeper parts of the cerebral tissue, followed by irradiation using light wavelengths with good tissue penetration properties, could allow more efficient PBM of the brain.


Subject(s)
Brain/radiation effects , Lanthanoid Series Elements/chemistry , Low-Level Light Therapy/methods , Nanoparticles/radiation effects , Animals , Brain/metabolism , Female , Humans , Lanthanoid Series Elements/radiation effects , Male , Nanoparticles/chemistry , Sensitivity and Specificity
11.
Photobiomodul Photomed Laser Surg ; 37(3): 159-167, 2019 Mar.
Article in English | MEDLINE | ID: mdl-31050946

ABSTRACT

Objective: We present a case report of reversal of cognitive impairment, olfactory dysfunction, and quality of life measures in a patient with cognitive decline after multi-modality photobiomodulation (PBM) therapy. Background: Transcranial and intranasal PBM has been introduced as a light-based therapeutic technique in which exposure to low levels of red to near-infrared (NIR) light stimulates neuronal function, leading to beneficial neurological effects. Materials and methods: Patient received twice-daily PBM therapy at home using three different wearable light-emitting diode (LED) devices. For the first week containing a mixture of continuous wave mode red (635 nm) and NIR (810 nm) LEDs, a prototype transcranial light helmet and a body pad were used. The body pad was placed on various areas on the lower back and the helmet was worn while seated. After the first week of treatment, an intranasal LED device, 10-Hz pulsed wave mode NIR (810 nm), was initiated in the left nostril twice daily. All three devices were applied simultaneously for an irradiation time of 25 min per session. Results: The patient showed a significant improvement in the Montreal Cognitive Assessment score from 18 to 24 and in the Working Memory Questionnaire score from 53 to 10. The cognitive enhancement was accompanied by reversal of olfactory dysfunction as measured by the Alberta Smell Test and peanut butter odor detection test. Quality-of-life measures improved and caregiver stress was reduced. No adverse effects were reported. Conclusions: PBM therapy may be a promising noninvasive approach for patients with neurodegenerative diseases.


Subject(s)
Alzheimer Disease/psychology , Alzheimer Disease/radiotherapy , Cognitive Dysfunction/therapy , Low-Level Light Therapy , Olfaction Disorders/therapy , Quality of Life , Alzheimer Disease/complications , Cognitive Dysfunction/etiology , Female , Humans , Middle Aged , Olfaction Disorders/etiology
12.
Front Cell Neurosci ; 13: 74, 2019.
Article in English | MEDLINE | ID: mdl-30983970

ABSTRACT

Disturbances in mitochondrial biogenesis and bioenergetics, combined with neuroinflammation, play cardinal roles in the cognitive impairment during aging that is further exacerbated by transient cerebral ischemia. Both near-infrared (NIR) photobiomodulation (PBM) and Coenzyme Q10 (CoQ10) administration are known to stimulate mitochondrial electron transport that potentially may reverse the effects of cerebral ischemia in aged animals. We tested the hypothesis that the effects of PBM and CoQ10, separately or in combination, improve cognition in a mouse model of transient cerebral ischemia superimposed on a model of aging. We modeled aging by 6-week administration of D-galactose (500 mg/kg subcutaneous) to mice. We subsequently induced transient cerebral ischemia by bilateral occlusion of the common carotid artery (BCCAO). We treated the mice with PBM (810 nm transcranial laser) or CoQ10 (500 mg/kg by gavage), or both, for 2 weeks after surgery. We assessed cognitive function by the Barnes and Lashley III mazes and the What-Where-Which (WWWhich) task. PBM or CoQ10, and both, improved spatial and episodic memory in the mice. Separately and together, the treatments lowered reactive oxygen species and raised ATP and general mitochondrial activity as well as biomarkers of mitochondrial biogenesis, including SIRT1, PGC-1α, NRF1, and TFAM. Neuroinflammatory responsiveness declined, as indicated by decreased iNOS, TNF-α, and IL-1ß levels with the PBM and CoQ10 treatments. Collectively, the findings of this preclinical study imply that the procognitive effects of NIR PBM and CoQ10 treatments, separately or in combination, are beneficial in a model of transient global brain ischemia superimposed on a model of aging in mice.

