1
|
Monteiro KKDS, Tomazoni SS, Albuquerque Pontes GM, Teixeira AM, Agra FADA, Alvim CB, Medeiros Brigato SL, Marcos RL, Dellê H, Serra AJ, Leal-Junior ECP. Effects of Short-, Medium-, and Long-Term Treatment Using Photobiomodulation Therapy Combined with Static Magnetic Field in Aging Rats. Biomedicines 2024; 12:990. [PMID: 38790953 PMCID: PMC11117574 DOI: 10.3390/biomedicines12050990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/05/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: We investigated the detrimental and protective effects of short-, medium, and long-term treatment with different doses of photobiomodulation therapy combined with static magnetic field (PBMT-sMF) during the aging process. (2) Methods: Rats were treated for 15, 30, and 60 weeks with 1, 3, 10, and 30 J of PBMT-sMF or a placebo control. In addition, eight young rats were not subjected to any procedure or treatment and were euthanized at six weeks old. Skin, muscle, bone, kidney, liver, and blood samples were analyzed. (3) Results: No differences between the groups in the morphology of the skin, muscle, and bone was observed. Glutamic pyruvic transaminase levels were increased in the placebo group after 30 and 60 weeks. Glutamic oxaloacetic transaminase levels were also increased in the placebo group after 30 weeks. An increase in creatinine in the PBMT-sMF 3, 10, and 30 J groups compared with that in the young control group was observed. No significant difference in urea levels between the groups was noted. Vascular endothelial growth factor increased in the PBMT-sMF 10 and 30 J groups after 15 weeks of treatment and in the PBMT-sMF 3 J after 60 weeks. Finally, vascular endothelial growth factor decreased in the PBMT-sMF 30 J group after 30 weeks of treatment. (4) Conclusions: PBMT-sMF did not have detrimental effects on the skin, muscle, bone, kidney, or liver after short-, medium-, and long-term treatments in aging rats. In addition, PBMT-sMF may have protective effects on the muscle tissue in aging rats after short- and long-term treatment.
Collapse
Affiliation(s)
- Kadma Karênina Damasceno Soares Monteiro
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Post-Graduate Program in Rehabilitation Sciences, Nove de Julho University, São Paulo 01504-001, Brazil
| | | | - Gianna Móes Albuquerque Pontes
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Post-Graduate Program in Rehabilitation Sciences, Nove de Julho University, São Paulo 01504-001, Brazil
| | - Adeilson Matias Teixeira
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Post-Graduate Program in Rehabilitation Sciences, Nove de Julho University, São Paulo 01504-001, Brazil
| | - Fernanda Aparecida de Araújo Agra
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Post-Graduate Program in Rehabilitation Sciences, Nove de Julho University, São Paulo 01504-001, Brazil
| | - Carolina Barros Alvim
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Post-Graduate Program in Rehabilitation Sciences, Nove de Julho University, São Paulo 01504-001, Brazil
| | - Sâmela Lopes Medeiros Brigato
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Post-Graduate Program in Rehabilitation Sciences, Nove de Julho University, São Paulo 01504-001, Brazil
| | - Rodrigo Labat Marcos
- Post-Graduate Program in Biophotonics, Nove de Julho University, São Paulo 01504-001, Brazil
| | - Humberto Dellê
- Post-Graduate Program in Medicine, Nove de Julho University, São Paulo 01504-001, Brazil
| | - Andrey Jorge Serra
- Post-Graduate Program in Cardiology, Federal University of São Paulo, São Paulo 04024-002, Brazil
| | - Ernesto Cesar Pinto Leal-Junior
- Laboratory of Phototherapy and Innovative Technologies in Health (LaPIT), Post-Graduate Program in Rehabilitation Sciences, Nove de Julho University, São Paulo 01504-001, Brazil
- ELJ Consultancy, São Paulo 04076-000, Brazil
| |
Collapse
|
2
|
Chamkouri H, Liu Q, Zhang Y, Chen C, Chen L. Brain photobiomodulation therapy on neurological and psychological diseases. JOURNAL OF BIOPHOTONICS 2024; 17:e202300145. [PMID: 37403428 DOI: 10.1002/jbio.202300145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/06/2023]
Abstract
Photobiomodulation (PBM) therapy is an innovative treatment for neurological and psychological conditions. Complex IV of the mitochondrial respiratory chain can be stimulated by red light, which increases ATP synthesis. In addition, the ion channels' light absorption causes the release of Ca2+, which activates transcription factors and changes gene expression. Neuronal metabolism is improved by brain PBM therapy, which also promotes synaptogenesis and neurogenesis as well as anti-inflammatory. Its depression-treating potential is attracting attention for other conditions, including Parkinson's disease and dementia. Giving enough dosage for optimum stimulation using the transcranial PBM technique is challenging because of the rapidly increasing attenuation of light transmission in tissue. Different strategies like intranasal and intracranial light delivery systems have been proposed to overcome this restriction. The most recent preclinical and clinical data on the effectiveness of brain PBM therapy are studied in this review article.
Collapse
Affiliation(s)
- Hossein Chamkouri
- School of Materials Science and Engineering, Hefei University of Technology, Hefei, China
| | - Qi Liu
- School of Materials Science and Engineering, Hefei University of Technology, Hefei, China
| | - Yuqin Zhang
- Department of Neurology, Anhui No. 2 Provincial People's Hospital, Hefei, China
| | - Changchun Chen
- Department of Neurology, Anhui No. 2 Provincial People's Hospital, Hefei, China
| | - Lei Chen
- School of Materials Science and Engineering, Hefei University of Technology, Hefei, China
- Intelligent manufacturing institute of HFUT, Hefei, China
| |
Collapse
|
3
|
Monteiro F, Carvalho Ó, Sousa N, Silva FS, Sotiropoulos I. Photobiomodulation and visual stimulation against cognitive decline and Alzheimer's disease pathology: A systematic review. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12249. [PMID: 36447479 PMCID: PMC9695760 DOI: 10.1002/trc2.12249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 12/01/2021] [Accepted: 12/15/2021] [Indexed: 11/27/2022]
Abstract
Introduction Given the ineffectiveness of the available drug treatment against Alzheimer disease (AD), light-based therapeutic modalities have been increasingly receiving attention with photobiomodulation (PBM) and, more recently, visual stimulation (VS) being among the most promising approaches. However, the PBM and VS light parameters tested so far, as well as their outcomes, vary a lot with conflicting results being reported. Methods Based on Scopus, PubMed, and Web of Science databases search, this systematic review summarizes, compares, and discusses 43 cell, animal, and human studies of PBM and VS related to cognitive decline and AD pathology. Results Preclinical work suggests that PBM with 640±30-nm light and VS at 40 Hz attenuates Aβ and Tau pathology and improves neuronal and synaptic plasticity with most studies pointing towards enhancement of degradation/clearance mechanisms in the brain of AD animal models. Despite the gap of the translational evidence for both modalities, the few human studies performed so far support the use of PBM at 810-870 nm light pulsing at 40 Hz for improving brain network connectivity and memory in older subjects and AD patients, while 40 Hz VS in humans seems to improve cognition; further clinical investigation is urgently required to clarify the beneficial impact of PBM and VS in AD patients. Discussion This review highlights PBM and VS as promising light-based therapeutic approaches against AD brain neuropathology and related cognitive decline, clarifying the most effective light parameters for further preclinical and clinical testing and use. Highlights Light-based brain stimulation produces neural entrainment and reverts neuronal damageBrain PBM and VS attenuate AD neuropathologyPMB and VS are suggested to improve cognitive performance in AD patients and animal modelsLight stimulation represents a promising therapeutic strategy against neurodegeneration.
