1
|
Ban S, Yi H, Park J, Huang Y, Yu KJ, Yeo WH. Advances in Photonic Materials and Integrated Devices for Smart and Digital Healthcare: Bridging the Gap Between Materials and Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2416899. [PMID: 39905874 DOI: 10.1002/adma.202416899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/06/2024] [Indexed: 02/06/2025]
Abstract
Recent advances in developing photonic technologies using various materials offer enhanced biosensing, therapeutic intervention, and non-invasive imaging in healthcare. Here, this article summarizes significant technological advancements in materials, photonic devices, and bio-interfaced systems, which demonstrate successful applications for impacting human healthcare via improved therapies, advanced diagnostics, and on-skin health monitoring. The details of required materials, necessary properties, and device configurations are described for next-generation healthcare systems, followed by an explanation of the working principles of light-based therapeutics and diagnostics. Next, this paper shares the recent examples of integrated photonic systems focusing on translation and immediate applications for clinical studies. In addition, the limitations of existing materials and devices and future directions for smart photonic systems are discussed. Collectively, this review article summarizes the recent focus and trends of technological advancements in developing new nanomaterials, light delivery methods, system designs, mechanical structures, material functionalization, and integrated photonic systems to advance human healthcare and digital healthcare.
Collapse
Affiliation(s)
- Seunghyeb Ban
- George W. Woodruff School of Mechanical Engineering, Wearable Intelligent Systems and Healthcare Center at the Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Hoon Yi
- George W. Woodruff School of Mechanical Engineering, Wearable Intelligent Systems and Healthcare Center at the Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jaejin Park
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, South Korea
| | - Yunuo Huang
- School of Industrial Design, Wearable Intelligent Systems and Healthcare Center at the Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Ki Jun Yu
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, South Korea
- The Biotech Center, Pohang University of Science and Technology (POSTECH), Gyeongbuk, 37673, South Korea
- Department of Electrical and Electronic Engineering, YU-Korea Institute of Science and Technology (KIST) Institute, Yonsei University, Seoul, 03722, South Korea
| | - Woon-Hong Yeo
- George W. Woodruff School of Mechanical Engineering, Wearable Intelligent Systems and Healthcare Center at the Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
2
|
Barrett DW, Davis RE, Siegel-Ramsay JE, Bichlmeier A, Almeida JRC, Gonzalez-Lima F. Cognitive improvement and prefrontal network interactions in individuals with remitted bipolar disorder after transcranial infrared laser stimulation. Front Psychiatry 2025; 16:1547230. [PMID: 39950176 PMCID: PMC11822565 DOI: 10.3389/fpsyt.2025.1547230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 02/16/2025] Open
Abstract
Background Converging evidence suggests that bipolar disorder (BD) involves mitochondrial dysfunction and prefrontal cortex (PFC) hypometabolism associated with cognitive impairment, which persists in remitted BD individuals. Transcranial infrared laser stimulation (TILS) provides safe, non-invasive brain stimulation that enhances PFC metabolism via photobiomodulation of mitochondrial respiration and tissue oxygenation. We tested the hypothesis that the neurocognitive deficits found in BD may be ameliorated by TILS treatments. Methods This is the first study to explore neurocognitive effects of repeated TILS administration in BD. Using an open-label design, 29 individuals with remitted BD received six weekly TILS treatments. Working memory and attention were assessed with trail-making and 2-back tasks sensitive to TILS cognitive effects in individuals with BD. Changes in PFC network interactions were measured with functional near-infrared spectroscopy (fNIRS) because this method can measure TILS effects on oxygen metabolism in the PFC of individuals with BD. Results Participants reported no adverse effects from treatment, confirming the safety of this intervention in individuals with BD. Cognitive test results showed that in people with remitted BD, TILS was effective at improving cognition, i.e., enhanced speed and accuracy in tasks reflecting cognitive flexibility, working memory, and attentional control. Antipsychotic medication improved TILS cognitive effects. The fNIRS results showed a significant reduction in PFC network correlations of oxygenated hemoglobin changes driven by cognitive task performance. The right-hemisphere frontopolar cortex showed greater TILS effects than its left-hemisphere counterpart. Conclusions Repeated TILS is a safe intervention to improve cognition in people with remitted BD. Continued antipsychotic medication may have contributed to the cognitive improvement. To confirm TILS efficacy, a sham-controlled, double-blinded randomized trial is needed. Clinical trial registration https://clinicaltrials.gov/, identifier NCT05354895.