13.
Lasers Med Sci ; 34(6): 1193-1200, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31011865

ABSTRACT

The aim of the present study was to investigate the electrophysiological effects of the photobiomodulation (PBM) by the quantitative electroencephalography (qEEG) as a diagnostic method. The neurotherapeutic potential of transcranial PBM has been recently investigated in preclinical and clinical studies. According to the PBM mechanisms of action on increasing the cerebral blood flow and the neuronal firing, a change may occur in cortical electrical activity after transcranial PBM that could be revealed in qEEG. A total of 30 participants (15 males and 15 females) were included in this experimental study in a convenience sampling method. A 19-channel EEG was obtained from subjects, before and after receiving sham or real 850-nm PBM by light emitting diode (LED) array on the right prefrontal cortex (PFC). An attentional task also was completed by the participant before and after the irradiation. Results presented that the effect of PBM on the reaction time was significant (p = 0.001) in favor of the real-treatment group (p < 0.05). For the absolute power, repeated-measures ANOVA showed a significant interaction of group × time × frequency (p = 0.04). In the real-treatment group, absolute power of delta band was significantly reduced in all electrodes (p < 0.05). Also, a similar significant interaction of group × time × frequency was seen for relative power (p = 0.04). Post-hoc analysis showed a significant decrease in delta band after PBM in the real treatment group (p < 0.05). The study presented that light irradiation with 850-nm LED source on right PFC could change brain electrical activity and has beneficial effects on attentional performance.


Subject(s)
Attention/physiology , Brain/physiology , Brain/radiation effects , Electrophysiological Phenomena , Infrared Rays , Low-Level Light Therapy , Adult , Behavior , Delta Rhythm , Electrodes , Electroencephalography , Female , Humans , Male , Reaction Time , Task Performance and Analysis , Young Adult
14.
Brain Res Bull ; 144: 213-222, 2019 01.
Article in English | MEDLINE | ID: mdl-30385146

ABSTRACT

This study was aimed to evaluate the effects of near-infrared (NIR) photobiomodulation (PBM) combined with coenzyme Q10 (CoQ10) on depressive-like behavior, cerebral oxidative stress, inflammation, and apoptosis markers in mice. To induce a depressive-like model, mice were subjected to sub-chronic restraint stress for 5 consecutive days. NIR PBM (810 nm laser, 33.3 J/cm2) and/or CoQ10 (500 mg/kg/day, gavage) were administered for five days concomitantly with immobilization. Behavior was evaluated by the forced swim test (FST), tail suspension test (TST), and open field test (OFT). Mitochondrial membrane potential as well as oxidative stress, neuroinflammatory, and markers of apoptosis were evaluated in the prefrontal cortex (PFC) and hippocampus (HIP). The serum levels of pro-inflammatory cytokines, cortisol, and corticosterone were also measured. PBM or CoQ10, or the combination, ameliorated depressive-like behaviors induced by restraint stress as indicated by decreased immobility time in both the FST and TST. PBM and/or CoQ10 treatments decreased lipid peroxidation and enhanced total antioxidant capacity (TAC), GSH levels, GPx and SOD activities in both brain areas. The neuroinflammatory response in the HIP and PFC was suppressed, as indicated by decreased NF-kB, p38, and JNK levels in PBM and/or CoQ10 groups. Intrinsic apoptosis biomarkers, BAX, Bcl-2, cytochrome c release, and caspase-3 and -9, were also significantly down-regulated by both treatments. Furthermore, both treatments decreased the elevated serum levels of cortisol, corticosterone, TNF-α, and IL-6 induced by restraint stress. Transcranial NIR PBM and CoQ10 therapies may be effective antidepressant strategies for the prevention of psychopathological and behavioral symptoms induced by stress.