Collapse
Affiliation(s)
- Francisca Monteiro
- Center for Microelectromechanical Systems (CMEMS)Campus AzurémUniversity of MinhoGuimarãesPortugal
- ICVS/3B's ‐ PT Government Associate LaboratoryBraga/GuimarãesPortugal
- LABBELS—Associate LaboratoryUniversity of MinhoGuimarãesPortugal
| | - Óscar Carvalho
- Center for Microelectromechanical Systems (CMEMS)Campus AzurémUniversity of MinhoGuimarãesPortugal
- LABBELS—Associate LaboratoryUniversity of MinhoGuimarãesPortugal
| | - Nuno Sousa
- ICVS/3B's ‐ PT Government Associate LaboratoryBraga/GuimarãesPortugal
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of MinhoCampus de GualtarBragaPortugal
| | - Filipe S. Silva
- Center for Microelectromechanical Systems (CMEMS)Campus AzurémUniversity of MinhoGuimarãesPortugal
- LABBELS—Associate LaboratoryUniversity of MinhoGuimarãesPortugal
| | - Ioannis Sotiropoulos
- ICVS/3B's ‐ PT Government Associate LaboratoryBraga/GuimarãesPortugal
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of MinhoCampus de GualtarBragaPortugal
- Institute of Biosciences and ApplicationsNCSR DemokritosAthensGreece
| |
Collapse
|
4
|
Kim H, Kim MJ, Kwon YW, Jeon S, Lee S, Kim C, Choi BT, Shin Y, Hong SW, Shin HK. Benefits of a Skull-Interfaced Flexible and Implantable Multilight Emitting Diode Array for Photobiomodulation in Ischemic Stroke. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104629. [PMID: 35076161 PMCID: PMC9008794 DOI: 10.1002/advs.202104629] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/08/2022] [Indexed: 05/05/2023]
Abstract
Photobiomodulation (PBM) has received attention due to its potential for improving tissue function and enhancing regeneration in stroke. A lightweight, compact, and simple system of miniaturized electronic devices consisting of packaged light-emitting diodes (LEDs) that incorporates a flexible substrate for in vivo brain PBM in a mouse model is developed. Using this device platform, the preventive and therapeutic effects of PBM affixed to the exposed skull of mice in the photothrombosis and middle cerebral artery occlusion stroke model are evaluated. Among the wavelength range of 630, 850, and 940 nm LED array, the PBM with 630-nm LED array is proved to be the most effective for reducing the infarction volume and neurological impairment after ischemic stroke. Moreover, the PBM with 630 nm LED array remarkably improves the capability of spatial learning and memory in the chronic poststroke phase, attenuates AIM2 inflammasome activation and inflammasome-mediated pyroptosis, and modulates microglial polarization in the hippocampus and cortex 7 days following ischemic stroke. Thus, PBM may prevent tissue and functional damage in acute ischemic injury, thereby attenuating the development of cognitive impairment after stroke.
Collapse
Affiliation(s)
- Hyunha Kim
- Department of Korean Medical ScienceGraduate Training Program of Korean Medical Therapeutics for Healthy‐AgingSchool of Korean MedicinePusan National UniversityYangsan50612Republic of Korea
- Department of Pharmacology and NeuroscienceCreighton University School of MedicineOmahaNE68178USA
| | - Min Jae Kim
- Department of Korean Medical ScienceGraduate Training Program of Korean Medical Therapeutics for Healthy‐AgingSchool of Korean MedicinePusan National UniversityYangsan50612Republic of Korea
| | - Young Woo Kwon
- Department of Nano‐Fusion TechnologyCollege of Nanoscience & NanotechnologyPusan National UniversityBusan46241Republic of Korea
| | - Sangheon Jeon
- Department of Congo‐Mechatronics EngineeringDepartment of Optics and Mechatronics EngineeringCollege of Nanoscience & NanotechnologyPusan National UniversityBusan46241Republic of Korea
| | - Seo‐Yeon Lee
- Department of PharmacologyWonkwang University School of MedicineIksan54538Republic of Korea
| | - Chang‐Seok Kim
- Department of Congo‐Mechatronics EngineeringDepartment of Optics and Mechatronics EngineeringCollege of Nanoscience & NanotechnologyPusan National UniversityBusan46241Republic of Korea
| | - Byung Tae Choi
- Department of Korean Medical ScienceGraduate Training Program of Korean Medical Therapeutics for Healthy‐AgingSchool of Korean MedicinePusan National UniversityYangsan50612Republic of Korea
| | - Yong‐Il Shin
- Department of Rehabilitation MedicineSchool of MedicinePusan National UniversityYangsan50612Republic of Korea
| | - Suck Won Hong
- Department of Congo‐Mechatronics EngineeringDepartment of Optics and Mechatronics EngineeringCollege of Nanoscience & NanotechnologyPusan National UniversityBusan46241Republic of Korea
| | - Hwa Kyoung Shin
- Department of Korean Medical ScienceGraduate Training Program of Korean Medical Therapeutics for Healthy‐AgingSchool of Korean MedicinePusan National UniversityYangsan50612Republic of Korea
| |
Collapse
|
5
|
Perez Garcia G, Perez GM, Otero-Pagan A, Abutarboush R, Kawoos U, De Gasperi R, Gama Sosa MA, Pryor D, Hof PR, Cook DG, Gandy S, Ahlers ST, Elder GA. Transcranial Laser Therapy Does Not Improve Cognitive and Post-Traumatic Stress Disorder-Related Behavioral Traits in Rats Exposed to Repetitive Low-Level Blast Injury. Neurotrauma Rep 2021; 2:548-563. [PMID: 34901948 PMCID: PMC8655798 DOI: 10.1089/neur.2021.0005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Many military veterans who experienced blast-related traumatic brain injuries (TBIs) in the conflicts in Iraq and Afghanistan suffer from chronic cognitive and mental health problems, including post-traumatic stress disorder (PTSD). Transcranial laser therapy (TLT) uses low-power lasers emitting light in the far- to near-infrared ranges. Beneficial effects of TLT have been reported in neurological and mental-health-related disorders in humans and animal models, including TBI. Rats exposed to repetitive low-level blast develop chronic cognitive and PTSD-related behavioral traits. We tested whether TLT treatment could reverse these traits. Rats received a 74.5-kPa blast or sham exposures delivered one per day for 3 consecutive days. Beginning at 34 weeks after blast exposure, the following groups of rats were treated with active or sham TLT: 1) Sham-exposed rats (n = 12) were treated with sham TLT; 2) blast-exposed rats (n = 13) were treated with sham TLT; and 3) blast-exposed rats (n = 14) were treated with active TLT. Rats received 5 min of TLT five times per week for 6 weeks (wavelength, 808 nm; power of irradiance, 240 mW). At the end of treatment, rats were tested in tasks found previously to be most informative (novel object recognition, novel object localization, contextual/cued fear conditioning, elevated zero maze, and light/dark emergence). TLT did not improve blast-related effects in any of these tests, and blast-exposed rats were worse after TLT in some anxiety-related measures. Based on these findings, TLT does not appear to be a promising treatment for the chronic cognitive and mental health problems that follow blast injury.
