1
|
Henderson TA. Can infrared light really be doing what we claim it is doing? Infrared light penetration principles, practices, and limitations. Front Neurol 2024; 15:1398894. [PMID: 39263274 PMCID: PMC11388112 DOI: 10.3389/fneur.2024.1398894] [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: 03/11/2024] [Accepted: 06/10/2024] [Indexed: 09/13/2024] Open
Abstract
Near infrared (NIR) light has been shown to provide beneficial treatment of traumatic brain injury (TBI) and other neurological problems. This concept has spawned a plethora of commercial entities and practitioners utilizing panels of light emitting diodes (LEDs) and promising to treat patients with TBI and other disorders, who are desperate for some treatment for their untreatable conditions. Unfortunately, an LED intended to deliver photonic energy to the human brain does not necessarily do what an LED pointed at a mouse brain does. There is a problem of scale. Extensive prior research has shown that infrared light from a 0.5-watt LED will not penetrate the scalp and skull of a human. Both the properties of NIR light and the manner in which it interacts with tissue are examined. Based on these principles, the shortcomings of current approaches to treating neurological disorders with NIR light are explored. Claims of clinical benefit from low-level LED-based devices are explored and the proof of concept challenged. To date, that proof is thin with marginal benefits which are largely transient. Extensive research has shown fluence at the level of the target tissue which falls within the range of 0.9 J/cm2 to 15 J/cm2 is most effective in activating the biological processes at the cellular level which underlie direct photobiomodulation. If low-level infrared light from LED devices is not penetrating the scalp and skull, then these devices certainly are not delivering that level of fluence to the neurons of the subjacent brain. Alternative mechanisms, such as remote photobiomodulation, which may underlie the small and transient benefits for TBI symptoms reported for low-power LED-based NIR studies are presented. Actionable recommendations for the field are offered.
Collapse
Affiliation(s)
- Theodore A Henderson
- Neuro-Luminance, Inc., Denver, CO, United States
- Neuro-Laser Foundation, Denver, CO, United States
- Dr. Theodore Henderson, Inc., Denver, CO, United States
- The Synaptic Space, Inc., Denver, CO, United States
- The International Society of Applied Neuroimaging (ISAN), Toronto, ON, Canada
| |
Collapse
|
2
|
Ranjbar K, Komaki A, Fayazi B, Zarrinkalam E. Coenzyme Q10 and exercise training reinstate middle cerebral artery occlusion-induced behavioral deficits and hippocampal long-term potentiation suppression in aging rats. Psychopharmacology (Berl) 2024; 241:1577-1594. [PMID: 38627309 DOI: 10.1007/s00213-024-06583-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 03/29/2024] [Indexed: 06/15/2024]
Abstract
RATIONAL Patients experience post-stroke cognitive impairment during aging. To date, no specific treatment solution has been reported for this disorder. OBJECTIVE The purpose of this study was to evaluate the effects of exercise training and coenzyme Q10 supplementation on middle cerebral artery occlusion (MCAO) induced behavioral impairment, long-term potentiation inhibition and cerebral infarction size in aging rats. METHODS Fifty aging male rats underwent MCAO surgery and were randomly distributed in to the following groups: 1-Sham, 2- control, 3- Coenzyme Q10, 4- Exercise training and 5- Exercise training with Q10 supplementation (Ex + Q10). Aerobic training groups were allowed to run on a treadmill for 12 weeks. Q10 (50 mg/kg) was administered intragastrically by gavage. Morris water maze, shuttle box and elevated plus maze tests were used to evaluate cognitive function. The population spike (PS) amplitude and slope of excitatory postsynaptic potentials (EPSP) in the dentate gyrus area were recorded as a result of perforant pathway electrical stimulation. RESULTS Our study showed that Q10 and aerobic training alone ameliorate spatial memory in the acquisition phase, but have no effect on spatial memory in the retention phase. Q10 and exercise training synergistically promoted spatial memory in the retention phase. Q10 and exercise training separately and simultaneously mitigated cerebral ischemia-induced passive avoidance memory impairment in acquisition and retention phases. The EPSP did not differ between the groups, but exercise training and Q10 ameliorate the PS amplitude in hippocampal responses to perforant path stimulation. Exercising and Q10 simultaneously reduced the cerebral infarction volume. CONCLUSION Collectively, the findings of the present study imply that 12 weeks of aerobic training and Q10 supplementation alone can simultaneously reverse cerebral ischemia induced neurobehavioral deficits via amelioration of synaptic plasticity and a reduction in cerebral infarction volume in senescent rats.
Collapse
Affiliation(s)
- Kamal Ranjbar
- Department of Physical Education and Sport Science, Bandar Abbas Branch, Islamic Azad University, Bandar Abbas, Iran.
| | - Alireza Komaki
- Neurophysiology Research Center, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Bayan Fayazi
- Department of Physical Education and Sport Science, Razi University, Kermanshah, Iran
| | - Ebrahim Zarrinkalam
- Department of Physical Education and Sport Science, Hamedan Branch, Islamic Azad University, Hamedan, Iran.
| |
Collapse
|
3
|
Nairuz T, Sangwoo-Cho, Lee JH. Photobiomodulation Therapy on Brain: Pioneering an Innovative Approach to Revolutionize Cognitive Dynamics. Cells 2024; 13:966. [PMID: 38891098 PMCID: PMC11171912 DOI: 10.3390/cells13110966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Photobiomodulation (PBM) therapy on the brain employs red to near-infrared (NIR) light to treat various neurological and psychological disorders. The mechanism involves the activation of cytochrome c oxidase in the mitochondrial respiratory chain, thereby enhancing ATP synthesis. Additionally, light absorption by ion channels triggers the release of calcium ions, instigating the activation of transcription factors and subsequent gene expression. This cascade of events not only augments neuronal metabolic capacity but also orchestrates anti-oxidant, anti-inflammatory, and anti-apoptotic responses, fostering neurogenesis and synaptogenesis. It shows promise for treating conditions like dementia, stroke, brain trauma, Parkinson's disease, and depression, even enhancing cognitive functions in healthy individuals and eliciting growing interest within the medical community. However, delivering sufficient light to the brain through transcranial approaches poses a significant challenge due to its limited penetration into tissue, prompting an exploration of alternative delivery methods such as intracranial and intranasal approaches. This comprehensive review aims to explore the mechanisms through which PBM exerts its effects on the brain and provide a summary of notable preclinical investigations and clinical trials conducted on various brain disorders, highlighting PBM's potential as a therapeutic modality capable of effectively impeding disease progression within the organism-a task often elusive with conventional pharmacological interventions.
Collapse
Affiliation(s)
| | | | - Jong-Ha Lee
- Department of Biomedical Engineering, Keimyung University, Daegu 42601, Republic of Korea; (T.N.); (S.-C.)
| |
Collapse
|
4
|
Farazi N, Salehi-Pourmehr H, Farajdokht F, Mahmoudi J, Sadigh-Eteghad S. Photobiomodulation combination therapy as a new insight in neurological disorders: a comprehensive systematic review. BMC Neurol 2024; 24:101. [PMID: 38504162 PMCID: PMC10949673 DOI: 10.1186/s12883-024-03593-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 03/04/2024] [Indexed: 03/21/2024] Open
Abstract
Preclinical and clinical studies have indicated that combining photobiomodulation (PBM) therapy with other therapeutic approaches may influence the treatment process in a variety of disorders. The purpose of this systematic review was to determine whether PBM-combined therapy provides additional benefits over monotherapies in neurologic and neuropsychiatric disorders. In addition, the review describes the most commonly used methods and PBM parameters in these conjunctional approaches.To accomplish this, a systematic search was conducted in Google Scholar, PubMed, and Scopus databases through January 2024. 95 potentially eligible articles on PBM-combined treatment strategies for neurological and neuropsychological disorders were identified, including 29 preclinical studies and 66 clinical trials.According to the findings, seven major categories of studies were identified based on disease type: neuropsychiatric diseases, neurodegenerative diseases, ischemia, nerve injury, pain, paresis, and neuropathy. These studies looked at the effects of laser therapy in combination with other therapies like pharmacotherapies, physical therapies, exercises, stem cells, and experimental materials on neurological disorders in both animal models and humans. The findings suggested that most combination therapies could produce synergistic effects, leading to better outcomes for treating neurologic and psychiatric disorders and relieving symptoms.These findings indicate that the combination of PBM may be a useful adjunct to conventional and experimental treatments for a variety of neurological and psychological disorders.
Collapse
Affiliation(s)
- Narmin Farazi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, 5166614756, Iran
| | - Hanieh Salehi-Pourmehr
- Research Center for Evidence-Based Medicine, Iranian EBM Centre: A Joanna Briggs Institute (JBI) Center of Excellence, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, 5166614756, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, 5166614756, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, 5166614756, Iran.
| |
Collapse
|
5
|
Lin H, Li D, Zhu J, Liu S, Li J, Yu T, Tuchin VV, Semyachkina-Glushkovskaya O, Zhu D. Transcranial photobiomodulation for brain diseases: review of animal and human studies including mechanisms and emerging trends. NEUROPHOTONICS 2024; 11:010601. [PMID: 38317779 PMCID: PMC10840571 DOI: 10.1117/1.nph.11.1.010601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 12/07/2023] [Accepted: 01/05/2024] [Indexed: 02/07/2024]
Abstract
The brain diseases account for 30% of all known diseases. Pharmacological treatment is hampered by the blood-brain barrier, limiting drug delivery to the central nervous system (CNS). Transcranial photobiomodulation (tPBM) is a promising technology for treating brain diseases, due to its effectiveness, non-invasiveness, and affordability. tPBM has been widely used in pre-clinical experiments and clinical trials for treating brain diseases, such as stroke and Alzheimer's disease. This review provides a comprehensive overview of tPBM. We summarize emerging trends and new discoveries in tPBM based on over one hundred references published in the past 20 years. We discuss the advantages and disadvantages of tPBM and highlight successful experimental and clinical protocols for treating various brain diseases. A better understanding of tPBM mechanisms, the development of guidelines for clinical practice, and the study of dose-dependent and personal effects hold great promise for progress in treating brain diseases.