Collapse
Affiliation(s)
- Douglas W. Barrett
- Departments of Psychology, Psychiatry and Behavioral Sciences, and Institute for Neuroscience, The University of Texas at Austin, Austin, TX, United States
| | - Roger E. Davis
- Departments of Psychology, Psychiatry and Behavioral Sciences, and Institute for Neuroscience, The University of Texas at Austin, Austin, TX, United States
| | - Jennifer E. Siegel-Ramsay
- Bipolar Disorder Center, Department of Psychiatry and Behavioral Sciences, Dell Medical School, Austin, TX, United States
| | - Amy Bichlmeier
- Bipolar Disorder Center, Department of Psychiatry and Behavioral Sciences, Dell Medical School, Austin, TX, United States
| | - Jorge R. C. Almeida
- Bipolar Disorder Center, Department of Psychiatry and Behavioral Sciences, Dell Medical School, Austin, TX, United States
| | - F. Gonzalez-Lima
- Departments of Psychology, Psychiatry and Behavioral Sciences, and Institute for Neuroscience, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
3
|
Frankowski DW, Ferrucci L, Arany PR, Bowers D, Eells JT, Gonzalez-Lima F, Lohr NL, Quirk BJ, Whelan HT, Lakatta EG. Light buckets and laser beams: mechanisms and applications of photobiomodulation (PBM) therapy. GeroScience 2025:10.1007/s11357-025-01505-z. [PMID: 39826026 DOI: 10.1007/s11357-025-01505-z] [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: 12/17/2024] [Accepted: 12/31/2024] [Indexed: 01/20/2025] Open
Abstract
Photobiomodulation (PBM) therapy, a non-thermal light therapy using nonionizing light sources, has shown therapeutic potential across diverse biological processes, including aging and age-associated diseases. In 2023, scientists from the National Institute on Aging (NIA) Intramural and Extramural programs convened a workshop on the topic of PBM to discuss various proposed mechanisms of PBM action, including the stimulation of mitochondrial cytochrome C oxidase, modulation of cell membrane transporters and receptors, and the activation of transforming growth factor-β1. They also reviewed potential therapeutic applications of PBM across a range of conditions, including cardiovascular disease, retinal disease, Parkinson's disease, and cognitive impairment. Workshop participants largely agreed that PBM holds immense potential as a safe and effective therapeutic approach for a wide range of age-related diseases and cognitive decline. While further research is needed to fully elucidate its mechanisms and optimize treatment protocols, the findings presented at the NIA workshop provide strong evidence for the continued investigation and clinical translation of this promising, inexpensive, safe technology, to aging and age-associated diseases. Here, we review the research presented and discussion held at the meeting. In addition, the text has been updated, where applicable, with recent research findings that have been made since the meeting occurred.