Collapse
Affiliation(s)
- Georgina Perez Garcia
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gissel M. Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Alena Otero-Pagan
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Rania Abutarboush
- Department of Neurotrauma, Naval Medical Research Center, Silver Spring, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Usmah Kawoos
- Department of Neurotrauma, Naval Medical Research Center, Silver Spring, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Rita De Gasperi
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Miguel A. Gama Sosa
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Dylan Pryor
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| | - Patrick R. Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - David G. Cook
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Sam Gandy
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Barbara and Maurice A. Deane Center for Wellness and Cognitive Health and the Mount Sinai NFL Neurological Care Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stephen T. Ahlers
- Department of Neurotrauma, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Gregory A. Elder
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Alzheimer's Disease Research Center and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, New York, USA
| |
Collapse
|
6
|
Baik JS, Lee TY, Kim NG, Pak K, Ko SH, Min JH, Shin YI. Effects of Photobiomodulation on Changes in Cognitive Function and Regional Cerebral Blood Flow in Patients with Mild Cognitive Impairment: A Pilot Uncontrolled Trial. J Alzheimers Dis 2021; 83:1513-1519. [PMID: 34420956 DOI: 10.3233/jad-210386] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Photobiomodulation (PBM) affects local blood flow regulation through nitric oxide generation, and various studies have reported on its effect on improving cognitive function in neurodegenerative diseases. However, the effect of PBM in the areas of the vertebral arteries (VA) and internal carotid arteries (ICA), which are the major blood-supplying arteries to the brain, has not been previously investigated. OBJECTIVE We aimed to determine whether irradiating PBM in the areas of the VA and ICA, which are the major blood-supplying arteries to the brain, improved regional cerebral blood flow (rCBF) and cognitive function. METHODS Fourteen patients with mild cognitive impairments were treated with PBM. Cognitive assessment and single-photon emission computed tomography were implemented at the baseline and at the end of PBM. RESULTS Regarding rCBF, statistically significant trends were found in the medial prefrontal cortex, lateral prefrontal cortex, anterior cingulate cortex, and occipital lateral cortex. Based on the cognitive assessments, statistically significant trends were found in overall cognitive function, memory, and frontal/executive function. CONCLUSION We confirmed the possibility that PBM treatment in the VA and ICA areas could positively affect cognitive function by increasing rCBF. A study with a larger sample size is needed to validate the potential of PBM.
Collapse
Affiliation(s)
- Ji Soo Baik
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Tae Young Lee
- Department of Psychiatry, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Nam Gyun Kim
- Medical Research Center of Color Seven. Seoul, Republic of Korea
| | - Kyoungjune Pak
- Department of Nuclear Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Sung-Hwa Ko
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea.,Department of Rehabilitation Medicine, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Ji Hong Min
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea.,Department of Rehabilitation Medicine, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Yong-Il Shin
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea.,Department of Rehabilitation Medicine & Institute of Medical Science, Pusan National University School of Medicine, Busan, Republic of Korea
| |
Collapse
|
7
|
Foo ASC, Soong TW, Yeo TT, Lim KL. Mitochondrial Dysfunction and Parkinson's Disease-Near-Infrared Photobiomodulation as a Potential Therapeutic Strategy. Front Aging Neurosci 2020; 12:89. [PMID: 32308618 PMCID: PMC7145956 DOI: 10.3389/fnagi.2020.00089] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/17/2020] [Indexed: 12/21/2022] Open
Abstract
As the main driver of energy production in eukaryotes, mitochondria are invariably implicated in disorders of cellular bioenergetics. Given that dopaminergic neurons affected in Parkinson's disease (PD) are particularly susceptible to energy fluctuations by their high basal energy demand, it is not surprising to note that mitochondrial dysfunction has emerged as a compelling candidate underlying PD. A recent approach towards forestalling dopaminergic neurodegeneration in PD involves near-infrared (NIR) photobiomodulation (PBM), which is thought to enhance mitochondrial function of stimulated cells through augmenting the activity of cytochrome C oxidase. Notwithstanding this, our understanding of the neuroprotective mechanism of PBM remains far from complete. For example, studies focusing on the effects of PBM on gene transcription are limited, and the mechanism through which PBM exerts its effects on distant sites (i.e., its "abscopal effect") remains unclear. Also, the clinical application of NIR in PD proves to be challenging. Efficacious delivery of NIR light to the substantia nigra pars compacta (SNpc), the primary site of disease pathology in PD, is fraught with technical challenges. Concerted efforts focused on understanding the biological effects of PBM and improving the efficiency of intracranial NIR delivery are therefore essential for its successful clinical translation. Nonetheless, PBM represents a potential novel therapy for PD. In this review, we provide an update on the role of mitochondrial dysfunction in PD and how PBM may help mitigate the neurodegenerative process. We also discussed clinical translation aspects of this treatment modality using intracranially implanted NIR delivery devices.
Collapse
Affiliation(s)
- Aaron Song Chuan Foo
- Department of Physiology, National University of Singapore, Singapore, Singapore
- Division of Neurosurgery, Department of Surgery, University Surgical Cluster, National University Hospital, Singapore, Singapore
| | - Tuck Wah Soong
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Tseng Tsai Yeo
- Division of Neurosurgery, Department of Surgery, University Surgical Cluster, National University Hospital, Singapore, Singapore
| | - Kah-Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Research, National Neuroscience Institute, Singapore, Singapore
| |
Collapse
|
8
|
Enengl J, Hamblin MR, Dungel P. Photobiomodulation for Alzheimer's Disease: Translating Basic Research to Clinical Application. J Alzheimers Dis 2020; 75:1073-1082. [PMID: 32390621 PMCID: PMC7369090 DOI: 10.3233/jad-191210] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2020] [Indexed: 12/30/2022]
Abstract
One of the challenges in translating new therapeutic approaches to the patient bedside lies in bridging the gap between scientists who are conducting basic laboratory research and medical practitioners who are not exposed to highly specialized journals. This review covers the literature on photobiomodulation therapy as a novel approach to prevent and treat Alzheimer's disease, aiming to bridge that gap by gathering together the terms and technical specifications into a single concise suggestion for a treatment protocol. In light of the predicted doubling in the number of people affected by dementia and Alzheimer's disease within the next 30 years, a treatment option which has already shown promising results in cell culture studies and animal models, and whose safety has already been proven in humans, must not be left in the dark. This review covers the mechanistic action of photobiomodulation therapy against Alzheimer's disease at a cellular level. Safe and effective doses have been found in animal models, and the first human case studies have provided reasons to undertake large-scale clinical trials. A brief discussion of the minimally effective and maximum tolerated dose concludes this review, and provides the basis for a successful translation from bench to bedside.