Collapse
Affiliation(s)
- Hao Lin
- Huazhong University of Science and Technology, Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics – Advanced Biomedical Imaging Facility, Wuhan, China
| | - Dongyu Li
- Huazhong University of Science and Technology, Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics – Advanced Biomedical Imaging Facility, Wuhan, China
- Huazhong University of Science and Technology, School of Optical Electronic Information, Wuhan, China
| | - Jingtan Zhu
- Huazhong University of Science and Technology, Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics – Advanced Biomedical Imaging Facility, Wuhan, China
| | - Shaojun Liu
- Huazhong University of Science and Technology, Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics – Advanced Biomedical Imaging Facility, Wuhan, China
| | - Jingting Li
- Huazhong University of Science and Technology, School of Engineering Sciences, Wuhan, China
| | - Tingting Yu
- Huazhong University of Science and Technology, Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics – Advanced Biomedical Imaging Facility, Wuhan, China
| | - Valery V. Tuchin
- Saratov State University, Science Medical Center, Saratov, Russia
- Research Center of Biotechnology of the Russian Academy of Sciences, Bach Institute of Biochemistry, Moscow, Russia
- Tomsk State University, Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk, Russia
| | - Oxana Semyachkina-Glushkovskaya
- Saratov State University, Science Medical Center, Saratov, Russia
- Humboldt University, Department of Physics, Berlin, Germany
| | - Dan Zhu
- Huazhong University of Science and Technology, Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics – Advanced Biomedical Imaging Facility, Wuhan, China
| |
Collapse
|
6
|
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: 4] [Impact Index Per Article: 4.0] [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
|
7
|
Hoh Kam J, Mitrofanis J. Glucose Improves the Efficacy of Photobiomodulation in Changing ATP and ROS Levels in Mouse Fibroblast Cell Cultures. Cells 2023; 12:2533. [PMID: 37947612 PMCID: PMC10648764 DOI: 10.3390/cells12212533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/16/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
In this study, we tested the idea that photobiomodulation-the application of red to near infrared light (~λ = 600-1300 nm) to body tissues-is more effective in influencing cell metabolism when glucose is readily available. To this end, we used a mouse fibroblast (L-929) cell culture model and had two sets of conditions: non-stressed (10% FBS (foetal bovine serum)) and stressed (1% FBS), both either with or without glucose. We treated (or not) cells with photobiomodulation using an 810 nm laser at 15 mW/cm2 (~7.2 J/cm2). Our results showed that photobiomodulation was neither cytotoxic nor effective in enhancing measures of cell viability and proliferation, together with protein levels in any of the cell cultures. Photobiomodulation was, however, effective in increasing adenosine triphosphate (ATP) and decreasing reactive oxygen species (ROS) levels and this was-most importantly-only in conditions where glucose was present; corresponding cultures that did not contain glucose did not show these changes. In summary, we found that the benefits of photobiomodulation, in particular in changing ATP and ROS levels, were induced only when there was glucose available. Our findings lay a template for further explorations into the mechanisms of photobiomodulation, together with having considerable experimental and clinical implications.
Collapse
Affiliation(s)
- Jaimie Hoh Kam
- Fonds de Dotation Clinatec, Grenoble Alpes University, 38000 Grenoble, France;
| | - John Mitrofanis
- Fonds de Dotation Clinatec, Grenoble Alpes University, 38000 Grenoble, France;
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| |
Collapse
|
8
|
Tang L, Jiang H, Sun M, Liu M. Pulsed transcranial photobiomodulation generates distinct beneficial neurocognitive effects compared with continuous wave transcranial light. Lasers Med Sci 2023; 38:203. [PMID: 37668791 DOI: 10.1007/s10103-023-03865-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/24/2023] [Indexed: 09/06/2023]
Abstract
Previous research has demonstrated the beneficial effect brought by transcranial photobiomodulation (tPBM). The present study is a further investigation of pulsed transcranial light delivery, from the perspective of wavelength, operation mode, and pulse frequency. A total of 56 healthy young adults (28 males and 28 females) were included in this randomized, sham-controlled experimental study. The wavelength of tPBM was 660 nm and 850 nm, and under each wavelength, subjects were randomly assigned to one of the following four treatments: (1) sham control; (2) continuous-wave (CW) tPBM; (3) pulsed-wave (PW) tPBM (40 Hz); and (4) PW tPBM (100 Hz). The tPBM duration was 8 min and the mean power density was fixed at 250 mW/cm2. Karolinska Sleepiness Scale (KSS) questionnaire, psychomotor vigilance task (PVT), and delayed match-to-sample (DMS) task were completed by subjects before and after the intervention to test whether PW tPBM produced distinct beneficial effects with measures of sleepiness, attention, and memory. 32-channel electroencephalography (EEG) signals were obtained from subjects before, during and after receiving tPBM or sham intervention. Paired sample T test showed that the KSS score, the number of correct responses of PVT, and DMS rate correct score (RCS) of PW tPBM groups improved significantly after intervention (p < 0.05). With regard to EEG analysis, paired one-way repeated ANOVA test showed that during the intervention of PW tPBM, the average power within the Gamma band was higher than the baseline (p < 0.05). Our study presented that PW tPBM could generate better beneficial cognitive effects and change brain electrical activity under certain circumstances.
Collapse
Affiliation(s)
- Luyao Tang
- Department of Light Sources and Illuminating Engineering, Fudan University, Shanghai, China
| | - Hui Jiang
- Institute of Future Lighting, Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Miao Sun
- Department of Light Sources and Illuminating Engineering, Fudan University, Shanghai, China
| | - Muqing Liu
- Department of Light Sources and Illuminating Engineering, Fudan University, Shanghai, China.
- Zhongshan Fudan Joint Innovation Center, Zhongshan, China.
| |
Collapse
|
9
|
Priyadarshi A, Keshri GK, Gupta A. Effect of combination of photobiomodulation 904 nm superpulsed laser therapy and Hippophae rhamnoides L. on third-degree burn wound healing. J Cosmet Dermatol 2023; 22:2492-2501. [PMID: 37272267 DOI: 10.1111/jocd.15806] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/28/2023] [Accepted: 04/23/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND Burn is a traumatic injury and aesthetic scarless repair poses a great challenge in area of cosmetic dermatology. Focus on multimode therapeutic strategies to promote healing of burns by regulating various stages of healing is warranted. Photobiomodulation therapy (PBMT), a non-invasive modality grabs the attention to repair impaired wounds. Seabuckthorn extract (SBTL-ALE) is known to possess antioxidant, anti-inflammation, and tissue-repair abilities. Current study aims to assess the effect of combination treatment of PBM 904 nm superpulsed laser and SBTL-ALE (2.5%) on repair of third-degree burn in rats. METHODS Rats were randomized into five groups: uninjured, control, SBTL-ALE, 904 nm PBMT, and combination. A transdermal burn wound was induced on the dorsal side of rats of all groups except the uninjured group and respective treatment was applied for 7 days postwounding. RESULTS Dual treatment increased wound area contraction compared to control and either treatment alone. Immunohistochemical analyses exhibited increased angiogenesis, dermal hydration, collagen synthesis, and maintained redox homeostasis as evidenced by enhanced expression (p < 0.05) of CD31, aquaporin3, collagen type 3, Nrf2, and HO1 in combination group compared with control. Conversely, pro-inflammatory and oxidative stress markers exhibited reduced (p < 0.05) TNF-α, IL-6, IL-1β, NOS-2, ROS levels, and increased catalase activity in combined treatment. Furthermore, energy metabolizing enzymes viz. citrate synthase, CCO, and ATP contents were substantially (p < 0.05) increased, and LDH activity was reduced in the combination group. CONCLUSIONS Dual treatment (PBMT + SBTL-ALE) prominently accelerates third-degree burn wound healing in rats, which could pave the path for multimode therapeutic strategies for the management of burns and dermal cosmetic care.
Collapse
Affiliation(s)
- Ashok Priyadarshi
- Pharmacology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, India
| | - Gaurav K Keshri
- Pharmacology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, India
| | - Asheesh Gupta
- Pharmacology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), DRDO, Timarpur, India
| |
Collapse
|
10
|
Covatti C, Mizobuti DS, Rocha GLD, da Silva HNM, de Lourenço CC, Pertille A, Pereira ECL, Minatel E. Low-Level Photobiomodulation Therapy Modulates H 2O 2 Production, TRPC-6, and PGC-1α Levels in the Dystrophic Muscle. Photobiomodul Photomed Laser Surg 2023; 41:389-401. [PMID: 37527194 DOI: 10.1089/photob.2022.0115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023] Open
Abstract
Objective: This study evaluated photobiomodulation therapy (PBMT) effects on the factors involved in mitochondrial biogenesis, on the mitochondrial respiratory complexes, and on the transient receptor potential canonical channels (such as TRPC-1 and TRPC-6) in in vitro (mdx muscle cells) and in vivo studies (gastrocnemius muscle) from mdx mice, the dystrophin-deficient model of Duchenne muscular dystrophy (DMD). Background: There is no successful treatment for DMD, therefore demanding search for new therapies that can improve the muscle role, the quality of life, and the survival of dystrophic patients. Methods: The dystrophic primary muscle cells received PBMT at 0.6 J and 5 J, and the dystrophic gastrocnemius muscle received PBMT at 0.6 J. Results: The dystrophic muscle cells treated with PBMT (0.6 J and 5 J) showed no cytotoxicity and significantly lower levels in hydrogen peroxide (H2O2) production. We also demonstrated, for the first time, the capacity of PBMT, at a low dose (0.6 J), in reducing the TRPC-6 content and in raising the peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) content in the dystrophic gastrocnemius muscle. Conclusions: PBMT modulates H2O2 production, TRPC-6, and PGC-1α content in the dystrophic muscle. These results suggest that laser therapy could act as an auxiliary therapy in the treatment of dystrophic patients.
Collapse
Affiliation(s)
- Caroline Covatti
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Daniela Sayuri Mizobuti
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Guilherme Luiz da Rocha
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Heloina Nathalliê Mariano da Silva
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Caroline Caramano de Lourenço
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Adriana Pertille
- Graduate Program in Science of Human Movement, Universidade Metodista de Piracicaba (UNIMEP), Piracicaba, São Paulo, Brazil
| | - Elaine Cristina Leite Pereira
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Universidade de Brasília (UnB), Faculdade de Ceilândia, Brasília, Distrito Federal, Brazil
| | - Elaine Minatel
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
11
|
Danciu OC, Holdhoff M, Peterson RA, Fischer JH, Liu LC, Wang H, Venepalli NK, Chowdhery R, Nicholas MK, Russell MJ, Fan TM, Hergenrother PJ, Tarasow TM, Dudek AZ. Phase I study of procaspase-activating compound-1 (PAC-1) in the treatment of advanced malignancies. Br J Cancer 2023; 128:783-792. [PMID: 36470974 PMCID: PMC9977881 DOI: 10.1038/s41416-022-02089-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Procaspase-3 (PC-3) is overexpressed in multiple tumour types and procaspase-activating compound 1 (PAC-1) directly activates PC-3 and induces apoptosis in cancer cells. This report describes the first-in-human, phase I study of PAC-1 assessing maximum tolerated dose, safety, and pharmacokinetics. METHODS Modified-Fibonacci dose-escalation 3 + 3 design was used. PAC-1 was administered orally at 7 dose levels (DL) on days 1-21 of a 28-day cycle. Dose-limiting toxicity (DLT) was assessed during the first two cycles of therapy, and pharmacokinetics analysis was conducted on days 1 and 21 of the first cycle. Neurologic and neurocognitive function (NNCF) tests were performed throughout the study. RESULTS Forty-eight patients were enrolled with 33 completing ≥2 cycles of therapy and evaluable for DLT. DL 7 (750 mg/day) was established as the recommended phase 2 dose, with grade 1 and 2 neurological adverse events noted, while NNCF testing showed stable neurologic and cognitive evaluations. PAC-1's t1/2 was 28.5 h after multi-dosing, and systemic drug exposures achieved predicted therapeutic concentrations. PAC-1 clinical activity was observed in patients with neuroendocrine tumour (NET) with 2/5 patients achieving durable partial response. CONCLUSIONS PAC-1 dose at 750 mg/day was recommended for phase 2 studies. Activity of PAC-1 in treatment-refractory NET warrants further investigation. CLINICAL TRIAL REGISTRATION Clinical Trials.gov: NCT02355535.