Subject(s)
Depression/therapy , Stress, Psychological/therapy , Ubiquinone/analogs & derivatives , Animals , Antidepressive Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Behavior, Animal/drug effects , Depression/chemically induced , Depressive Disorder/pathology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Low-Level Light Therapy/methods , Male , Mice , Mice, Inbred BALB C , Neuroimmunomodulation/drug effects , Oxidative Stress/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Ubiquinone/metabolism , Ubiquinone/pharmacology
15.
J Vis Exp ; (141)2018 11 18.
Article in English | MEDLINE | ID: mdl-30507909

ABSTRACT

Transcranial photobiomodulation is a potential innovative noninvasive therapeutic approach for improving brain bioenergetics, brain function in a wide range of neurological and psychiatric disorders, and memory enhancement in age-related cognitive decline and neurodegenerative diseases. We describe a laboratory protocol for transcranial photobiomodulation therapy (PBMT) in mice. Aged BALB/c mice (18 months old) are treated with a 660 nm laser transcranially, once daily for 2 weeks. Laser transmittance data shows that approximately 1% of the incident red light on the scalp reaches a 1 mm depth from the cortical surface, penetrating the dorsal hippocampus. Treatment outcomes are assessed by two methods: a Barnes maze test, which is a hippocampus-dependent spatial learning and memory task evaluation, and measuring hippocampal ATP levels, which is used as a bioenergetics index. The results from the Barnes task show an enhancement of the spatial memory in laser-treated aged mice when compared with age-matched controls. Biochemical analysis after laser treatment indicates increased hippocampal ATP levels. We postulate that the enhancement of memory performance is potentially due to an improvement in hippocampal energy metabolism induced by the red laser treatment. The observations in mice could be extended to other animal models since this protocol could potentially be adapted to other species frequently used in translational neuroscience, such as rabbit, cat, dog, or monkey. Transcranial photobiomodulation is a safe and cost-effective modality which may be a promising therapeutic approach in age-related cognitive impairment.


Subject(s)
Aging/physiology , Cognitive Dysfunction/therapy , Hippocampus/physiology , Low-Level Light Therapy/methods , Maze Learning/physiology , Aging/radiation effects , Animals , Cognition/physiology , Cognitive Dysfunction/physiopathology , Hippocampus/radiation effects , Maze Learning/radiation effects , Memory Disorders/physiopathology , Memory Disorders/therapy , Mice , Mice, Inbred BALB C , Organ Culture Techniques , Spatial Memory/physiology
16.
Iran J Basic Med Sci ; 21(8): 781-786, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30186563

ABSTRACT

OBJECTIVES: Chronic stress has been linked to the pathophysiology of mood disorders including anxiety and depression. In this study, we aimed to investigate the effect of troxerutin (TRX), as a flavonol, on stress-induced anxiety and depression. MATERIALS AND METHODS: 56 animals were randomly divided into seven groups (n=8 per group) as follows: control, saline, TRX 50, TRX 150, TRX 300, Diazepam, and Imipramine. Chronic mild stress (CMS) was induced by restraining animals in Plexiglas cylinders for 1 hr each day for 25 consecutive days. Different doses (50, 150, and 300 mg/kg, oral gavage) of troxerutin was gavaged for 14 consecutive days. At the end of treatments, anxiety- and depressive-like behaviors were tested using elevated plus-maze (EPM), open field test (OFT), and forced swimming test (FST). RESULTS: CMS significantly increased immobility (P<0.05) and decreased swimming (P<0.01) time in FST. However, different doses of troxerutin significantly decreased immobility (P<0.01) and increased swimming (P<0.001) time. CMS also significantly (P<0.01) decreased the percentage of open arm entrance (%OAE), whereas troxerutin significantly increased both %OAE and percentage of open arm time (%OAT) in the EPM. Moreover, CMS significantly decreased time spent in the center (P<0.001) and the number of center entrances (P<0.01) in the OFT. However, troxerutin significantly increased time spent in the center and number of the entrances crossing. Furthermore, CMS significantly increased serum cortisol levels and troxerutin decreased it. CONCLUSION: Troxerutin demonstrated anxiolytic- and antidepressant-like activities in rodents, which supports the use of herbal medicine in the mood disorders.