Collapse
Affiliation(s)
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Peter Dungel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| |
Collapse
|
9
|
Esenaliev RO, Petrov IY, Petrov Y, Guptarak J, Boone DR, Mocciaro E, Weisz H, Parsley MA, Sell SL, Hellmich H, Ford JM, Pogue C, DeWitt D, Prough DS, Micci MA. Nano-Pulsed Laser Therapy Is Neuroprotective in a Rat Model of Blast-Induced Neurotrauma. J Neurotrauma 2018; 35:1510-1522. [PMID: 29562823 PMCID: PMC5998828 DOI: 10.1089/neu.2017.5249] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
We have developed a novel, non-invasive nano-pulsed laser therapy (NPLT) system that combines the benefits of near-infrared laser light (808 nm) and ultrasound (optoacoustic) waves, which are generated with each short laser pulse within the tissue. We tested NPLT in a rat model of blast-induced neurotrauma (BINT) to determine whether transcranial application of NPLT provides neuroprotective effects. The laser pulses were applied on the intact rat head 1 h after injury using a specially developed fiber-optic system. Vestibulomotor function was assessed on post-injury days (PIDs) 1–3 on the beam balance and beam walking tasks. Cognitive function was assessed on PIDs 6–10 using a working memory Morris water maze (MWM) test. BDNF and caspase-3 messenger RNA (mRNA) expression was measured by quantitative real-time PCR (qRT-PCR) in laser-captured cortical neurons. Microglia activation and neuronal injury were assessed in brain sections by immunofluorescence using specific antibodies against CD68 and active caspase-3, respectively. In the vestibulomotor and cognitive (MWM) tests, NPLT-treated animals performed significantly better than the untreated blast group and similarly to sham animals. NPLT upregulated mRNA encoding BDNF and downregulated the pro-apoptotic protein caspase-3 in cortical neurons. Immunofluorescence demonstrated that NPLT inhibited microglia activation and reduced the number of cortical neurons expressing activated caspase-3. NPLT also increased expression of BDNF in the hippocampus and the number of proliferating progenitor cells in the dentate gyrus. Our data demonstrate a neuroprotective effect of NPLT and prompt further studies aimed to develop NPLT as a therapeutic intervention after traumatic brain injury (TBI).
Collapse
Affiliation(s)
- Rinat O Esenaliev
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas.,2 Department of Neuroscience and Cell Biology, University of Texas Medical Branch , Galveston, Texas.,3 Center for Biomedical Engineering, University of Texas Medical Branch , Galveston, Texas
| | - Irene Y Petrov
- 3 Center for Biomedical Engineering, University of Texas Medical Branch , Galveston, Texas
| | - Yuriy Petrov
- 3 Center for Biomedical Engineering, University of Texas Medical Branch , Galveston, Texas
| | - Jutatip Guptarak
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Debbie R Boone
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Emanuele Mocciaro
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Harris Weisz
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Margaret A Parsley
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Stacy L Sell
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Helen Hellmich
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Jonathan M Ford
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Connor Pogue
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Douglas DeWitt
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Donald S Prough
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Maria-Adelaide Micci
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| |
Collapse
|
10
|
Brain Photobiomodulation Therapy: a Narrative Review. Mol Neurobiol 2018; 55:6601-6636. [PMID: 29327206 DOI: 10.1007/s12035-017-0852-4] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/19/2017] [Indexed: 12/20/2022]
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.
Collapse
|
11
|
Moro C, Torres N, Arvanitakis K, Cullen K, Chabrol C, Agay D, Darlot F, Benabid AL, Mitrofanis J. No evidence for toxicity after long-term photobiomodulation in normal non-human primates. Exp Brain Res 2017; 235:3081-3092. [PMID: 28744621 DOI: 10.1007/s00221-017-5048-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/24/2017] [Indexed: 11/28/2022]
Abstract
In this study, we explored the effects of a longer term application, up to 12 weeks, of photobiomodulation in normal, naïve macaque monkeys. Monkeys (n = 5) were implanted intracranially with an optical fibre device delivering photobiomodulation (red light, 670 nm) to a midline midbrain region. Animals were then aldehyde-fixed and their brains were processed for immunohistochemistry. In general, our results showed that longer term intracranial application of photobiomodulation had no adverse effects on the surrounding brain parenchyma or on the nearby dopaminergic cell system. We found no evidence for photobiomodulation generating an inflammatory glial response or neuronal degeneration near the implant site; further, photobiomodulation did not induce an abnormal activation or mitochondrial stress in nearby cells, nor did it cause an abnormal arrangement of the surrounding vasculature (endothelial basement membrane). Finally, because of our interest in Parkinson's disease, we noted that photobiomodulation had no impact on the number of midbrain dopaminergic cells and the density of their terminations in the striatum. In summary, we found no histological basis for any major biosafety concerns associated with photobiomodulation delivered by our intracranial approach and our findings set a key template for progress onto clinical trial on patients with Parkinson's disease.
Collapse
Affiliation(s)
- Cécile Moro
- University of Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000, Grenoble, France
| | - Napoleon Torres
- University of Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000, Grenoble, France
| | | | - Karen Cullen
- Department of Anatomy F13, University of Sydney, Camperdown, 2006, Australia
| | - Claude Chabrol
- University of Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000, Grenoble, France
| | - Diane Agay
- University of Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000, Grenoble, France
| | - Fannie Darlot
- University of Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000, Grenoble, France
| | - Alim-Louis Benabid
- University of Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000, Grenoble, France
| | - John Mitrofanis
- Department of Anatomy F13, University of Sydney, Camperdown, 2006, Australia.
| |
Collapse
|
12
|
Moro C, El Massri N, Darlot F, Torres N, Chabrol C, Agay D, Auboiroux V, Johnstone DM, Stone J, Mitrofanis J, Benabid AL. Effects of a higher dose of near-infrared light on clinical signs and neuroprotection in a monkey model of Parkinson's disease. Brain Res 2016; 1648:19-26. [PMID: 27396907 DOI: 10.1016/j.brainres.2016.07.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/05/2016] [Accepted: 07/06/2016] [Indexed: 11/16/2022]
Abstract
We have reported previously that intracranial application of near-infrared light (NIr) - when delivered at the lower doses of 25J and 35J - reduces clinical signs and offers neuroprotection in a subacute MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) monkey model of Parkinson's disease. In this study, we explored whether a higher NIr dose (125J) generated beneficial effects in the same MPTP monkey model (n=15). We implanted an NIr (670nm) optical fibre device within a midline region of the midbrain in macaque monkeys, close to the substantia nigra of both sides. MPTP injections (1.8-2.1mg/kg) were made over a five day period, during which time the NIr device was turned on and left on continuously throughout the ensuing three week survival period. Monkeys were evaluated clinically and their brains processed for immunohistochemistry and stereology. Our results showed that the higher NIr dose did not have any toxic impact on cells at the midbrain implant site. Further, this NIr dose resulted in a higher number of nigral tyrosine hydroxylase immunoreactive cells when compared to the MPTP group. However, the higher NIr dose monkeys showed little evidence for an increase in mean clinical score, number of nigral Nissl-stained cells and density of striatal tyrosine hydroxylase terminations. In summary, the higher NIr dose of 125J was not as beneficial to MPTP-treated monkeys as compared to the lower doses of 25J and 35J, boding well for strategies of NIr dose delivery and device energy consumption in a future clinical trial.