Collapse
Affiliation(s)
- Oana C Danciu
- Division of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
- Clinical Trials Office, University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| | - Matthias Holdhoff
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | | | - James H Fischer
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Li C Liu
- Division of Epidemiology and Biostatistics, School of Public Health, University of Illinois at Chicago, Chicago, IL, USA
| | - Heng Wang
- Division of Epidemiology and Biostatistics, School of Public Health, University of Illinois at Chicago, Chicago, IL, USA
| | - Neeta K Venepalli
- Division of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Rozina Chowdhery
- Division of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - M Kelly Nicholas
- Division of Hematology/Oncology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Meredith J Russell
- Clinical Trials Office, University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Timothy M Fan
- Vanquish Oncology, Inc., Champaign, IL, USA
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana-Champaign, IL, USA
- Cancer Center at Illinois, Urbana-Champaign, IL, USA
- Institute for Genomic Biology, University of Illinois, Urbana-Champaign, IL, USA
| | - Paul J Hergenrother
- Vanquish Oncology, Inc., Champaign, IL, USA
- Cancer Center at Illinois, Urbana-Champaign, IL, USA
- Institute for Genomic Biology, University of Illinois, Urbana-Champaign, IL, USA
- Department of Chemistry, University of Illinois, Urbana-Champaign, IL, USA
| | | | - Arkadiusz Z Dudek
- HealthPartners Institute, Regions Cancer Care Center, St. Paul, MN, USA
- Vanquish Oncology, Inc., Champaign, IL, USA
| |
Collapse
|
12
|
Stevens AR, Hadis M, Milward M, Ahmed Z, Belli A, Palin W, Davies DJ. Photobiomodulation in Acute Traumatic Brain Injury: A Systematic Review and Meta-Analysis. J Neurotrauma 2023; 40:210-227. [PMID: 35698294 DOI: 10.1089/neu.2022.0140] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Photobiomodulation (PBM) is a therapeutic modality that has gained increasing interest in neuroscience applications, including acute traumatic brain injury (TBI). Its proposed mechanisms for therapeutic effect when delivered to the injured brain include antiapoptotic and anti-inflammatory effects. This systematic review summarizes the available evidence for the value of PBM in improving outcomes in acute TBI and presents a meta-analysis of the pre-clinical evidence for neurological severity score (NSS) and lesion size in animal models of TBI. A systematic review of the literature was performed, with searches and data extraction performed independently in duplicate by two authors. Eighteen published articles were identified for inclusion: seventeen pre-clinical studies of in vivo animal models and one clinical study in human patients. The available human study supports safety and feasibility of PBM in acute moderate TBI. For pre-clinical studies, meta-analysis for NSS and lesion size were found to favor intervention versus control. Subgroup analysis based on PBM parameter variables for these outcomes was performed. Favorable parameters were identified as: wavelengths in the region of 665 nm and 810 nm; time to first administration of PBM ≤4 h; total number of daily treatments ≤3. No differences were identified between pulsed and continuous wave modes or energy delivery. Mechanistic substudies within included in vivo studies are presented and were found to support hypotheses of antiapoptotic, anti-inflammatory, and pro-proliferative effects, and a modulation of cellular metabolism. This systematic review provides substantial meta-analysis evidence of the benefits of PBM on functional and histological outcomes of TBI in in vivo mammalian models. Study design and PBM parameters should be closely considered for future human clinical studies.
Collapse
Affiliation(s)
- Andrew Robert Stevens
- Department of Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, and University of Birmingham, Edgbaston, Birmingham, United Kingdom.,NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham, Birmingham, United Kingdom.,Phototherapy Research Group, School of Dentistry, University of Birmingham, Birmingham, United Kingdom
| | - Mohammed Hadis
- Phototherapy Research Group, School of Dentistry, University of Birmingham, Birmingham, United Kingdom
| | - Michael Milward
- Phototherapy Research Group, School of Dentistry, University of Birmingham, Birmingham, United Kingdom
| | - Zubair Ahmed
- Department of Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, and University of Birmingham, Edgbaston, Birmingham, United Kingdom.,NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham, Birmingham, United Kingdom.,Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Antonio Belli
- Department of Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, and University of Birmingham, Edgbaston, Birmingham, United Kingdom.,NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham, Birmingham, United Kingdom.,Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - William Palin
- Phototherapy Research Group, School of Dentistry, University of Birmingham, Birmingham, United Kingdom
| | - David James Davies
- Department of Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, and University of Birmingham, Edgbaston, Birmingham, United Kingdom.,NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham, Birmingham, United Kingdom.,Phototherapy Research Group, School of Dentistry, University of Birmingham, Birmingham, United Kingdom.,Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| |
Collapse
|
13
|
Cheung MC, Lee TL, Sze SL, Chan AS. Photobiomodulation improves frontal lobe cognitive functions and mental health of older adults with non-amnestic mild cognitive impairment: Case studies. Front Psychol 2023; 13:1095111. [PMID: 36704674 PMCID: PMC9871821 DOI: 10.3389/fpsyg.2022.1095111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/16/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction This study investigated the effects of transcranial photobiomodulation (tPBM) on improving the frontal lobe cognitive functions and mental health of older adults. Methods Three older adults with mild cognitive impairment (MCI) of the non-amnestic type received 18-session tPBM stimulation for 9 weeks and were assessed with neuropsychological tests of memory and executive functions and standardized questionnaires on depressive and anxiety symptoms, global cognitive functions, and daily functioning abilities before and after tPBM stimulation. Results At baseline, their intrusion and/or perseveration errors in a verbal memory test and a fluency test, as measures of the frontal lobe cognitive functions, were in the borderline to severely impaired range at baseline. After tPBM stimulation, the three older adults showed various levels of improvement in their frontal lobe cognitive functions. One older adult's intrusion and perseveration errors improved from the <1st-2nd percentile (moderately to severely impaired range) to the 41st-69th percentile (average range), another older adult's intrusion errors improved from the 11th percentile to the 83rd percentile, and the third older adult's intrusion errors improved from the 5th percentile to the 56th percentile. Moreover, improvements in their anxiety and/or depressive symptoms were also observed. One older adult's depressive and anxiety symptoms improved from the severe range at baseline to the mild range after the intervention. The other two older adults' depressive symptoms improved from the mild range at baseline to the normal range after the intervention. Discussion These findings provide preliminary support for the potential of tPBM to improve the frontal lobe cognitive functions and mental health of older adults with MCI. Given the small sample size of only three older adults and the absence of a placebo control group, larger randomized controlled studies are needed to confirm its potential.
Collapse
Affiliation(s)
- Mei-Chun Cheung
- Department of Social Work, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China,Research Center for Neuropsychological Well-Being, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Tsz-Lok Lee
- Research Center for Neuropsychological Well-Being, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China,Neuropsychology Laboratory, Department of Psychology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Sophia L. Sze
- Research Center for Neuropsychological Well-Being, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China,Neuropsychology Laboratory, Department of Psychology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Agnes S. Chan
- Research Center for Neuropsychological Well-Being, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China,Neuropsychology Laboratory, Department of Psychology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China,*Correspondence: Agnes S. Chan, ✉
| |
Collapse
|
14
|
Dos Santos Cardoso F, Mansur FCB, Araújo BHS, Gonzalez-Lima F, Gomes da Silva S. Photobiomodulation Improves the Inflammatory Response and Intracellular Signaling Proteins Linked to Vascular Function and Cell Survival in the Brain of Aged Rats. Mol Neurobiol 2021; 59:420-428. [PMID: 34708330 DOI: 10.1007/s12035-021-02606-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 10/15/2021] [Indexed: 11/29/2022]
Abstract
Photobiomodulation is a non-pharmacological tool widely used to reduce inflammation in many tissues. However, little is known about its effects on the inflammatory response in the aged brain. We conducted the study to examine anti-inflammatory effects of photobiomodulation in aging brains. We used aged rats (20 months old) with control (handled, laser off) or transcranial laser (660 nm wavelength, 100 mW power) treatments for 10 consecutive days and evaluated the level of inflammatory cytokines and chemokines, and the expression and activation of intracellular signaling proteins in the cerebral cortex and the hippocampus. Inflammatory analysis showed that aged rats submitted to transcranial laser treatment had increased levels of IL-1alpha and decreased levels of IL-5 in the cerebral cortex. In the hippocampus, the laser treatment increased the levels of IL-1alpha and decreased levels of IL-5, IL-18, and fractalkine. Regarding the intracellular signaling proteins, a reduction in the ERK and p38 expression and an increase in the STAT3 and ERK activation were observed in the cerebral cortex of aged rats from the laser group. In addition, the laser treatment increased the hippocampal expression of p70S6K, STAT3, and p38 of aged rats. Taken together, our data indicate that transcranial photobiomodulation can improve the inflammatory response and the activation of intracellular signaling proteins linked to vascular function and cell survival in the aged brain.
Collapse
Affiliation(s)
- Fabrízio Dos Santos Cardoso
- Núcleo de Pesquisas Tecnológicas, Universidade de Mogi das Cruzes, Av. Cândido Xavier de Almeida e Souza, 200, Mogi das Cruzes, SP, 08780-911, Brazil. .,Department of Psychology and Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA. .,Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brasil.
| | | | - Bruno Henrique Silva Araújo
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa Em Energia E Materiais (CNPEM), Campinas, SP, Brazil
| | - F Gonzalez-Lima
- Department of Psychology and Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - Sérgio Gomes da Silva
- Núcleo de Pesquisas Tecnológicas, Universidade de Mogi das Cruzes, Av. Cândido Xavier de Almeida e Souza, 200, Mogi das Cruzes, SP, 08780-911, Brazil. .,Centro Universitário UNIFAMINAS (UNIFAMINAS), Muriaé, MG, Brazil. .,Hospital Do Câncer de Muriaé, Fundação Cristiano Varella (FCV), Muriaé, MG, Brazil.
| |
Collapse
|
15
|
Tao L, Liu Q, Zhang F, Fu Y, Zhu X, Weng X, Han H, Huang Y, Suo Y, Chen L, Gao X, Wei X. Microglia modulation with 1070-nm light attenuates Aβ burden and cognitive impairment in Alzheimer's disease mouse model. LIGHT, SCIENCE & APPLICATIONS 2021; 10:179. [PMID: 34493703 PMCID: PMC8423759 DOI: 10.1038/s41377-021-00617-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/29/2021] [Accepted: 08/17/2021] [Indexed: 05/05/2023]
Abstract
Photobiomodulation, by utilizing low-power light in the visible and near-infrared spectra to trigger biological responses in cells and tissues, has been considered as a possible therapeutic strategy for Alzheimer's disease (AD), while its specific mechanisms have remained elusive. Here, we demonstrate that cognitive and memory impairment in an AD mouse model can be ameliorated by 1070-nm light via reducing cerebral β-amyloid (Aβ) burden, the hallmark of AD. The glial cells, including microglia and astrocytes, play important roles in Aβ clearance. Our results show that 1070-nm light pulsed at 10 Hz triggers microglia rather than astrocyte responses in AD mice. The 1070-nm light-induced microglia responses with alteration in morphology and increased colocalization with Aβ are sufficient to reduce Aβ load in AD mice. Moreover, 1070-nm light pulsed at 10 Hz can reduce perivascular microglia and promote angiogenesis to further enhance Aβ clearance. Our study confirms the important roles of microglia and cerebral vessels in the use of 1070-nm light for the treatment of AD mice and provides a framework for developing a novel therapeutic approach for AD.