17.
Neuropeptides ; 71: 61-69, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30054019

ABSTRACT

Medial prefrontal cortex (mPFC) ischemia affects post-stroke cognitive outcomes. We aimed to investigate the effects of different doses and routes of cerebrolysin (CBL) on the structural synaptic plasticity and cognitive function after mPFC ischemia in mice. Thence, CBL (1, 2.5 ml/kg/i.p./daily) or (1 ml/kg/i.n./daily), were administrated in photothrombotic mouse model of mPFC ischemia for two weeks. Episodic and spatial memories were assessed by the What-Where-Which (WWWhich) and Barnes tasks. Growth-associated protein 43 (GAP-43), postsynaptic density-95 (PSD-95), and synaptophysin (SYN) levels were measured in the lesioned area using western blot analysis. Dendritic arbors, spine densities, and morphology were assessed via Golgi-Cox staining. Treatment with 2.5 ml/kg/i.p. and 1 ml/kg/i.n. doses attenuated mPFC ischemia-induced episodic and spatial memories impairment. Results showed an obvious increase in the GAP-43, PSD-95 and SYN levels and improvement in the structural synaptic indexes in lesioned area induced by the same doses and routes of CBL. In conclusion, we found that specific doses/routes of CBL have positive effects on the structural synaptic plasticity and cognitive outcomes after mPFC ischemia.


Subject(s)
Amino Acids/administration & dosage , Brain Ischemia/drug therapy , Cognition/drug effects , Neuronal Plasticity/drug effects , Prefrontal Cortex/drug effects , Amino Acids/therapeutic use , Animals , Behavior, Animal/drug effects , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , GAP-43 Protein/metabolism , Male , Maze Learning/drug effects , Mice , Mice, Inbred BALB C , Neurons/drug effects , Neurons/metabolism , Prefrontal Cortex/blood supply , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Spatial Memory/drug effects , Synaptophysin/metabolism
18.
Brain Res ; 1682: 36-43, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29307593

ABSTRACT

Sleep deprivation (SD) causes oxidative stress in the hippocampus and subsequent memory impairment. In this study, the effect of near-infrared (NIR) photobiomodulation (PBM) on learning and memory impairment induced by acute SD was investigated. The mice were subjected to an acute SD protocol for 72 h. Simultaneously, NIR PBM using a laser at 810 nm was delivered (once a day for 3 days) transcranially to the head to affect the entire brain of mice. The Barnes maze and the What-Where-Which task were used to assess spatial and episodic-like memories. The hippocampal levels of antioxidant enzymes and oxidative stress biomarkers were evaluated. The results showed that NIR PBM prevented cognitive impairment induced by SD. Moreover, NIR PBM therapy enhanced the antioxidant status and increased mitochondrial activity in the hippocampus of SD mice. Our findings revealed that hippocampus-related mitochondrial damage and extensive oxidative stress contribute to the occurrence of memory impairment. In contrast, NIR PBM reduced hippocampal oxidative damage, supporting the ability of 810 nm laser light to improve the antioxidant defense system and maintain mitochondrial survival. This confirms that non-invasive transcranial NIR PBM therapy ameliorates hippocampal dysfunction, which is reflected in enhanced memory function.


Subject(s)
Hippocampus/metabolism , Low-Level Light Therapy/methods , Memory Disorders , Oxidative Stress/physiology , Sleep Deprivation/complications , Aldehydes/metabolism , Animals , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Lipid Peroxidation/physiology , Lipid Peroxidation/radiation effects , Male , Malondialdehyde/metabolism , Maze Learning/physiology , Memory Disorders/etiology , Memory Disorders/pathology , Memory Disorders/therapy , Mice , Mice, Inbred BALB C , Mitochondria/pathology , Oxidative Stress/radiation effects , Reactive Oxygen Species/metabolism , Sleep Deprivation/pathology , Thiobarbituric Acid Reactive Substances/metabolism , Time Factors
19.
Mol Neurobiol ; 55(8): 6601-6636, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29327206