Collapse
Affiliation(s)
- Cécile Moro
- CLINATEC, EJ Safra Centre, CEA, LETI, University of Grenoble, Alpes F38000, France.
| | | | - Fannie Darlot
- CLINATEC, EJ Safra Centre, CEA, LETI, University of Grenoble, Alpes F38000, France.
| | - Napoleon Torres
- CLINATEC, EJ Safra Centre, CEA, LETI, University of Grenoble, Alpes F38000, France.
| | - Claude Chabrol
- CLINATEC, EJ Safra Centre, CEA, LETI, University of Grenoble, Alpes F38000, France.
| | - Diane Agay
- CLINATEC, EJ Safra Centre, CEA, LETI, University of Grenoble, Alpes F38000, France.
| | - Vincent Auboiroux
- CLINATEC, EJ Safra Centre, CEA, LETI, University of Grenoble, Alpes F38000, France.
| | | | - Jonathan Stone
- Dept of Physiology F13, University of Sydney, 2006, Australia.
| | | | - Alim-Louis Benabid
- CLINATEC, EJ Safra Centre, CEA, LETI, University of Grenoble, Alpes F38000, France.
| |
Collapse
|
13
|
Salehpour F, Rasta SH, Mohaddes G, Sadigh-Eteghad S, Salarirad S. Therapeutic effects of 10-HzPulsed wave lasers in rat depression model: A comparison between near-infrared and red wavelengths. Lasers Surg Med 2016; 48:695-705. [DOI: 10.1002/lsm.22542] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2016] [Indexed: 01/28/2023]
Affiliation(s)
- Farzad Salehpour
- Neurosciences Research Center (NSRC); Tabriz University of Medical Sciences; Tabriz 51666 Iran
- Department of Medical Physics; Tabriz University of Medical Sciences; Tabriz 51666 Iran
| | - Seyed Hossein Rasta
- Neurosciences Research Center (NSRC); Tabriz University of Medical Sciences; Tabriz 51666 Iran
- Department of Medical Bioengineering; Tabriz University of Medical Sciences; Tabriz 51666 Iran
- Department of Medical Physics; Tabriz University of Medical Sciences; Tabriz 51666 Iran
- School of Medical Sciences; University of Aberdeen; Aberdeen AB24 5DT United Kingdom
| | - Gisou Mohaddes
- Neurosciences Research Center (NSRC); Tabriz University of Medical Sciences; Tabriz 51666 Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC); Tabriz University of Medical Sciences; Tabriz 51666 Iran
| | - Sima Salarirad
- School of Medical Sciences; University of Aberdeen; Aberdeen AB24 5DT United Kingdom
- Department of Psychiatry; Tabriz University of Medical Sciences; Tabriz 51666 Iran
| |
Collapse
|
14
|
Johnstone DM, Moro C, Stone J, Benabid AL, Mitrofanis J. Turning On Lights to Stop Neurodegeneration: The Potential of Near Infrared Light Therapy in Alzheimer's and Parkinson's Disease. Front Neurosci 2016; 9:500. [PMID: 26793049 PMCID: PMC4707222 DOI: 10.3389/fnins.2015.00500] [Citation(s) in RCA: 206] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/15/2015] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's and Parkinson's disease are the two most common neurodegenerative disorders. They develop after a progressive death of many neurons in the brain. Although therapies are available to treat the signs and symptoms of both diseases, the progression of neuronal death remains relentless, and it has proved difficult to slow or stop. Hence, there is a need to develop neuroprotective or disease-modifying treatments that stabilize this degeneration. Red to infrared light therapy (λ = 600-1070 nm), and in particular light in the near infrared (NIr) range, is emerging as a safe and effective therapy that is capable of arresting neuronal death. Previous studies have used NIr to treat tissue stressed by hypoxia, toxic insult, genetic mutation and mitochondrial dysfunction with much success. Here we propose NIr therapy as a neuroprotective or disease-modifying treatment for Alzheimer's and Parkinson's patients.
Collapse
Affiliation(s)
| | - Cécile Moro
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus Grenoble, France
| | - Jonathan Stone
- Department of Physiology, University of Sydney Sydney, NSW, Australia
| | - Alim-Louis Benabid
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus Grenoble, France
| | - John Mitrofanis
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus Grenoble, France
| |
Collapse
|
15
|
Safety assessment of trans-tympanic photobiomodulation. Lasers Med Sci 2016; 31:323-33. [PMID: 26738500 DOI: 10.1007/s10103-015-1851-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 12/08/2015] [Indexed: 12/16/2022]
Abstract
We evaluated functional and morphological changes after trans-tympanic laser application using several different powers of photobiomodulation (PBM). The left (L) ears of 17 rats were irradiated for 30 min daily over 14 days using a power density of 909.1 (group A, 5040 J), 1136.4 (group B, 6300 J), and 1363.6 (group C, 7560 J) mW/cm(2). The right (N) ears served as controls. The safety of PBM was determined by endoscopic findings, auditory brainstem response (ABR) thresholds, and histological images of hair cells using confocal microscopy, and light microscopic images of the external auditory canal (EAC) and tympanic membrane (TM). Endoscopic findings revealed severe inflammation in the TM of C group; no other group showed damage in the TM. No significant difference in ABR threshold was found in the PBM-treated groups (excluding the group with TM damage). Confocal microscopy showed no histological difference between the AL and AN, or BL and BN groups. However, light microscopy showed more prominent edema, inflammation, and vascular congestion in the TM of BL ears. This study found a dose-response relationship between laser power parameters and TM changes. These results will be useful for defining future allowance criteria for trans-tympanic laser therapies.
Collapse
|
16
|
Darlot F, Moro C, El Massri N, Chabrol C, Johnstone DM, Reinhart F, Agay D, Torres N, Bekha D, Auboiroux V, Costecalde T, Peoples CL, Anastascio HDT, Shaw VE, Stone J, Mitrofanis J, Benabid AL. Near-infrared light is neuroprotective in a monkey model of Parkinson disease. Ann Neurol 2015; 79:59-75. [PMID: 26456231 DOI: 10.1002/ana.24542] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/07/2015] [Accepted: 10/08/2015] [Indexed: 11/12/2022]
Abstract
OBJECTIVE To examine whether near-infrared light (NIr) treatment reduces clinical signs and/or offers neuroprotection in a subacute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) monkey model of Parkinson disease. METHODS We implanted an optical fiber device that delivered NIr (670 nm) to the midbrain of macaque monkeys, close to the substantia nigra of both sides. MPTP injections (1.5-2.1mg/kg) were made over a 5- to 7-day period, during which time the NIr device was turned on. This was then followed by a 3-week survival period. Monkeys were evaluated clinically (eg, posture, bradykinesia) and behaviorally (open field test), and their brains were processed for immunohistochemistry and stereology. RESULTS All monkeys in the MPTP group developed severe clinical and behavioral impairment (mean clinical scores = 21-34; n = 11). By contrast, the MPTP-NIr group developed much less clinical and behavioral impairment (n = 9); some monkeys developed moderate clinical signs (mean scores = 11-15; n = 3), whereas the majority--quite remarkably--developed few clinical signs (mean scores = 1-6; n = 6). The monkeys that developed moderate clinical signs had hematic fluid in their optical fibers at postmortem, presumably limiting NIr exposure and overall clinical improvement. NIr was not toxic to brain tissue and offered neuroprotection to dopaminergic cells and their terminations against MPTP insult, particularly in animals that developed few clinical signs. INTERPRETATION Our findings indicate NIr to be an effective therapeutic agent in a primate model of the disease and create the template for translation into clinical trials.