Collapse
Affiliation(s)
- Lechan Tao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qi Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Fuli Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yuting Fu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xi Zhu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xiaofu Weng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Hongbin Han
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China
- Department of Radiology, Peking University Third Hospital, Beijing, 100191, China
- Key Lab of Magnetic Resonance Imaging Device and Technique, Beijing, 100191, China
| | - Yong Huang
- Zhejiang Brainhealth Medical Technology Co., Ltd, Hangzhou, 314400, China
| | - Yuanzhen Suo
- Biomedical Pioneering Innovation Center, Peking University, Beijing, 100871, China
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
- Tianqiao and Chrissy Chen Institute for Clinical Translational Research, Huashan Hospital, Shanghai, 200040, China.
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xunbin Wei
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China.
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.
- Biomedical Engineering Department, Peking University, Beijing, 100081, China.
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
16
|
Superpulsed 904 nm laser photobiomodulation combined with coenzyme Q10 synergistically augment burn wound healing. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2021. [DOI: 10.1016/j.jpap.2021.100053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
17
|
Li W, Song Y, Liang X, Zhou Y, Xu M, Lu Q, Wang X, Li N. Mutual-reinforcing sonodynamic therapy against Rheumatoid Arthritis based on sparfloxacin sonosensitizer doped concave-cubic rhodium nanozyme. Biomaterials 2021; 276:121063. [PMID: 34391020 DOI: 10.1016/j.biomaterials.2021.121063] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 12/12/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease associated with synovitis and cartilage destruction. Ultrasound (US)-driven sonodynamic therapy (SDT) possess a good application prospect in RA therapy because of its non-invasiveness and strong tissue penetration capabilities, which can kill activated synovial inflammatory cells. Nevertheless, the tiny accumulation of sonosensitizers in the joints and the hypoxic synovial microenvironment severely limit the therapeutic effect of SDT. Hence, we developed a sonosensitizer spafloxacin (SPX) doped and human serum albumin (HSA) loaded concave-cubic rhodium (Rh) nanozyme (Rh/SPX-HSA) to realize mutual-reinforcing SDT during ultrasonic activation. On the one hand, SPX would cause mitochondrial dysfunction by inducing excessive reactive oxygen species (ROS) production, thus suppressing fibroblast-like synoviocyte (FLS) under US conditions. On the other hand, concave-cubic rhodium was utilized as a nanozyme with endogenous peroxidase (POD) and catalase (CAT)-like enzyme activities, which not only relieved the hypoxia of the joint to resist angiogenesis, but also enormously ascended the SDT efficacy by rising 1O2 levels. Interestingly, the activity of nanozymes was also improved by the ultrasonic cavitation effect, thereby realizing mutual-reinforcing SDT. Overall, our strategy provided Rh-based to achieve effective SDT under hypoxic microenvironment, which offered a promising prospect for highly efficient treatment of RA.
Collapse
Affiliation(s)
- Wen Li
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, PR China
| | - Yilin Song
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, PR China
| | - Xiaoyang Liang
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, PR China
| | - Yue Zhou
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, PR China
| | - Min Xu
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, PR China
| | - Qiang Lu
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, PR China
| | - Xinxing Wang
- Tianjin Institute of Environmental and Operational Medicine, 1 Dali Road, Heping District, 300050, Tianjin, PR China.
| | - Nan Li
- Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, PR China.
| |
Collapse
|
18
|
Yang M, Yang Z, Wang P, Sun Z. Current application and future directions of photobiomodulation in central nervous diseases. Neural Regen Res 2021; 16:1177-1185. [PMID: 33269767 PMCID: PMC8224127 DOI: 10.4103/1673-5374.300486] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/20/2020] [Accepted: 05/25/2020] [Indexed: 02/05/2023] Open
Abstract
Photobiomodulation using light in the red or near-infrared region is an innovative treatment strategy for a wide range of neurological and psychological conditions. Photobiomodulation can promote neurogenesis and elicit anti-apoptotic, anti-inflammatory and antioxidative responses. Its therapeutic effects have been demonstrated in studies on neurological diseases, peripheral nerve injuries, pain relief and wound healing. We conducted a comprehensive literature review of the application of photobiomodulation in patients with central nervous system diseases in February 2019. The NCBI PubMed database, EMBASE database, Cochrane Library and ScienceDirect database were searched. We reviewed 95 papers and analyzed. Photobiomodulation has wide applicability in the treatment of stroke, traumatic brain injury, Parkinson's disease, Alzheimer's disease, major depressive disorder, and other diseases. Our analysis provides preliminary evidence that PBM is an effective therapeutic tool for the treatment of central nervous system diseases. However, additional studies with adequate sample size are needed to optimize treatment parameters.
Collapse
Affiliation(s)
- Muyue Yang
- Shanghai Jiao Tong University, Shanghai, China
| | - Zhen Yang
- Core Facility of West China Hospital, Chengdu, Sichuan Province, China
| | - Pu Wang
- Department of Rehabilitation Medicine, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Zhihui Sun
- Department of Psychosomatic Medicine, The People’s Hospital of Suzhou New District, Suzhou, Jiangsu Province, China
| |
Collapse
|
19
|
Méndez M, Fidalgo C, Arias JL, Arias N. Methylene blue and photobiomodulation recover cognitive impairment in hepatic encephalopathy through different effects on cytochrome c-oxidase. Behav Brain Res 2021; 403:113164. [PMID: 33549685 DOI: 10.1016/j.bbr.2021.113164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 01/02/2021] [Accepted: 02/01/2021] [Indexed: 10/22/2022]
Abstract
Mitochondrial dysfunction plays a central role in hepatic encephalopathy (HE), due to changes in enzyme cytochrome c-oxidase (CCO), causing a decline in brain metabolism. We used an HE animal model and applied intracranial administration of methylene blue (MB) and transcranial photobiomodulation (PBM), both targeting CCO, to determine their differential effects on recovering cognition. Five groups of rats were used: sham-operated group + saline (SHAM + SAL, n = 6), hepatic encephalopathy + SAL (HE + SAL, n = 7), SHAM + methylene blue (SHAM + MB, n = 7), HE + MB (n = 7), HE + PBM (n = 7). PBM animals were exposed transcranially to 670 +/- 10 nm LED light at a dose of 9 J/cm2 once a day for 7 days, and the MB and SAL groups were injected with 2.2 μg/0.5 μL in the accumbens. Cognitive dysfunction was evaluated on a striatal stimulus-response task using the Morris water maze. Our results showed cognitive improvement in the HE group when treated with MB. This improvement was accompanied by a decrease in CCO activity in the prefrontal cortex, dorsal striatum, and dorsal hippocampus. When comparing MB and PBM, we found that, although both treatments effectively improved the HE-memory deficit, there was a differential effect on CCO. A general decrease in CCO activity was found in the prefrontal and entorhinal cortices, dorsal striatum, and hippocampus when PBM, compared to MB, was applied. Our results suggest that mitochondrial dysfunction and brain metabolic decline in HE might involve CCO alteration and can be improved by administering MB and PBM.
Collapse
Affiliation(s)
- Marta Méndez
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad de Oviedo, Plaza Feijoo s/n, Oviedo, 33003, Spain; INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
| | - Camino Fidalgo
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain; Departamento de Psicología y Sociología, IIS Aragón, Universidad de Zaragoza, Ciudad Escolar s/n, Teruel, 44003, Spain
| | - Jorge L Arias
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad de Oviedo, Plaza Feijoo s/n, Oviedo, 33003, Spain; INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
| | - Natalia Arias
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain; UK Dementia Research Institute, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK.
| |
Collapse
|
20
|
Bonvicini JFS, Basso FG, de Souza Costa CA, Soares CJ, Turrioni AP. Photobiomodulation effect of red LED (630 nm) on the free radical levels produced by pulp cells under stress conditions. Lasers Med Sci 2021; 37:607-617. [PMID: 33826014 DOI: 10.1007/s10103-021-03309-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 03/28/2021] [Indexed: 12/13/2022]
Abstract
The aim of this study was to assess the ability of red light emitting diodes (LED) to modulate oxidative stress in human dental pulp fibroblasts (HDPFs) when different irradiation parameters are employed. Cells from primary teeth were seeded (100,000 cells/well) in 24-well plates in culture medium (DMEM). At 24 h after incubation, the culture medium was replaced with DMEM containing 10 μg/mL lipopolysaccharide (LPS). Thereafter, the cells were irradiated (LED 630 nm, 0.04 W/cm2 and 0.08 W/cm2) at 0 J/cm2 (control group), 4 J/cm2, 15 J/cm2, and 30 J/cm2; and their viability (MTT assay), number (Trypan Blue), synthesis of nitric oxide (NO) (Griess reagent), and reactive oxygen species (ROS) (fluorescence probe, DCFH-DA) were assessed. The Kruskal-Wallis and Mann-Whitney statistical tests using Bonferroni correction were employed (significance level of 5%). Compared to that in control fibroblasts, increased viability was observed in HDPFs exposed to LPS and irradiated with 15 J/cm2 and 30 J/cm2 at 0.04 W/cm2 and 4 J/cm2 and 15 J/cm2 at 0.08 W/cm2 (p < 0.05). Exposure to 4 J/cm2 at 0.04 W/cm2 and 15 J/cm2 and 30 J/cm2 at 0.08 W/cm2 modulated the oxidative stress in cells relative to that observed in non-irradiated LPS-treated pulp cells (p < 0.05). It was concluded that the irradiation strategies of using red LED with radiant exposures of 15 J/cm2 and 30 J/cm2 at 0.04 W/cm2 and 15 J/cm2 at 0.08 W/cm2 were the best parameters to decrease NO and ROS concentration and to stimulate viability of HDPFs exposed to LPS challenge.
Collapse
Affiliation(s)
- Jéssica Fernanda Sena Bonvicini
- Department of Pediatric Dentistry, School of Dentistry, Federal University of Uberlândia, UFU, Av. Pará 1720, Umuarama, Uberlândia, MG, 38400902, Brazil
| | | | | | - Carlos José Soares
- Department of Operative Dentistry and Dental Materials, Federal University of Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Ana Paula Turrioni
- Department of Pediatric Dentistry, School of Dentistry, Federal University of Uberlândia, UFU, Av. Pará 1720, Umuarama, Uberlândia, MG, 38400902, Brazil.
| |
Collapse
|
21
|
Markoulli M, Chandramohan N, Papas EB. Photobiomodulation (low-level light therapy) and dry eye disease. Clin Exp Optom 2021; 104:561-566. [PMID: 33689636 DOI: 10.1080/08164622.2021.1878866] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Dry eye disease is one of the most common, chief-complaints presenting in clinical practice, with a prevalence of up to 50%. Evaporative dry eye, as a result of meibomian gland dysfunction, is thought to be the biggest component factor. Treatments for meibomian gland dysfunction aim to restore tear film homoeostasis and include warm compress therapy, eyelid hygiene, in-office meibomian gland expression and lipid-containing, artificial tears. A recent introduction to the in-office treatments available for meibomian gland dysfunction has been low-level light therapy, also known as photobiomodulation. The technique involves applying red, or near infra-red, radiation using low-power light sources and is suggested to promote tissue repair, decrease inflammation, and relieve pain. This work aims to review the available literature on the efficacy and safety of photobiomodulation in meibomian gland dysfunction and dry eye disease, as well as what is currently known about its mechanism of action.