Collapse
Affiliation(s)
| | | | | | | | - Janis T Eells
- University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | | | - Nicole L Lohr
- The University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | |
Collapse
|
4
|
Lee K, Chun M, Jung B, Kim Y, Yang C, Choi J, Cha J, Lee SH, Im CH. Machine-Learning-Based Prediction of Photobiomodulation Effects on Older Adults With Cognitive Decline Using Functional Near-Infrared Spectroscopy. IEEE Trans Neural Syst Rehabil Eng 2024; 32:3710-3718. [PMID: 39331542 DOI: 10.1109/tnsre.2024.3469284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2024]
Abstract
Transcranial photobiomodulation (tPBM) has been widely studied for its potential to enhance cognitive functions of the elderly. However, its efficacy varies, with some individuals exhibiting no significant response to the treatment. Considering these inconsistencies, we introduce a machine learning approach aimed at distinguishing between individuals that respond and do not respond to tPBM treatment based on functional near-infrared spectroscopy (fNIRS) acquired before the treatment. We measured nine cognitive scores and recorded fNIRS data from 62 older adults with cognitive decline (43 experimental and 19 control subjects). The experimental group underwent tPBM intervention over a span of 12 weeks. Based on the comparison of the global cognitive score (GCS), merging the nine cognitive scores into a single representation, acquired before and after tPBM treatment, we classified all participants as responders or non-responders to tPBM with a threshold for the GCS change. The fNIRS data were recorded during the resting state, recognition memory task (RMT), Stroop task, and verbal fluency task. A regularized support vector machine was utilized to classify the responders and non-responders to tPBM. The most promising performance of our machine learning model was observed when using the fNIRS data collected during the RMT, which yielded an accuracy of 0.8537, an F1-score of 0.8421, sensitivity of 0.7619, and specificity of 0.95. To the best of our knowledge, this is the first study to demonstrate the feasibility of predicting the tPBM efficacy. Our approach is expected to contribute to more efficient treatment planning by excluding ineffective treatment options.
Collapse
|
5
|
Oliveira de Andrade Filho V, Amarante MOC, Gonzalez-Lima F, Gomes da Silva S, Cardoso FDS. Systematic review of photobiomodulation for multiple sclerosis. Front Neurol 2024; 15:1465621. [PMID: 39329016 PMCID: PMC11424438 DOI: 10.3389/fneur.2024.1465621] [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: 07/16/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
Background Multiple sclerosis (MS) is an inflammatory chronic autoimmune and neurodegenerative disorder of the brain and spinal cord, resulting in loss of motor, sensorial, and cognitive function. Among the non-pharmacological interventions for several brain conditions, photobiomodulation (PBM) has gained attention in medical society for its neuroprotective effects. We systematically reviewed the effects of PBM on MS. Methods We conducted a systematic search on the bibliographic databases (PubMed and ScienceDirect) with the keywords based on MeSH terms: PBM, low-level laser therapy, multiple sclerosis, autoimmune encephalomyelitis, demyelination, and progressive multiple sclerosis. Data search was limited from 2012 to July 2024. We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The initial systematic search identified 126 articles. Of these, 68 articles were removed by duplicity and 50 by screening. Thus, 8 studies satisfied the inclusion criteria. Results The reviewed studies showed that PBM modulates brain markers linked to inflammation, oxidative stress, and apoptosis. Improvements in motor, sensorial, and cognitive functions in MS patients were also observed after PBM therapy. No study reported adverse effects of PBM. Conclusion These findings suggest the potential of PBM as a promising non-pharmacological intervention for the management of MS, although further research is needed to standardize PBM protocols and assess its long-term effects.