Collapse
Affiliation(s)
- Fannie Darlot
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, Grenoble, France
| | - Cécile Moro
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, Grenoble, France
| | - Nabil El Massri
- Departments of Anatomy, University of Sydney, Sydney, New South Wales, Australia
| | - Claude Chabrol
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, Grenoble, France
| | - Daniel M Johnstone
- Departments of Physiology, University of Sydney, Sydney, New South Wales, Australia
| | - Florian Reinhart
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, Grenoble, France
| | - Diane Agay
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, Grenoble, France
| | - Napoleon Torres
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, Grenoble, France
| | - Dhaïf Bekha
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, Grenoble, France
| | - Vincent Auboiroux
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, Grenoble, France
| | - Thomas Costecalde
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, Grenoble, France
| | - Cassandra L Peoples
- Departments of Anatomy, University of Sydney, Sydney, New South Wales, Australia
| | | | - Victoria E Shaw
- Departments of Anatomy, University of Sydney, Sydney, New South Wales, Australia
| | - Jonathan Stone
- Departments of Physiology, University of Sydney, Sydney, New South Wales, Australia
| | - John Mitrofanis
- Departments of Anatomy, University of Sydney, Sydney, New South Wales, Australia
| | - Alim-Louis Benabid
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, Grenoble, France
| |
Collapse
|
17
|
Tedford CE, DeLapp S, Jacques S, Anders J. Quantitative analysis of transcranial and intraparenchymal light penetration in human cadaver brain tissue. Lasers Surg Med 2015; 47:312-22. [PMID: 25772014 DOI: 10.1002/lsm.22343] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2015] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND OBJECTIVE Photobiomodulation (PBM) also known as low-level light therapy has been used successfully for the treatment of injury and disease of the nervous system. The use of PBM to treat injury and diseases of the brain requires an in-depth understanding of light propagation through tissues including scalp, skull, meninges, and brain. This study investigated the light penetration gradients in the human cadaver brain using a Transcranial Laser System with a 30 mm diameter beam of 808 nm wavelength light. In addition, the wavelength-dependence of light scatter and absorbance in intraparenchymal brain tissue using 660, 808, and 940 nm wavelengths was investigated. STUDY DESIGN/MATERIAL AND METHODS Intact human cadaver heads (n = 8) were obtained for measurement of light propagation through the scalp/skull/meninges and into brain tissue. The cadaver heads were sectioned in either the transverse or mid-sagittal. The sectioned head was mounted into a cranial fixture with an 808 nm wavelength laser system illuminating the head from beneath with either pulsed-wave (PW) or continuous-wave (CW) laser light. A linear array of nine isotropic optical fibers on a 5 mm pitch was inserted into the brain tissue along the optical axis of the beam. Light collected from each fiber was delivered to a multichannel power meter. As the array was lowered into the tissue, the power from each probe was recorded at 5 mm increments until the inner aspect of the dura mater was reached. Intraparenchymal light penetration measurements were made by delivering a series of wavelengths (660, 808, and 940 nm) through a separate optical fiber within the array, which was offset from the array line by 5 mm. Local light penetration was determined and compared across the selected wavelengths. RESULTS Unfixed cadaver brains provide good anatomical localization and reliable measurements of light scatter and penetration in the CNS tissues. Transcranial application of 808 nm wavelength light penetrated the scalp, skull, meninges, and brain to a depth of approximately 40 mm with an effective attenuation coefficient for the system of 2.22 cm(-1) . No differences were observed in the results between the PW and CW laser light. The intraparenchymal studies demonstrated less absorption and scattering for the 808 nm wavelength light compared to the 660 or 940 nm wavelengths. CONCLUSIONS Transcranial light measurements of unfixed human cadaver brains allowed for determinations of light penetration variables. While unfixed human cadaver studies do not reflect all the conditions seen in the living condition, comparisons of light scatter and penetration and estimates of fluence levels can be used to establish further clinical dosing. The 808 nm wavelength light demonstrated superior CNS tissue penetration.
Collapse
|
18
|
Henderson TA, Morries LD. Near-infrared photonic energy penetration: can infrared phototherapy effectively reach the human brain? Neuropsychiatr Dis Treat 2015; 11:2191-208. [PMID: 26346298 PMCID: PMC4552256 DOI: 10.2147/ndt.s78182] [Citation(s) in RCA: 227] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury (TBI) is a growing health concern effecting civilians and military personnel. Research has yielded a better understanding of the pathophysiology of TBI, but effective treatments have not been forthcoming. Near-infrared light (NIR) has shown promise in animal models of both TBI and stroke. Yet, it remains unclear if sufficient photonic energy can be delivered to the human brain to yield a beneficial effect. This paper reviews the pathophysiology of TBI and elaborates the physiological effects of NIR in the context of this pathophysiology. Pertinent aspects of the physical properties of NIR, particularly in regards to its interactions with tissue, provide the background for understanding this critical issue of light penetration through tissue. Our recent tissue studies demonstrate no penetration of low level NIR energy through 2 mm of skin or 3 cm of skull and brain. However, at 10-15 W, 0.45%-2.90% of 810 nm light penetrated 3 cm of tissue. A 15 W 810 nm device (continuous or non-pulsed) NIR delivered 2.9% of the surface power density. Pulsing at 10 Hz reduced the dose of light delivered to the surface by 50%, but 2.4% of the surface energy reached the depth of 3 cm. Approximately 1.22% of the energy of 980 nm light at 10-15 W penetrated to 3 cm. These data are reviewed in the context of the literature on low-power NIR penetration, wherein less than half of 1% of the surface energy could reach a depth of 1 cm. NIR in the power range of 10-15 W at 810 and 980 nm can provide fluence within the range shown to be biologically beneficial at 3 cm depth. A companion paper reviews the clinical data on the treatment of patients with chronic TBI in the context of the current literature.