Collapse
Affiliation(s)
- Maria Markoulli
- School of Optometry & Vision Science, University of New South Wales, Sydney, Australia
| | - Nivaasheni Chandramohan
- School of Optometry & Vision Science, University of New South Wales, Sydney, Australia.,Macular Degeneration Foundation Australia, Sydney, Australia
| | - Eric B Papas
- School of Optometry & Vision Science, University of New South Wales, Sydney, Australia
| |
Collapse
|
22
|
You J, Bragin A, Liu H, Li L. Preclinical studies of transcranial photobiomodulation in the neurological diseases. TRANSLATIONAL BIOPHOTONICS 2021. [DOI: 10.1002/tbio.202000024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Jing You
- Department of Biomedical Engineering University of North Texas Denton Texas USA
| | - Anatol Bragin
- Department of Neurology University of California Los Angeles Los Angeles California USA
- Brain Research Institute University of California Los Angeles Los Angeles California USA
| | - Hanli Liu
- Department of Bioengineering University of Texas at Arlington Arlington Texas USA
| | - Lin Li
- Department of Biomedical Engineering University of North Texas Denton Texas USA
- Department of Neurology University of California Los Angeles Los Angeles California USA
| |
Collapse
|
23
|
Yang J, Wang L, Wu MX. 830 nm photobiomodulation therapy promotes engraftment of human umbilical cord blood-derived hematopoietic stem cells. Sci Rep 2020; 10:19671. [PMID: 33184429 PMCID: PMC7661704 DOI: 10.1038/s41598-020-76760-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 11/02/2020] [Indexed: 11/26/2022] Open
Abstract
Human umbilical cord blood (hUCB)-derived hematopoietic stem cells (HSCs) are an important source for HSCs in allogeneic HSC transplantation, but a limited number and a low efficacy of engraftment greatly restrict their clinical use. Here, we report the ability of photobiomodulation therapy (PBMT) to significantly enhance the engraftment efficacy of hUCB HSCs and progenitor cells (HSPCs). hUCB CD34+ cells were illuminated at a fluence of 2 J/cm2 with a near-infrared light (830 nm) transmitted by an array of light-emitting diodes (LED) prior to infusion of NOD/SCID-IL2Rγ−/− mice. The pre-treatment resulted in a threefold higher of the mean percentage of human CD45+ cells in the periphery of the mice compared to sham-treated CD34+ cells. The enhanced engraftment may result from a PBMT-mediated increase of intracellular reactive oxygen species (ROS) levels and Src protein phosphorylation in CD34+ cells. The two events were causally related as suggested by the finding that elevation of ROS by hydrogen peroxide increased Src phosphorylation, while ROS reduction by N-acetyl cysteine partially reversed the phosphorylation. The investigation demonstrates that PBMT can promote engraftment of hUCB HPSCs, at least in part, via ROS-mediated Src signaling pathway. PBMT can be potentially a safe, convenient, and cost-effective modality to improve hematological reconstitution in patients.
Collapse
Affiliation(s)
- Jingke Yang
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Li Wang
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Mei X Wu
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
24
|
Photobiomodulation as a promising new tool in the management of psychological disorders: A systematic review. Neurosci Biobehav Rev 2020; 119:242-254. [PMID: 33069687 DOI: 10.1016/j.neubiorev.2020.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/21/2020] [Accepted: 10/09/2020] [Indexed: 12/18/2022]
Abstract
Photobiomodulation is a brain modulation technique that has become a promising treatment for multiple pathologies. This systematic review collects studies up to 2019 about the beneficial effects of photobiomodulation as a therapy for treating psychological disorders and a tool for modulating cognitive processes. This technique is mostly used for the treatment of depression and stress, as well as to study its effects on psychological variables in healthy subjects. Despite the lack of parameters used, photobiomodulation seems to achieve enough brain penetration to produce beneficial effects in healthy subjects and patients with multiple pathologies. The best parameters are the wavelengths of 810 nm for the treatment of depression and 1064 nm for cognitive enhancement, along with a scalp irradiance of 250 mW/cm2 and a scalp yield of 60 J/cm2. It weekly application on the bilateral prefrontal area and the default mode network seems to be ideal for the maintenance of the effects. Photobiomodulation could be used as an effective and safe therapy for the treatment of multiple psychological pathologies.
Collapse
|
25
|
Arias JL, Mendez M, Martínez JÁ, Arias N. Differential effects of photobiomodulation interval schedules on brain cytochrome c-oxidase and proto-oncogene expression. NEUROPHOTONICS 2020; 7:045011. [PMID: 33313338 PMCID: PMC7723391 DOI: 10.1117/1.nph.7.4.045011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/28/2020] [Indexed: 06/12/2023]
Abstract
Significance: Transcranial photobiomodulation (PBM) is a noninvasive neuromodulation technique capable of producing changes in the mitochondrial cytochrome c-oxidase (CCO) activity of neurons. Although the application of PBM in clinical practice and as a neurophysiological tool is increasing, less is known about how different treatment time intervals may result in different outcomes. Aim: We evaluated the effects of different PBM treatment intervals on brain metabolic activity through the CCO and proto-oncogene expression (c-Fos). Approach: We studied PBM effects on brain CCO and c-Fos expression in three groups of animals: Control (CN, n = 8 ), long interval PBM treatment (LI, n = 5 ), and short interval PBM treatment (SI, n = 5 ). Results: Increased CCO activity in the LI group, compared to the SI and CN groups, was found in the prefrontal cortices, dorsal and ventral striatum, and hippocampus. Regarding c-Fos expression, we found a significant increase in the SI group compared to LI and CN, whereas LI showed increased c-Fos expression compared to CN in the cingulate and infralimbic cortices. Conclusions: We show the effectiveness of different PBM interval schedules in increasing brain metabolic activity or proto-oncogene expression.
Collapse
Affiliation(s)
- Jorge L. Arias
- University of Oviedo, Neuroscience Laboratory, Department of Psychology, Oviedo, Spain
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
| | - Marta Mendez
- University of Oviedo, Neuroscience Laboratory, Department of Psychology, Oviedo, Spain
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
| | - Juan Ángel Martínez
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
- University of Oviedo, Escuela Politécnica de Gijón, Departamento Ingeniería Eléctrica, Electrónica, Computadores y Sistemas, Gijón, Spain
| | - Natalia Arias
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
- Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King´s College London, Department of Basic and Clinical Neuroscience, London, United Kingdom
| |
Collapse
|
26
|
Boudreau MW, Peh J, Hergenrother PJ. Procaspase-3 Overexpression in Cancer: A Paradoxical Observation with Therapeutic Potential. ACS Chem Biol 2019; 14:2335-2348. [PMID: 31260254 PMCID: PMC6858495 DOI: 10.1021/acschembio.9b00338] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many anticancer strategies rely on the promotion of apoptosis in cancer cells as a means to shrink tumors. Crucial for apoptotic function are executioner caspases, most notably caspase-3, that proteolyze a variety of proteins, inducing cell death. Paradoxically, overexpression of procaspase-3 (PC-3), the low-activity zymogen precursor to caspase-3, has been reported in a variety of cancer types. Until recently, this counterintuitive overexpression of a pro-apoptotic protein in cancer has been puzzling. Recent studies suggest subapoptotic caspase-3 activity may promote oncogenic transformation, a possible explanation for the enigmatic overexpression of PC-3. Herein, the overexpression of PC-3 in cancer and its mechanistic basis is reviewed; collectively, the data suggest the potential for exploitation of PC-3 overexpression with PC-3 activators as a targeted anticancer strategy.
Collapse
Affiliation(s)
- Matthew W. Boudreau
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| | - Jessie Peh
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| | - Paul J. Hergenrother
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| |
Collapse
|
27
|
Serrage H, Heiskanen V, Palin WM, Cooper PR, Milward MR, Hadis M, Hamblin MR. Under the spotlight: mechanisms of photobiomodulation concentrating on blue and green light. Photochem Photobiol Sci 2019; 18:1877-1909. [PMID: 31183484 PMCID: PMC6685747 DOI: 10.1039/c9pp00089e] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/30/2019] [Indexed: 12/31/2022]
Abstract
Photobiomodulation (PBM) describes the application of light at wavelengths ranging from 400-1100 nm to promote tissue healing, reduce inflammation and promote analgesia. Traditionally, red and near-infra red (NIR) light have been used therapeutically, however recent studies indicate that other wavelengths within the visible spectrum could prove beneficial including blue and green light. This review aims to evaluate the literature surrounding the potential therapeutic effects of PBM with particular emphasis on the effects of blue and green light. In particular focus is on the possible primary and secondary molecular mechanisms of PBM and also evaluation of the potential effective parameters for application both in vitro and in vivo. Studies have reported that PBM affects an array of molecular targets, including chromophores such as signalling molecules containing flavins and porphyrins as well as components of the electron transport chain. However, secondary mechanisms tend to converge on pathways induced by increases in reactive oxygen species (ROS) production. Systematic evaluation of the literature indicated 72% of publications reported beneficial effects of blue light and 75% reported therapeutic effects of green light. However, of the publications evaluating the effects of green light, reporting of treatment parameters was uneven with 41% failing to report irradiance (mW cm-2) and 44% failing to report radiant exposure (J cm-2). This review highlights the potential of PBM to exert broad effects on a range of different chromophores within the body, dependent upon the wavelength of light applied. Emphasis still remains on the need to report exposure and treatment parameters, as this will enable direct comparison between different studies and hence enable the determination of the full potential of PBM.
Collapse
Affiliation(s)
- Hannah Serrage
- College of Medical and Dental Sciences, University of Birmingham, UK.
| | | | | | | | | | | | | |
Collapse
|
28
|
Meynaghizadeh-Zargar R, Sadigh-Eteghad S, Mohaddes G, Salehpour F, Rasta SH. Effects of transcranial photobiomodulation and methylene blue on biochemical and behavioral profiles in mice stress model. Lasers Med Sci 2019; 35:573-584. [PMID: 31372913 DOI: 10.1007/s10103-019-02851-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 07/23/2019] [Indexed: 12/19/2022]
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.
Collapse
Affiliation(s)
- Reza Meynaghizadeh-Zargar
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, 51666, Iran.,Department of Medical Physics, Tabriz University of Medical Sciences, Tabriz, 51666, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, 51666, Iran
| | - Gisou Mohaddes
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, 51666, Iran
| | - Farzad Salehpour
- Neurosciences Research Center (NSRC), 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 Physics, Tabriz University of Medical Sciences, Tabriz, 51666, Iran. .,Department of Medical Bioengineering, Tabriz University of Medical Sciences, Tabriz, 51666, Iran. .,School of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, UK.
| |
Collapse
|
29
|
Facchin F, Canaider S, Tassinari R, Zannini C, Bianconi E, Taglioli V, Olivi E, Cavallini C, Tausel M, Ventura C. Physical energies to the rescue of damaged tissues. World J Stem Cells 2019; 11:297-321. [PMID: 31293714 PMCID: PMC6600852 DOI: 10.4252/wjsc.v11.i6.297] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/24/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023] Open
Abstract
Rhythmic oscillatory patterns sustain cellular dynamics, driving the concerted action of regulatory molecules, microtubules, and molecular motors. We describe cellular microtubules as oscillators capable of synchronization and swarming, generating mechanical and electric patterns that impact biomolecular recognition. We consider the biological relevance of seeing the inside of cells populated by a network of molecules that behave as bioelectronic circuits and chromophores. We discuss the novel perspectives disclosed by mechanobiology, bioelectromagnetism, and photobiomodulation, both in term of fundamental basic science and in light of the biomedical implication of using physical energies to govern (stem) cell fate. We focus on the feasibility of exploiting atomic force microscopy and hyperspectral imaging to detect signatures of nanomotions and electromagnetic radiation (light), respectively, generated by the stem cells across the specification of their multilineage repertoire. The chance is reported of using these signatures and the diffusive features of physical waves to direct specifically the differentiation program of stem cells in situ, where they already are resident in all the tissues of the human body. We discuss how this strategy may pave the way to a regenerative and precision medicine without the needs for (stem) cell or tissue transplantation. We describe a novel paradigm based upon boosting our inherent ability for self-healing.