Collapse
Affiliation(s)
| | | | - Francisco Gonzalez-Lima
- Department of Psychology and Institute for Neuroscience, University of Texas at Austin, Austin, TX, United States
| | - Sérgio Gomes da Silva
- Centro Universitário FAMINAS, Muriaé, MG, Brazil
- Hospital do Câncer de Muriaé, Fundação Cristiano Varella (FCV), Muriaé, MG, Brazil
- Centro Universitário Redentor (UniREDENTOR/Afya), Itaperuna, RJ, Brazil
| | - Fabrízio Dos Santos Cardoso
- Hospital do Câncer de Muriaé, Fundação Cristiano Varella (FCV), Muriaé, MG, Brazil
- Centro Universitário Redentor (UniREDENTOR/Afya), Itaperuna, RJ, Brazil
| |
Collapse
|
6
|
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
|
7
|
Yokomizo S, Kopp T, Roessing M, Morita A, Lee S, Cho S, Ogawa E, Komai E, Inoue K, Fukushi M, Feil S, Kim HH, Bragin DE, Gerashchenko D, Huang PL, Kashiwagi S, Atochin DN. Near-Infrared II Photobiomodulation Preconditioning Ameliorates Stroke Injury via Phosphorylation of eNOS. Stroke 2024; 55:1641-1649. [PMID: 38572660 PMCID: PMC11126363 DOI: 10.1161/strokeaha.123.045358] [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: 06/18/2023] [Accepted: 02/28/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND The current management of patients with stroke with intravenous thrombolysis and endovascular thrombectomy is effective only when it is timely performed on an appropriately selected but minor fraction of patients. The development of novel adjunctive therapy is highly desired to reduce morbidity and mortality with stroke. Since endothelial dysfunction is implicated in the pathogenesis of stroke and is featured with suppressed endothelial nitric oxide synthase (eNOS) with concomitant nitric oxide deficiency, restoring endothelial nitric oxide represents a promising approach to treating stroke injury. METHODS This is a preclinical proof-of-concept study to determine the therapeutic effect of transcranial treatment with a low-power near-infrared laser in a mouse model of ischemic stroke. The laser treatment was performed before the middle cerebral artery occlusion with a filament. To determine the involvement of eNOS phosphorylation, unphosphorylatable eNOS S1176A knock-in mice were used. Each measurement was analyzed by a 2-way ANOVA to assess the effect of the treatment on cerebral blood flow with laser Doppler flowmetry, eNOS phosphorylation by immunoblot analysis, and stroke outcomes by infarct volumes and neurological deficits. RESULTS Pretreatment with a 1064-nm laser at an irradiance of 50 mW/cm2 improved cerebral blood flow, eNOS phosphorylation, and stroke outcomes. CONCLUSIONS Near-infrared II photobiomodulation could offer a noninvasive and low-risk adjunctive therapy for stroke injury. This new modality using a physical parameter merits further consideration to develop innovative therapies to prevent and treat a wide array of cardiovascular diseases.
Collapse
Affiliation(s)
- Shinya Yokomizo
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital, 114 16th Street, Charlestown, MA, 02129, USA
- Department of Radiological Science, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa, Tokyo 116-8551, Japan
| | - Timo Kopp
- Interfaculty Institute of Biochemistry (IFIB), University of Tübingen, Auf der Morgenstelle 34, Tübingen 72076, Germany
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital 149 13 Street, Charlestown, MA 02129, USA
| | - Malte Roessing
- Interfaculty Institute of Biochemistry (IFIB), University of Tübingen, Auf der Morgenstelle 34, Tübingen 72076, Germany
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital 149 13 Street, Charlestown, MA 02129, USA
| | - Atsuyo Morita
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital 149 13 Street, Charlestown, MA 02129, USA
| | - Seeun Lee
- Stroke and Neurovascular Regulation Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
- School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Suin Cho
- Stroke and Neurovascular Regulation Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
- School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Emiyu Ogawa
- School of Allied Health Science, Kitasato University, 1-15-1 Kitasato Minami-ku Sagamihara, Kanagawa, Japan
| | - Eri Komai
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital 149 13 Street, Charlestown, MA 02129, USA
| | - Kazumasa Inoue
- Department of Radiological Science, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa, Tokyo 116-8551, Japan
| | - Masahiro Fukushi
- Department of Radiological Science, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa, Tokyo 116-8551, Japan
| | - Susanne Feil