Collapse
Affiliation(s)
- Theodore A Henderson
- The Synaptic Space, Centennial, CO, USA ; Neuro-Laser Foundation, Lakewood, CO, USA
| | | |
Collapse
|
19
|
Giacci MK, Wheeler L, Lovett S, Dishington E, Majda B, Bartlett CA, Thornton E, Harford-Wright E, Leonard A, Vink R, Harvey AR, Provis J, Dunlop SA, Hart NS, Hodgetts S, Natoli R, Van Den Heuvel C, Fitzgerald M. Differential effects of 670 and 830 nm red near infrared irradiation therapy: a comparative study of optic nerve injury, retinal degeneration, traumatic brain and spinal cord injury. PLoS One 2014; 9:e104565. [PMID: 25105800 PMCID: PMC4126771 DOI: 10.1371/journal.pone.0104565] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 07/10/2014] [Indexed: 01/23/2023] Open
Abstract
Red/near-infrared irradiation therapy (R/NIR-IT) delivered by laser or light-emitting diode (LED) has improved functional outcomes in a range of CNS injuries. However, translation of R/NIR-IT to the clinic for treatment of neurotrauma has been hampered by lack of comparative information regarding the degree of penetration of the delivered irradiation to the injury site and the optimal treatment parameters for different CNS injuries. We compared the treatment efficacy of R/NIR-IT at 670 nm and 830 nm, provided by narrow-band LED arrays adjusted to produce equal irradiance, in four in vivo rat models of CNS injury: partial optic nerve transection, light-induced retinal degeneration, traumatic brain injury (TBI) and spinal cord injury (SCI). The number of photons of 670 nm or 830 nm light reaching the SCI injury site was 6.6% and 11.3% of emitted light respectively. Treatment of rats with 670 nm R/NIR-IT following partial optic nerve transection significantly increased the number of visual responses at 7 days after injury (P ≤ 0.05); 830 nm R/NIR-IT was partially effective. 670 nm R/NIR-IT also significantly reduced reactive species and both 670 nm and 830 nm R/NIR-IT reduced hydroxynonenal immunoreactivity (P ≤ 0.05) in this model. Pre-treatment of light-induced retinal degeneration with 670 nm R/NIR-IT significantly reduced the number of Tunel+ cells and 8-hydroxyguanosine immunoreactivity (P ≤ 0.05); outcomes in 830 nm R/NIR-IT treated animals were not significantly different to controls. Treatment of fluid-percussion TBI with 670 nm or 830 nm R/NIR-IT did not result in improvements in motor or sensory function or lesion size at 7 days (P>0.05). Similarly, treatment of contusive SCI with 670 nm or 830 nm R/NIR-IT did not result in significant improvements in functional recovery or reduced cyst size at 28 days (P>0.05). Outcomes from this comparative study indicate that it will be necessary to optimise delivery devices, wavelength, intensity and duration of R/NIR-IT individually for different CNS injury types.
Collapse
Affiliation(s)
- Marcus K. Giacci
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Australia
- School of Animal Biology, The University of Western Australia, Crawley, Australia
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Australia
| | - Lachlan Wheeler
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Australia
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Australia
| | - Sarah Lovett
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Australia
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Australia
| | - Emma Dishington
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Australia
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Australia
| | - Bernadette Majda
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Australia
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Australia
| | - Carole A. Bartlett
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Australia
- School of Animal Biology, The University of Western Australia, Crawley, Australia
| | - Emma Thornton
- School of Medical Sciences, The University of Adelaide, Adelaide, Australia
| | | | - Anna Leonard
- School of Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Robert Vink
- School of Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Alan R. Harvey
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Australia
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Australia
| | - Jan Provis
- ANU Medical School and John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Sarah A. Dunlop
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Australia
- School of Animal Biology, The University of Western Australia, Crawley, Australia
| | - Nathan S. Hart
- School of Animal Biology, The University of Western Australia, Crawley, Australia
- Neuroecology Group, The Oceans Institute, The University of Western Australia, Crawley, Australia
| | - Stuart Hodgetts
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Australia
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Crawley, Australia
| | - Riccardo Natoli
- ANU Medical School and John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | | | - Melinda Fitzgerald
- Experimental and Regenerative Neurosciences, The University of Western Australia, Crawley, Australia
- School of Animal Biology, The University of Western Australia, Crawley, Australia
- * E-mail:
| |
Collapse
|
20
|
Moro C, Massri NE, Torres N, Ratel D, De Jaeger X, Chabrol C, Perraut F, Bourgerette A, Berger M, Purushothuman S, Johnstone D, Stone J, Mitrofanis J, Benabid AL. Photobiomodulation inside the brain: a novel method of applying near-infrared light intracranially and its impact on dopaminergic cell survival in MPTP-treated mice. J Neurosurg 2014; 120:670-83. [DOI: 10.3171/2013.9.jns13423] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
Previous experimental studies have documented the neuroprotection of damaged or diseased cells after applying, from outside the brain, near-infrared light (NIr) to the brain by using external light-emitting diodes (LEDs) or laser devices. In the present study, the authors describe an effective and reliable surgical method of applying to the brain, from inside the brain, NIr to the brain. They developed a novel internal surgical device that delivers the NIr to brain regions very close to target damaged or diseased cells. They suggest that this device will be useful in applying NIr within the large human brain, particularly if the target cells have a very deep location.
Methods
An optical fiber linked to an LED or laser device was surgically implanted into the lateral ventricle of BALB/c mice or Sprague-Dawley rats. The authors explored the feasibility of the internal device, measured the NIr signal through living tissue, looked for evidence of toxicity at doses higher than those required for neuroprotection, and confirmed the neuroprotective effect of NIr on dopaminergic cells in the substantia nigra pars compacta (SNc) in an acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of Parkinson disease in mice.
Results
The device was stable in freely moving animals, and the NIr filled the cranial cavity. Measurements showed that the NIr intensity declined as distance from the source increased across the brain (65% per mm) but was detectable up to 10 mm away. At neuroprotective (0.16 mW) and much higher (67 mW) intensities, the NIr caused no observable behavioral deficits, nor was there evidence of tissue necrosis at the fiber tip, where radiation was most intense. Finally, the intracranially delivered NIr protected SNc cells against MPTP insult; there were consistently more dopaminergic cells in MPTP-treated mice irradiated with NIr than in those that were not irradiated.
Conclusions
In summary, the authors showed that NIr can be applied intracranially, does not have toxic side effects, and is neuroprotective.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jonathan Stone
- 3Physiology, University of Sydney, New South Wales, Australia
| | | | | |
Collapse
|
21
|
Tuby H, Hertzberg E, Maltz L, Oron U. Long-term safety of low-level laser therapy at different power densities and single or multiple applications to the bone marrow in mice. Photomed Laser Surg 2013; 31:269-73. [PMID: 23675984 DOI: 10.1089/pho.2012.3395] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE The purpose of this study was to determine the long-term safety effect of low-level laser therapy (LLLT) to the bone marrow (BM) in mice. BACKGROUND DATA LLLT has been shown to have a photobiostimulatory effect on various cellular processes and on stem cells. It was recently shown that applying LLLT to BM in rats post-myocardial infarction caused a marked reduction of scar tissue formation in the heart. METHODS Eighty-three mice were divided into five groups: control sham-treated and laser-treated at measured density of either 4, 10, 18, or 40 mW/cm(2) at the BM level. The laser was applied to the exposed flat medial part of the tibia 8 mm from the knee joint for 100 sec. Mice were monitored for 8 months and then killed, and histopathology was performed on various organs. RESULTS No histological differences were observed in the liver, kidneys, brain or BM of the laser-treated mice as compared with the sham-treated, control mice. Moreover, no neoplasmic response in the tissues was observed in the laser-treated groups as compared with the control, sham-treated mice. There were no significant histopathological differences among the same organs under different laser treatment regimes in response to the BM-derived mesenchymal stem cell proliferation following LLLT to the BM. CONCLUSIONS LLLT applied multiple times either at the optimal dose (which induces photobiostimulation of stem cells in the BM), or at a higher dose (such as five times the optimal dose), does not cause histopathological changes or neoplasmic response in various organs in mice, as examined over a period of 8 months.