Collapse
Affiliation(s)
- Federica Facchin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), School of Medicine, University of Bologna, Bologna 40100, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Silvia Canaider
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), School of Medicine, University of Bologna, Bologna 40100, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Riccardo Tassinari
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Chiara Zannini
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Eva Bianconi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Valentina Taglioli
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Elena Olivi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Claudia Cavallini
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | | | - Carlo Ventura
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), School of Medicine, University of Bologna, Bologna 40100, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| |
Collapse
|
30
|
Elblbesy MA. Comparative In Vitro Study: Examining 635 nm Laser and 265 nm Ultraviolet Interaction with Blood. PHOTOBIOMODULATION PHOTOMEDICINE AND LASER SURGERY 2019; 37:342-348. [PMID: 31188088 DOI: 10.1089/photob.2018.4611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Objective: This study represents a viable assessment of the effect of the low-level laser (LLL) of 635 nm and ultraviolet (UV) of 265 nm on biophysical properties of blood. Materials and methods: Blood samples were divided into two main groups: one for irradiation by LLL and the other for irradiation by UV. Each group was divided into three aliquots. First aliquot: whole blood was exposed to radiation. The second aliquot: erythrocytes were exposed to radiation and resuspended in autologous plasma. The third aliquot: plasma was exposed to radiation, and erythrocytes were resuspended in it. The following parameters were measured after irradiation by LLL and UV for all aliquots: whole blood viscosity, microscopic aggregation index, deformation index, and Zeta potential. Results: A decrease in whole blood viscosity due to irradiation by LLL was observed. To the contrary, an increase in whole blood viscosity due to irradiation by UV was detected. A significant reduction in erythrocytes' aggregation was observed as a result of LLL and UV radiation. Erythrocytes' deformability was strongly affected by UV radiation, while there was no significant effect from LLL. Another noticeable change observed was an increase in Zeta potential due to UV and a decrease in Zeta potential values, as a result of LLL irradiation. Conclusions: It can be concluded from this study that LLL and UV can be used to change some biological processes, as well as cellular properties.
Collapse
Affiliation(s)
- Mohamed A Elblbesy
- Department of Medical Biophysics, Medical Research Institute, Alexandria University, Alexandria Governorate, Egypt
| |
Collapse
|
31
|
Salehpour F, Farajdokht F, Mahmoudi J, Erfani M, Farhoudi M, Karimi P, Rasta SH, Sadigh-Eteghad S, Hamblin MR, Gjedde A. Photobiomodulation and Coenzyme Q 10 Treatments Attenuate Cognitive Impairment Associated With Model of Transient Global Brain Ischemia in Artificially Aged Mice. Front Cell Neurosci 2019; 13:74. [PMID: 30983970 PMCID: PMC6434313 DOI: 10.3389/fncel.2019.00074] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 02/14/2019] [Indexed: 01/11/2023] Open
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-1β 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.
Collapse
Affiliation(s)
- Farzad Salehpour
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Physics, Tabriz University of Medical Sciences, Tabriz, Iran
- ProNeuroLIGHT LLC, Phoenix, AZ, United States
| | - Fereshteh Farajdokht
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marjan Erfani
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Higher Educational Institute of Rab-Rashid, Tabriz, Iran
| | - Mehdi Farhoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouran Karimi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Hossein Rasta
- Department of Medical Physics, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Bioengineering, Tabriz University of Medical Sciences, Tabriz, Iran
- School of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
- Department of Dermatology, Harvard Medical School, Boston, MA, United States
- Harvard-MIT Health Sciences and Technology, Cambridge, MA, United States
| | - Albert Gjedde
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Departments of Clinical Research and Nuclear Medicine, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology & Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
32
|
Yue X, Mei Y, Zhang Y, Tong Z, Cui D, Yang J, Wang A, Wang R, Fei X, Ai L, Di Y, Luo H, Li H, Luo W, Lu Y, Li R, Duan C, Gao G, Yang H, Sun B, He R, Song W, Han H, Tong Z. New insight into Alzheimer's disease: Light reverses Aβ-obstructed interstitial fluid flow and ameliorates memory decline in APP/PS1 mice. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2019; 5:671-684. [PMID: 31720368 PMCID: PMC6838540 DOI: 10.1016/j.trci.2019.09.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Pharmacological therapies to treat Alzheimer's disease (AD) targeting "Aβ" have failed for over 100 years. Low levels of laser light can disassemble Aβ. In this study, we investigated the mechanisms that Aβ-blocked extracellular space (ECS) induces memory disorders in APP/PS1 transgenic mice and addressed whether red light (RL) at 630 nm rescues cognitive decline by reducing Aβ-disturbed flow of interstitial fluid (ISF). METHODS We compared the heating effects on the brains of rats illuminated with laser light at 630, 680, and 810 nm for 40 minutes, respectively. Then, a light-emitting diode with red light at 630 nm (LED-RL) was selected to illuminate AD mice. The changes in the structure of ECS in the cortex were examined by fluorescent double labeling. The volumes of ECS and flow speed of ISF were quantified by magnetic resonance imaging. Spatial memory behaviors in mice were evaluated by the Morris water maze. Then, the brains were sampled for biochemical analysis. RESULTS RL at 630 nm had the least heating effects than other wavelengths associated with ~49% penetration ratio into the brains. For the molecular mechanisms, Aβ could induce formaldehyde (FA) accumulation by inactivating FA dehydrogenase. Unexpectedly, in turn, FA accelerated Aβ deposition in the ECS. However, LED-RL treatment not only directly destroyed Aβ assembly in vitro and in vivo but also activated FA dehydrogenase to degrade FA and attenuated FA-facilitated Aβ aggregation. Subsequently, LED-RL markedly smashed Aβ deposition in the ECS, recovered the flow of ISF, and rescued cognitive functions in AD mice. DISCUSSION Aβ-obstructed ISF flow is the direct reason for the failure of the developed medicine delivery from superficial into the deep brain in the treatment of AD. The phototherapy of LED-RL improves memory by reducing Aβ-blocked ECS and suggests that it is a promising noninvasive approach to treat AD.
Collapse
Affiliation(s)
- Xiangpei Yue
- Laboratory of Alzheimer's Optoelectric Therapy, Alzheimer's Disease Center, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yufei Mei
- Laboratory of Alzheimer's Optoelectric Therapy, Alzheimer's Disease Center, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Yun Zhang
- Department of Psychiatry, Townsend Family Laboratories, The University of British Columbia, Vancouver, Canada
| | - Zheng Tong
- School of Engineering, Mechanical Engineering with Renewable Energy. Old College, The University of Edinburgh, Edinburgh, United Kingdom
- Nanjing University of Aeronautics and Astronautics, Institute of Aeronautics and Astronautics, Aircraft Design and Engineering, Nanjing, China
| | - Dehua Cui
- Department of Radiology, Peking University Third Hospital, Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China
| | - Jun Yang
- Department of Radiology, Peking University Third Hospital, Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China
| | - Aibo Wang
- Department of Radiology, Peking University Third Hospital, Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China
| | - Rui Wang
- Department of Radiology, Peking University Third Hospital, Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China
| | - Xuechao Fei
- Laboratory of Alzheimer's Optoelectric Therapy, Alzheimer's Disease Center, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Li Ai
- Laboratory of Alzheimer's Optoelectric Therapy, Alzheimer's Disease Center, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yalan Di
- Laboratory of Alzheimer's Optoelectric Therapy, Alzheimer's Disease Center, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Hongjun Luo
- Central Laboratory, Shantou University Medical College, Guangdong, China
| | - Hui Li
- Central Laboratory, Shantou University Medical College, Guangdong, China
| | - Wenhong Luo
- Central Laboratory, Shantou University Medical College, Guangdong, China
| | - Yu Lu
- Section of Environmental Biomedicine, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, China
| | - Rui Li
- Section of Environmental Biomedicine, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Central China Normal University, Wuhan, China
| | - Chunli Duan
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ge Gao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Hui Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Binggui Sun
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Rongqiao He
- Laboratory of Alzheimer's Optoelectric Therapy, Alzheimer's Disease Center, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
- State Key Laboratory of Brain & Cognitive Science, Institute of Biophysics, CAS Key Laboratory of Mental Health, University of Chinese Academy of Sciences (UCAS), Beijing, China
| | - Weihong Song
- Department of Psychiatry, Townsend Family Laboratories, The University of British Columbia, Vancouver, Canada
- Corresponding author. Tel: 604-822-8019; Fax: 604-822-7981.
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China
- Corresponding author. Tel: +86-010-82266972; Fax: +86-010-82265962.
| | - Zhiqian Tong
- Laboratory of Alzheimer's Optoelectric Therapy, Alzheimer's Disease Center, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
- Corresponding author. Tel: +86-010-83950362; Fax: +86-010-83950363.
| |
Collapse
|
33
|
Wu MX, Hamblin MR. Photobiomodulation and mitochondria for traumatic brain injury in mouse models. PHOTOBIOMODULATION IN THE BRAIN 2019:169-187. [DOI: 10.1016/b978-0-12-815305-5.00013-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
34
|
Near-infrared photobiomodulation combined with coenzyme Q 10 for depression in a mouse model of restraint stress: reduction in oxidative stress, neuroinflammation, and apoptosis. Brain Res Bull 2018; 144:213-222. [PMID: 30385146 DOI: 10.1016/j.brainresbull.2018.10.010] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/18/2018] [Accepted: 10/25/2018] [Indexed: 12/19/2022]
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.
Collapse
|
35
|
Yadav A, Verma S, Keshri GK, Gupta A. Combination of medicinal honey and 904 nm superpulsed laser-mediated photobiomodulation promotes healing and impedes inflammation, pain in full-thickness burn. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2018; 186:152-159. [DOI: 10.1016/j.jphotobiol.2018.07.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/05/2018] [Accepted: 07/07/2018] [Indexed: 01/09/2023]
|
36
|
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: 248] [Impact Index Per Article: 35.4] [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
|
37
|
Salehpour F, Farajdokht F, Erfani M, Sadigh-Eteghad S, Shotorbani SS, Hamblin MR, Karimi P, Rasta SH, Mahmoudi J. Transcranial near-infrared photobiomodulation attenuates memory impairment and hippocampal oxidative stress in sleep-deprived mice. Brain Res 2018; 1682:36-43. [PMID: 29307593 DOI: 10.1016/j.brainres.2017.12.040] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 12/23/2017] [Accepted: 12/29/2017] [Indexed: 12/15/2022]
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.