ABSTRACT

Brain photobiomodulation (PBM) therapy using red to near-infrared (NIR) light is an innovative treatment for a wide range of neurological and psychological conditions. Red/NIR light is able to stimulate complex IV of the mitochondrial respiratory chain (cytochrome c oxidase) and increase ATP synthesis. Moreover, light absorption by ion channels results in release of Ca2+ and leads to activation of transcription factors and gene expression. Brain PBM therapy enhances the metabolic capacity of neurons and stimulates anti-inflammatory, anti-apoptotic, and antioxidant responses, as well as neurogenesis and synaptogenesis. Its therapeutic role in disorders such as dementia and Parkinson's disease, as well as to treat stroke, brain trauma, and depression has gained increasing interest. In the transcranial PBM approach, delivering a sufficient dose to achieve optimal stimulation is challenging due to exponential attenuation of light penetration in tissue. Alternative approaches such as intracranial and intranasal light delivery methods have been suggested to overcome this limitation. This article reviews the state-of-the-art preclinical and clinical evidence regarding the efficacy of brain PBM therapy.


Subject(s)
Brain/pathology , Low-Level Light Therapy , Animals , Apoptosis , Dose-Response Relationship, Radiation , Humans , Neurogenesis , Oxidative Stress
20.
Neurobiol Aging ; 58: 140-150, 2017 10.
Article in English | MEDLINE | ID: mdl-28735143

ABSTRACT

Mitochondrial function plays a key role in the aging-related cognitive impairment, and photoneuromodulation of mitochondria by transcranial low-level laser therapy (LLLT) may contribute to its improvement. This study focused on the transcranial LLLT effects on the D-galactose (DG)-induced mitochondrial dysfunction, apoptosis, and cognitive impairment in mice. For this purpose, red and near-infrared (NIR) laser wavelengths (660 and 810 nm) at 2 different fluencies (4 and 8 J/cm2) at 10-Hz pulsed wave mode were administrated transcranially 3 d/wk in DG-received (500 mg/kg/subcutaneous) mice model of aging for 6 weeks. Spatial and episodic-like memories were assessed by the Barnes maze and What-Where-Which (WWWhich) tasks. Brain tissues were analyzed for mitochondrial function including active mitochondria, adenosine triphosphate, and reactive oxygen species levels, as well as membrane potential and cytochrome c oxidase activity. Apoptosis-related biomarkers, namely, Bax, Bcl-2, and caspase-3 were evaluated by Western blotting method. Laser treatments at wavelengths of 660 and 810 nm at 8 J/cm2 attenuated DG-impaired spatial and episodic-like memories. Also, results showed an obvious improvement in the mitochondrial function aspects and modulatory effects on apoptotic markers in aged mice. However, same wavelengths at the fluency of 4 J/cm2 had poor effect on the behavioral and molecular indexes in aging model. This data indicates that transcranial LLLT at both of red and NIR wavelengths at the fluency of 8 J/cm2 has a potential to ameliorate aging-induced mitochondrial dysfunction, apoptosis, and cognitive impairment.


Subject(s)
Brain/metabolism , Cognitive Aging , Cognitive Dysfunction/etiology , Cognitive Dysfunction/therapy , Galactose/adverse effects , Low-Level Light Therapy/methods , Mitochondria/metabolism , Mitochondrial Diseases/etiology , Mitochondrial Diseases/radiotherapy , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cognitive Dysfunction/pathology , Cognitive Dysfunction/psychology , Disease Models, Animal , Male , Memory, Episodic , Mice, Inbred BALB C , Mitochondria/physiology , Mitochondria/radiation effects , Mitochondrial Diseases/pathology , Mitochondrial Diseases/psychology , Reactive Oxygen Species/metabolism , Spatial Memory
SELECTION OF CITATIONS
SEARCH DETAIL
...