Collapse
Affiliation(s)
- Hana Tuby
- Department of Zoology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University Tel-Aviv, Israel
| | | | | | | |
Collapse
|
22
|
Oron A, Oron U, Streeter J, De Taboada L, Alexandrovich A, Trembovler V, Shohami E. Near infrared transcranial laser therapy applied at various modes to mice following traumatic brain injury significantly reduces long-term neurological deficits. J Neurotrauma 2012; 29:401-7. [PMID: 22040267 DOI: 10.1089/neu.2011.2062] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Near-infrared transcranial laser therapy (TLT) has been found to modulate various biological processes including traumatic brain injury (TBI). Following TBI in mice, in this study we assessed the possibility of various near-infrared TLT modes (pulsed versus continuous) in producing a beneficial effect on the long-term neurobehavioral outcome and brain lesions of these mice. TBI was induced by a weight-drop device, and neurobehavioral function was assessed from 1 h to 56 days post-trauma using the Neurological Severity Score (NSS). The extent of recovery is expressed as the difference in NSS (dNSS), the difference between the initial score and that at any other later time point. An 808-nm Ga-Al-As diode laser was employed transcranially 4, 6, or 8 h post-trauma to illuminate the entire cortex of the brain. Mice were divided into several groups of 6-8 mice: one control group that received a sham treatment and experimental groups that received either TLT continuous wave (CW) or pulsed wave (PW) mode transcranially. MRI was taken prior to sacrifice at 56 days post-injury. From 5-28 days post-TBI, the NSS of the laser-treated mice were significantly lower (p<0.05) than those of the non-laser-treated control mice. The percentage of surviving mice that demonstrated full recovery at 56 days post-CHI (NSS=0, as in intact mice) was the highest (63%) in the group that had received TLT in the PW mode at 100 Hz. In addition, magnetic resonance imaging (MRI) analysis demonstrated significantly smaller infarct lesion volumes in laser-treated mice compared to controls. Our data suggest that non-invasive TLT of mice post-TBI provides a significant long-term functional neurological benefit, and that the pulsed laser mode at 100 Hz is the preferred mode for such treatment.
Collapse
Affiliation(s)
- Amir Oron
- Department of Zoology, Tel Aviv University, Faculty of Life Sciences, Tel Aviv 69978, Israel.
| | | | | | | | | | | | | |
Collapse
|
23
|
The Translation Procedure of Low-Level Laser Therapy in Acute Ischemic Stroke: A Nonpharmaceutics Noninvasive Method. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
24
|
Rhee CK, He P, Jung JY, Ahn JC, Chung PS, Suh MW. Effect of low-level laser therapy on cochlear hair cell recovery after gentamicin-induced ototoxicity. Lasers Med Sci 2011; 27:987-92. [PMID: 22138884 DOI: 10.1007/s10103-011-1028-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 10/28/2011] [Indexed: 10/14/2022]
Abstract
Cochlear hair cells are the sensory receptors of the auditory system. It is well established that antibiotic drugs such as gentamicin can damage hair cells and cause hearing loss. Rescuing hair cells after ototoxic injury is an important issue in hearing recovery. Although many studies have indicated a positive effect of low-level laser therapy (LLLT) on neural cell survival, there has been no study on the effects of LLLT on cochlear hair cells. Therefore, the aim of this study was to elucidate the effects of LLLT on hair cell survival following gentamicin exposure in organotypic cultures of the cochlea of rats. The cochlea cultures were then divided into a control group (n = 8), a laser-only group (n = 8), a gentamicin-only group (n = 8) and a gentamicin plus laser group (n = 7). The control cultures were allowed to grow continuously for 11 days. The laser-only cultures were irradiated with a laser with a wavelength of 810 nm at 8 mW/cm(2) for 60 min per day (0.48 J/cm(2)) for 6 days. The gentamicin groups were exposed to 1 mM gentamicin for 48 h and allowed to recover (gentamicin-only group) or allowed to recover with daily irradiation (gentamicin plus laser group). The hair cells in all groups were stained with FM1-43 and counted every 3 days. The number of hair cells was significantly larger in the gentamicin plus laser group than in the gentamicin-only group. The number of hair cells was larger in the laser-only group than in the control group, but the difference did not reach statistical significance. These results suggest that LLLT may promote hair cell survival following gentamicin damage in the cochlea. This is the first study in the literature that has demonstrated the beneficial effect of LLLT on the recovery of cochlear hair cells.
Collapse
Affiliation(s)
- Chung-Ku Rhee
- Department of Otolaryngology-Head & Neck Surgery, Dankook University College of Medicine, Cheonan, Korea
| | | | | | | | | | | |
Collapse
|
25
|
Chung H, Dai T, Sharma SK, Huang YY, Carroll JD, Hamblin MR. The nuts and bolts of low-level laser (light) therapy. J Periodontal Implant Sci 2011; 40:105-10. [PMID: 20607054 PMCID: PMC2895515 DOI: 10.5051/jpis.2010.40.3.105] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Accepted: 04/20/2010] [Indexed: 01/11/2023] Open
Abstract
Soon after the discovery of lasers in the 1960s it was realized that laser therapy had the potential to improve wound healing and reduce pain, inflammation and swelling. In recent years the field sometimes known as photobiomodulation has broadened to include light-emitting diodes and other light sources, and the range of wavelengths used now includes many in the red and near infrared. The term "low level laser therapy" or LLLT has become widely recognized and implies the existence of the biphasic dose response or the Arndt-Schulz curve. This review will cover the mechanisms of action of LLLT at a cellular and at a tissular level and will summarize the various light sources and principles of dosimetry that are employed in clinical practice. The range of diseases, injuries, and conditions that can be benefited by LLLT will be summarized with an emphasis on those that have reported randomized controlled clinical trials. Serious life-threatening diseases such as stroke, heart attack, spinal cord injury, and traumatic brain injury may soon be amenable to LLLT therapy.
Collapse
Affiliation(s)
- Hoon Chung
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
26
|
Chung H, Dai T, Sharma SK, Huang YY, Carroll JD, Hamblin MR. The nuts and bolts of low-level laser (light) therapy. Ann Biomed Eng 2011; 40:516-33. [PMID: 22045511 DOI: 10.1007/s10439-011-0454-7] [Citation(s) in RCA: 832] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 10/20/2011] [Indexed: 01/03/2023]
Abstract
Soon after the discovery of lasers in the 1960s it was realized that laser therapy had the potential to improve wound healing and reduce pain, inflammation and swelling. In recent years the field sometimes known as photobiomodulation has broadened to include light-emitting diodes and other light sources, and the range of wavelengths used now includes many in the red and near infrared. The term "low level laser therapy" or LLLT has become widely recognized and implies the existence of the biphasic dose response or the Arndt-Schulz curve. This review will cover the mechanisms of action of LLLT at a cellular and at a tissular level and will summarize the various light sources and principles of dosimetry that are employed in clinical practice. The range of diseases, injuries, and conditions that can be benefited by LLLT will be summarized with an emphasis on those that have reported randomized controlled clinical trials. Serious life-threatening diseases such as stroke, heart attack, spinal cord injury, and traumatic brain injury may soon be amenable to LLLT therapy.
Collapse
Affiliation(s)
- Hoon Chung
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|