Collapse
Affiliation(s)
- Farzad Salehpour
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Physics, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marjan Erfani
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran; Higher Academic Education Institute of Rab-Rashid, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, United States; Department of Dermatology, Harvard Medical School, Boston, MA 02115, United States; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, United States
| | - Pouran Karimi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Hossein Rasta
- Department of Medical Physics, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Bioengineering, Tabriz University of Medical Sciences, Tabriz, Iran; School of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Javad Mahmoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
38
|
Zhang Q, Dong T, Li P, Wu MX. Noninvasive low-level laser therapy for thrombocytopenia. Sci Transl Med 2017; 8:349ra101. [PMID: 27464749 DOI: 10.1126/scitranslmed.aaf4964] [Citation(s) in RCA: 275] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/23/2016] [Indexed: 11/02/2022]
Abstract
Thrombocytopenia is a common hematologic disorder that is managed primarily by platelet transfusions. We report here that noninvasive whole-body illumination with a special near-infrared laser cures acute thrombocytopenia triggered by γ-irradiation within 2 weeks in mice, as opposed to a 5-week recovery time required in controls. The low-level laser (LLL) also greatly accelerated platelet regeneration in the presence of anti-CD41 antibody that binds and depletes platelets, and prevented a severe drop in platelet count caused by a common chemotherapeutic drug. Mechanistically, LLL stimulated mitochondrial biogenesis specifically in megakaryocytes owing to polyploidy of the cells. LLL also protected megakaryocytes from mitochondrial injury and apoptosis under stress. The multifaceted effects of LLL on mitochondria bolstered megakaryocyte maturation; facilitated elongation, branching, and formation of proplatelets; and doubled the number of platelets generated from individual megakaryocytes in mice. LLL-mediated platelet biogenesis depended on megakaryopoiesis and was inversely correlated with platelet counts, which kept platelet biogenesis in check and effectively averted thrombosis even after repeated uses, in sharp contrast to all current agents that stimulate the differentiation of megakaryocyte progenitors from hematopoietic stem cells independently of platelet counts. This safe, drug-free, donor-independent modality represents a paradigm shift in the prophylaxis and treatment of thrombocytopenia.
Collapse
Affiliation(s)
- Qi Zhang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA. Department of Dermatology, Harvard Medical School, Boston, MA 02114, USA
| | - Tingting Dong
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA. Department of Dermatology, Harvard Medical School, Boston, MA 02114, USA
| | - Peiyu Li
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA. Department of Dermatology, Harvard Medical School, Boston, MA 02114, USA
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA. Department of Dermatology, Harvard Medical School, Boston, MA 02114, USA. Affiliated faculty member of the Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02115, USA.
| |
Collapse
|
39
|
Hamblin MR. Photobiomodulation for traumatic brain injury and stroke. J Neurosci Res 2017; 96:731-743. [PMID: 29131369 DOI: 10.1002/jnr.24190] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 10/04/2017] [Accepted: 10/04/2017] [Indexed: 12/12/2022]
Abstract
There is a notable lack of therapeutic alternatives for what is fast becoming a global epidemic of traumatic brain injury (TBI). Photobiomodulation (PBM) employs red or near-infrared (NIR) light (600-1100nm) to stimulate healing, protect tissue from dying, increase mitochondrial function, improve blood flow, and tissue oxygenation. PBM can also act to reduce swelling, increase antioxidants, decrease inflammation, protect against apoptosis, and modulate microglial activation state. All these mechanisms of action strongly suggest that PBM delivered to the head should be beneficial in cases of both acute and chronic TBI. Most reports have used NIR light either from lasers or from light-emitting diodes (LEDs). Many studies in small animal models of acute TBI have found positive effects on neurological function, learning and memory, and reduced inflammation and cell death in the brain. There is evidence that PBM can help the brain repair itself by stimulating neurogenesis, upregulating BDNF synthesis, and encouraging synaptogenesis. In healthy human volunteers (including students and healthy elderly women), PBM has been shown to increase regional cerebral blood flow, tissue oxygenation, and improve memory, mood, and cognitive function. Clinical studies have been conducted in patients suffering from the chronic effects of TBI. There have been reports showing improvement in executive function, working memory, and sleep. Functional magnetic resonance imaging has shown modulation of activation in intrinsic brain networks likely to be damaged in TBI (default mode network and salience network).
Collapse
Affiliation(s)
- Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA.,Department of Dermatology, Harvard Medical School, Boston, MA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA
| |
Collapse
|
40
|
Abstract
Psoriasis is an autoimmune inflammatory skin disease. In the past several decades, phototherapy has been widely used to treat stable psoriatic lesions, including trunk, scalp, arms and legs, and partial nail psoriasis. A variety of light/lasers with different mechanisms of action have been developed for psoriasis including ultraviolet B (UVB), psoralen ultraviolet A (PUVA), pulsed dye laser (PDL), photodynamic therapy (PDT), intense pulsed light (IPL), light-emitting diodes (LED), and so on. Because light/laser each has specific therapeutic and adverse effects, it is important to adequately choose the sources and parameters in management of psoriasis with different pathogenic sites, severities, and duration of the disorder. This review aims at providing most updated clinic information to physicians about how to select light/laser sources and individual therapeutic regimens. To date, UV light is primarily for stable plaque psoriasis and PDL for topical psoriatic lesions with small area, both of which are safe and effective. On the other hand, PUVA has better curative effects than UVB for managing refractory psoriasis plaques, if its side effects can be better controlled. PDL provides optimal outcomes on nail psoriasis compared with other lasers. Although the trails of low-level light/laser therapy (LLLT) are still small, the near infrared (NIR) and visible red light with low energy show promise for treating psoriasis due to its strong penetration and encouraging photobiomodulation. IPL is rarely reported for psoriasis treatment, but PDT-IPL has been found to offer a moderate effect on nail psoriasis. In brief, various phototherapies have been used either in different combinations or as monotherapy. The modality has become a mainstay in the treatment of mild-to-moderate psoriasis without systemic adverse events in today's clinical practice.
Collapse
|
41
|
Thunshelle C, Hamblin MR. Transcranial Low-Level Laser (Light) Therapy for Brain Injury. Photomed Laser Surg 2017; 34:587-598. [PMID: 28001759 DOI: 10.1089/pho.2015.4051] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Low-level laser therapy (LLLT) or photobiomodulation (PBM) is a possible treatment for brain injury, including traumatic brain injury (TBI). METHODS We review the fundamental mechanisms at the cellular and molecular level and the effects on the brain are discussed. There are several contributing processes that have been proposed to lead to the beneficial effects of PBM in treating TBI such as stimulation of neurogenesis, a decrease in inflammation, and neuroprotection. Both animal and clinical trials for ischemic stroke are outlined. A number of articles have shown how transcranial LLLT (tLLLT) is effective at increasing memory, learning, and the overall neurological performance in rodent models with TBI. RESULTS Our laboratory has conducted three different studies on the effects of tLLLT on mice with TBI. The first studied pulsed against continuous laser irradiation, finding that 10 Hz pulsed was the best. The second compared four different wavelengths, discovering only 660 and 810 nm to have any effectiveness, whereas 732 and 980 nm did not. The third looked at varying regimens of daily laser treatments (1, 3, and 14 days) and found that 14 laser applications was excessive. We also review several studies of the effects of tLLLT on neuroprogenitor cells, brain-derived neurotrophic factor and synaptogenesis, immediate early response knockout mice, and tLLLT in combination therapy with metabolic inhibitors. CONCLUSIONS Finally, some clinical studies in TBI patients are covered.
Collapse
Affiliation(s)
- Connor Thunshelle
- 1 Harvard College , Cambridge, Massachusetts.,2 Wellman Center for Photomedicine , Massachusetts General Hospital, Boston, Massachusetts
| | - Michael R Hamblin
- 2 Wellman Center for Photomedicine , Massachusetts General Hospital, Boston, Massachusetts.,3 Department of Dermatology, Harvard Medical School , Boston, Massachusetts.,4 Harvard-MIT Division of Health Sciences and Technology , Cambridge, Massachusetts
| |
Collapse
|
42
|
Osier N, Dixon CE. Mini Review of Controlled Cortical Impact: A Well-Suited Device for Concussion Research. Brain Sci 2017; 7:E88. [PMID: 28726717 PMCID: PMC5532601 DOI: 10.3390/brainsci7070088] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 07/12/2017] [Accepted: 07/18/2017] [Indexed: 01/25/2023] Open
Abstract
Mild traumatic brain injury (mTBI) is increasingly recognized as a significant public health problem which warrants additional research. Part of the effort to understand mTBI and concussion includes modeling in animals. Controlled cortical impact (CCI) is a commonly employed and well-characterized model of experimental TBI that has been utilized for three decades. Today, several commercially available pneumatic- and electromagnetic-CCI devices exist as do a variety of standard and custom injury induction tips. One of CCI's strengths is that it can be scaled to a number of common laboratory animals. Similarly, the CCI model can be used to produce graded TBI ranging from mild to severe. At the mild end of the injury spectrum, CCI has been applied in many ways, including to study open and closed head mTBI, repeated injuries, and the long-term deficits associated with mTBI and concussion. The purpose of this mini-review is to introduce the CCI model, discuss ways the model can be applied to study mTBI and concussion, and compare CCI to alternative pre-clinical TBI models.
Collapse
Affiliation(s)
- Nicole Osier
- School of Nursing, Holistic Adult Health Division, University of Texas at Austin, Austin, TX 78701, USA.
- Dell Medical School, Department of Neurology, University of Texas at Austin, Austin, TX 78701, USA.
| | - C Edward Dixon
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA.
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15260, USA.
- VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA.
| |
Collapse
|
43
|
Salehpour F, Ahmadian N, Rasta SH, Farhoudi M, Karimi P, Sadigh-Eteghad S. Transcranial low-level laser therapy improves brain mitochondrial function and cognitive impairment in D-galactose-induced aging mice. Neurobiol Aging 2017; 58:140-150. [PMID: 28735143 DOI: 10.1016/j.neurobiolaging.2017.06.025] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 05/11/2017] [Accepted: 06/29/2017] [Indexed: 12/11/2022]
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.
Collapse
Affiliation(s)
- Farzad Salehpour
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Physics, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nahid Ahmadian
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Hossein Rasta
- Department of Medical Physics, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Bioengineering, Tabriz University of Medical Sciences, Tabriz, Iran; School of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouran Karimi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
44
|
Beirne K, Rozanowska M, Votruba M. Photostimulation of mitochondria as a treatment for retinal neurodegeneration. Mitochondrion 2017; 36:85-95. [PMID: 28499983 DOI: 10.1016/j.mito.2017.05.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 02/15/2017] [Accepted: 05/08/2017] [Indexed: 01/01/2023]
Abstract
Absorption of photon energy by neuronal mitochondria leads to numerous downstream neuroprotective effects. Red and near infrared (NIR) light are associated with significantly less safety concerns than light of shorter wavelengths and they are therefore, the optimal choice for irradiating the retina. Potent neuroprotective effects have been demonstrated in various models of retinal damage, by red/NIR light, with limited data from human studies showing its ability to improve visual function. Improved neuronal mitochondrial function, increased blood flow to neural tissue, upregulation of cell survival mediators and restoration of normal microglial function have all been proposed as potential underlying mechanisms of red/NIR light.