- Interfaculty Institute of Biochemistry (IFIB), University of Tübingen, Auf der Morgenstelle 34, Tübingen 72076, Germany
| | - Hyung-Hwan Kim
- Stroke and Neurovascular Regulation Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Denis E. Bragin
- Lovelace Biomedical Research Institute, 2425 Ridgecrest Dr. SE, Albuquerque, NM 87108, USA
- Department of Neurology, The University of New Mexico School of Medicine, MSC08 4720, 1 UNM, Albuquerque, NM 87131, USA
| | - Dmitry Gerashchenko
- Department of Psychiatry, Boston VA Medical Center West Roxbury, Veterans Affairs Boston Healthcare System and Harvard Medical School, 1400 VFW Pkwy, West Roxbury, MA 02132, USA
| | - Paul L. Huang
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital 149 13 Street, Charlestown, MA 02129, USA
| | - Satoshi Kashiwagi
- Department of Psychiatry, Boston VA Medical Center West Roxbury, Veterans Affairs Boston Healthcare System and Harvard Medical School, 1400 VFW Pkwy, West Roxbury, MA 02132, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13 Street, Charlestown, MA, 02129, USA
| | - Dmitriy N. Atochin
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital 149 13 Street, Charlestown, MA 02129, USA
- Department of Psychiatry, Boston VA Medical Center West Roxbury, Veterans Affairs Boston Healthcare System and Harvard Medical School, 1400 VFW Pkwy, West Roxbury, MA 02132, USA
| |
Collapse
|
8
|
Fernandes F, Oliveira S, Monteiro F, Gasik M, Silva FS, Sousa N, Carvalho Ó, Catarino SO. Devices used for photobiomodulation of the brain-a comprehensive and systematic review. J Neuroeng Rehabil 2024; 21:53. [PMID: 38600582 PMCID: PMC11007916 DOI: 10.1186/s12984-024-01351-8] [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: 10/18/2023] [Accepted: 04/02/2024] [Indexed: 04/12/2024] Open
Abstract
A systematic review was conducted to determine the trends in devices and parameters used for brain photobiomodulation (PBM). The revised studies included clinical and cadaveric approaches, in which light stimuli were applied to the head and/or neck. PubMed, Scopus, Web of Science and Google Scholar databases were used for the systematic search. A total of 2133 records were screened, from which 97 were included in this review. The parameters that were extracted and analysed in each article were the device design, actuation area, actuation site, wavelength, mode of operation, power density, energy density, power output, energy per session and treatment time. To organize device information, 11 categories of devices were defined, according to their characteristics. The most used category of devices was laser handpieces, which relate to 21% of all devices, while 28% of the devices were not described. Studies for cognitive function and physiological characterisation are the most well defined ones and with more tangible results. There is a lack of consistency when reporting PBM studies, with several articles under defining the stimulation protocol, and a wide variety of parameters used for the same health conditions (e.g., Alzheimer's or Parkinson's disease) resulting in positive outcomes. Standardization for the report of these studies is warranted, as well as sham-controlled comparative studies to determine which parameters have the greatest effect on PBM treatments for different neurological conditions.
Collapse
Affiliation(s)
- Filipa Fernandes
- Center for Micro-ElectroMechanical Systems (CMEMS-UMINHO), University of Minho, Guimarães, Portugal.
| | - Sofia Oliveira
- Center for Micro-ElectroMechanical Systems (CMEMS-UMINHO), University of Minho, Guimarães, Portugal
| | - Francisca Monteiro
- Center for Micro-ElectroMechanical Systems (CMEMS-UMINHO), University of Minho, Guimarães, Portugal
| | - Michael Gasik
- Department of Chemical and Metallurgical Engineering, School of Chemical Engineering, Aalto University Foundation, Espoo, Finland
| | - Filipe S Silva
- Center for Micro-ElectroMechanical Systems (CMEMS-UMINHO), University of Minho, Guimarães, Portugal
- LABBELS-Associate Laboratory, Braga, Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), University of Minho, 4710-057, Braga, Portugal
- ICVS/3BS, PT Government Associate Laboratory, 4710-057, Braga, Portugal
- 2CA-Braga, CVS/3BS, PT Government Associate Laboratory, 4710-057, Braga, Portugal
| | - Óscar Carvalho
- Center for Micro-ElectroMechanical Systems (CMEMS-UMINHO), University of Minho, Guimarães, Portugal
- LABBELS-Associate Laboratory, Braga, Guimarães, Portugal
| | - Susana O Catarino
- Center for Micro-ElectroMechanical Systems (CMEMS-UMINHO), University of Minho, Guimarães, Portugal.