Collapse
Affiliation(s)
- Kathy Beirne
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK; Cardiff Institute for Tissue Engineering and Repair, Cardiff University, Cardiff, UK.
| | - Malgorzata Rozanowska
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK; Cardiff Institute for Tissue Engineering and Repair, Cardiff University, Cardiff, UK.
| | - Marcela Votruba
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK; Cardiff Institute for Tissue Engineering and Repair, Cardiff University, Cardiff, UK; Cardiff Eye Unit, University Hospital of Wales, Cardiff, UK.
| |
Collapse
|
45
|
Lee HI, Lee SW, Kim SY, Kim NG, Park KJ, Choi BT, Shin YI, Shin HK. Pretreatment with light-emitting diode therapy reduces ischemic brain injury in mice through endothelial nitric oxide synthase-dependent mechanisms. Biochem Biophys Res Commun 2017; 486:945-950. [PMID: 28347821 DOI: 10.1016/j.bbrc.2017.03.131] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 03/23/2017] [Indexed: 11/26/2022]
Abstract
Photostimulation with low-level light emitting diode therapy (LED-T) modulates neurological and psychological functions. The purpose of this study was to evaluate the effects of LED-T pretreatment on the mouse brain after ischemia/reperfusion and to investigate the underlying mechanisms. Ischemia/reperfusion brain injury was induced by middle cerebral artery occlusion. The mice received LED-T twice a day for 2 days prior to cerebral ischemia. After reperfusion, the LED-T group showed significantly smaller infarct and edema volumes, fewer behavioral deficits compared to injured mice that did not receive LED-T and significantly higher cerebral blood flow compared to the vehicle group. We observed lower levels of endothelial nitric oxide synthase (eNOS) phosphorylation in the injured mouse brains, but significantly higher eNOS phosphorylation in LED-T-pretreated mice. The enhanced phospho-eNOS was inhibited by LY294002, indicating that the effects of LED-T on the ischemic brain could be attributed to the upregulation of eNOS phosphorylation through the phosphoinositide 3-kinase (PI3K)/Akt pathway. Moreover, no reductions in infarct or edema volume were observed in LED-T-pretreated eNOS-deficient (eNOS-/-) mice. Collectively, we found that pretreatment with LED-T reduced the amount of ischemia-induced brain damage. Importantly, we revealed that these effects were mediated by the stimulation of eNOS phosphorylation via the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Hae In Lee
- Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Sae-Won Lee
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Graduate Training Program of Korean Medicine for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - So Young Kim
- Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Nam Gyun Kim
- Medical Research Center of Color Seven, Seoul 06719, Republic of Korea
| | - Kyoung-Jun Park
- Medical Research Center of Color Seven, Seoul 06719, Republic of Korea
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Graduate Training Program of Korean Medicine for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Yong-Il Shin
- Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Gyeongnam 50612, Republic of Korea.
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Graduate Training Program of Korean Medicine for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea.
| |
Collapse
|
46
|
Xuan W, Huang L, Hamblin MR. Repeated transcranial low-level laser therapy for traumatic brain injury in mice: biphasic dose response and long-term treatment outcome. JOURNAL OF BIOPHOTONICS 2016; 9:1263-1272. [PMID: 26990361 PMCID: PMC5025344 DOI: 10.1002/jbio.201500336] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/10/2016] [Accepted: 02/11/2016] [Indexed: 05/02/2023]
Abstract
We previously showed that near-infrared laser photobiomodulation (PBM) (810 nm, CW, 18 J/cm2 , 25 mW/cm2 ) delivered to the mouse daily for 3-days after a controlled cortical impact traumatic brain injury (TBI) gave a significant improvement in neurological/cognitive function. However the same parameters delivered 14X daily gave significantly less benefit. This biphasic dose response intrigued us, and we decided to follow the mice that received 3X or 14X laser treatments out to 56-days post-TBI. We found the 14X group showed worse neurological function than the no-treatment TBI group at 2-weeks, but started to improve steadily during the next 6-weeks, and by 56-days were significantly better than the no-treatment TBI mice, but still worse than the 3X mice. A marker of activated glial cells (GFAP) was significantly increased in the brain regions (compared to both untreated TBI and 3X groups) at 4-weeks in the 14X group, but the GFAP had fallen to low levels in both 3X and 14X groups by 8-weeks. We conclude that an excessive number of laser-treatments delivered to mice can temporarily inhibit the process of brain repair stimulated by tPBM, but then the inhibitory effect ceases, and brain repair can resume. The mechanism may be temporary induction of reactive gliosis.
Collapse
Affiliation(s)
- Weijun Xuan
- Dept of Otorhinolaryngology, Head and Neck Surgery, Ruikang Clinical Medical College, Guangxi University of Chinese Medicine, Nanning, China
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
| | - Liyi Huang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| |
Collapse
|
47
|
Low-level light treatment ameliorates immune thrombocytopenia. Sci Rep 2016; 6:38238. [PMID: 27901126 PMCID: PMC5128784 DOI: 10.1038/srep38238] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/07/2016] [Indexed: 11/08/2022] Open
Abstract
Immune thrombocytopenia (ITP) is an immune-mediated acquired bleeding disorder characterized by abnormally low platelet counts. We reported here the ability of low-level light treatment (LLLT) to alleviate ITP in mice. The treatment is based on noninvasive whole body illumination 30 min a day for a few consecutive days by near infrared light (830 nm) transmitted by an array of light-emitting diodes (LEDs). LLLT significantly lifted the nadir of platelet counts and restored tail bleeding time when applied to two passive ITP models induced by anti-CD41 antibody. The anti-platelet antibody hindered megakaryocyte differentiation from the progenitors, impaired proplatelet and platelet formation, and induced apoptosis of platelets. These adverse effects of anti-CD41 antibody were all mitigated by LLLT to varying degrees, owing to its ability to enhance mitochondrial biogenesis and activity in megakaryocytes and preserve mitochondrial functions in platelets in the presence of the antibody. The observations argue not only for contribution of mitochondrial stress to the pathology of ITP, but also clinical potentials of LLLT as a safe, simple, and cost-effective modality of ITP.
Collapse
|
48
|
Shining light on the head: Photobiomodulation for brain disorders. BBA CLINICAL 2016; 6:113-124. [PMID: 27752476 PMCID: PMC5066074 DOI: 10.1016/j.bbacli.2016.09.002] [Citation(s) in RCA: 314] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 09/27/2016] [Accepted: 09/29/2016] [Indexed: 12/21/2022]
Abstract
Photobiomodulation (PBM) describes the use of red or near-infrared light to stimulate, heal, regenerate, and protect tissue that has either been injured, is degenerating, or else is at risk of dying. One of the organ systems of the human body that is most necessary to life, and whose optimum functioning is most worried about by humankind in general, is the brain. The brain suffers from many different disorders that can be classified into three broad groupings: traumatic events (stroke, traumatic brain injury, and global ischemia), degenerative diseases (dementia, Alzheimer's and Parkinson's), and psychiatric disorders (depression, anxiety, post traumatic stress disorder). There is some evidence that all these seemingly diverse conditions can be beneficially affected by applying light to the head. There is even the possibility that PBM could be used for cognitive enhancement in normal healthy people. In this transcranial PBM (tPBM) application, near-infrared (NIR) light is often applied to the forehead because of the better penetration (no hair, longer wavelength). Some workers have used lasers, but recently the introduction of inexpensive light emitting diode (LED) arrays has allowed the development of light emitting helmets or "brain caps". This review will cover the mechanisms of action of photobiomodulation to the brain, and summarize some of the key pre-clinical studies and clinical trials that have been undertaken for diverse brain disorders.
Collapse
|
49
|
Dong T, Zhi L, Bhayana B, Wu MX. Cortisol-induced immune suppression by a blockade of lymphocyte egress in traumatic brain injury. J Neuroinflammation 2016; 13:197. [PMID: 27561600 PMCID: PMC5000452 DOI: 10.1186/s12974-016-0663-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/18/2016] [Indexed: 12/16/2022] Open
Abstract
Background Acute traumatic brain injury (TBI) represents one of major causes of mortality and disability in the USA. Neuroinflammation has been regarded both beneficial and detrimental, probably in a time-dependent fashion. Methods To address a role for neuroinflammation in brain injury, C57BL/6 mice were subjected to a closed head mild TBI (mTBI) by a standard controlled cortical impact, along with or without treatment of sphingosine 1-phosphate (S1P) or rolipram, after which the brain tissue of the impact site was evaluated for cell morphology via histology, inflammation by qRT-PCR and T cell staining, and cell death with Caspase-3 and TUNEL staining. Circulating lymphocytes were quantified by flow cytometry, and plasma hydrocortisone was analyzed by LC-MS/MS. To investigate the mechanism whereby cortisol lowered the number of peripheral T cells, T cell egress was tracked in lymph nodes by intravital confocal microscopy after hydrocortisone administration. Results We detected a decreased number of circulating lymphocytes, in particular, T cells soon after mTBI, which was inversely correlated with a transient and robust increase of plasma cortisol. The transient lymphocytopenia might be caused by cortisol in part via a blockade of lymphocyte egress as demonstrated by the ability of cortisol to inhibit T cell egress from the secondary lymphoid tissues. Moreover, exogenous hydrocortisone severely suppressed periphery lymphocytes in uninjured mice, whereas administering an egress-promoting agent S1P normalized circulating T cells in mTBI mice and increased T cells in the injured brain. Likewise, rolipram, a cAMP phosphodiesterase inhibitor, was also able to elevate cAMP levels in T cells in the presence of hydrocortisone in vitro and abrogate the action of cortisol in mTBI mice. The investigation demonstrated that the number of circulating T cells in the early phase of TBI was positively correlated with T cell infiltration and inflammatory responses as well as cell death at the cerebral cortex and hippocampus beneath the impact site. Conclusions Decreases in intracellular cAMP might be part of the mechanism behind cortisol-mediated blockade of T cell egress. The study argues strongly for a protective role of cortisol-induced immune suppression in the early stage of TBI.
Collapse
Affiliation(s)
- Tingting Dong
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA
| | - Liang Zhi
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA
| | - Brijesh Bhayana
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA.
| |
Collapse
|
50
|
Mao Z, Wu JH, Dong T, Wu MX. Additive enhancement of wound healing in diabetic mice by low level light and topical CoQ10. Sci Rep 2016; 6:20084. [PMID: 26830658 PMCID: PMC4735721 DOI: 10.1038/srep20084] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 12/15/2015] [Indexed: 12/26/2022] Open
Abstract
Diabetes, a highly prevalent disease that affects 9.3% of Americans, often leads to severe complications and slow wound healing. Preclinical studies have suggested that low level light therapy (LLLT) can accelerate wound healing in diabetic subjects, but significant improvements must be made to overcome the absence of persuasive evidence for its clinical use. We demonstrate here that LLLT can be combined with topical Coenzyme Q10 (CoQ10) to heal wounds in diabetic mice significantly faster than LLLT alone, CoQ10 alone, or controls. LLLT followed by topical CoQ10 enhanced wound healing by 68~103% in diabetic mice in the first week and more than 24% in the second week compared with untreated controls. All wounds were fully healed in two weeks following the dual treatment, in contrast to only 50% wounds or a fewer being fully healed for single or sham treatment. The accelerated healing was corroborated by at least 50% higher hydroxyproline levels, and tripling cell proliferation rates in LLLT and CoQ10 treated wounds over controls. The beneficial effects on wound healing were probably attributed to additive enhancement of ATP production by LLLT and CoQ10 treatment. The combination of LLLT and topical CoQ10 is safe and convenient, and merits further clinical study.
Collapse
Affiliation(s)
- Zhigang Mao
- ShangHai 9th People Hospital, Shanghai Jiao Tong University School of Medicine, ShangHai, China.,Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA 02114
| | - Jeffrey H Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA 02114
| | - Tingting Dong
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA 02114
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, Boston, MA 02114
| |
Collapse
|