- LABBELS-Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
9
|
Waight JL, Arias N, Jiménez-García AM, Martini M. From functional neuroimaging to neurostimulation: fNIRS devices as cognitive enhancers. Behav Res Methods 2024; 56:2227-2242. [PMID: 37507648 PMCID: PMC10990990 DOI: 10.3758/s13428-023-02144-y] [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] [Accepted: 05/12/2023] [Indexed: 07/30/2023]
Abstract
Functional near-infrared spectroscopy (fNIRS) relies on near-infrared (NIR) light for changes in tissue oxygenation. For decades, this technique has been used in neuroscience to measure cortical activity. However, recent research suggests that NIR light directed to neural populations can modulate their activity through "photobiomodulation" (PBM). Yet, fNIRS is being used exclusively as a measurement tool. By adopting cognitive tests sensitive to prefrontal functioning, we show that a 'classical' fNIRS device, placed in correspondence of the prefrontal cortices of healthy participants, induces faster RTs and better accuracy in some of the indexes considered. A well-matched control group, wearing the same but inactive device, did not show any improvement. Hence, our findings indicate that the 'standard' use of fNIRS devices generates PBM impacting cognition. The neuromodulatory power intrinsic in that technique has been so far completely overlooked, and future studies will need to take this into account.
Collapse
Affiliation(s)
- Jason Lee Waight
- School of Psychology, University of East London, E15 4LZ, London, UK
| | - Natalia Arias
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Denmark Hill, London, SE5 8AF, UK.
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33005, Oviedo, Spain.
- Health Research Institute of the Principality of Asturias (ISPA), 33011, Oviedo, Spain.
- BRABE Group, Department of Psychology, Faculty of Life and Natural Sciences, University of Nebrija, C/del Hostal, 28248, Madrid, Spain.
| | - Ana M Jiménez-García
- BRABE Group, Department of Psychology, Faculty of Life and Natural Sciences, University of Nebrija, C/del Hostal, 28248, Madrid, Spain
| | - Matteo Martini
- School of Psychology, University of East London, E15 4LZ, London, UK.
- Department of Humanities, Letters, Cultural Heritage and Educational Studies, via Arpi, 71121, Foggia, Italy.
| |
Collapse
|
10
|
Joshi H, Sinha P, Bowers D, John JP. Dose response of transcranial near infrared light stimulation on brain functional connectivity and cognition in older adults-A randomized comparison. JOURNAL OF BIOPHOTONICS 2024; 17:e202300215. [PMID: 37776079 DOI: 10.1002/jbio.202300215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/16/2023] [Accepted: 09/27/2023] [Indexed: 10/01/2023]
Abstract
Photobiomodulation, also called low-level light therapy, has been reported in animal studies to have an effect on brain activity and cognition. However, studies in humans regarding its effect on cognition and brain functional connectivity, and the required dose threshold for achieving the same have been very limited. We compared the effects of different doses of photobiomodulation (PBM) on cognition and resting state brain functional connectivity in 25 cognitively normal adults aged 55-70 years. They were randomized to a single session of the sham group, "low-dose" and "high-dose" groups receiving NIR light with transcranial fluence of 26 and 52 J/cm2 respectively, and intranasal fluence of 9 and 18 J/cm2 respectively. There was a significant increase in resting state functional connectivity of the left superior frontal gyrus (SFG) with the left planum temporale (PT), p = 0.0016, and with the left inferior frontal gyrus, pars triangularis, p = 0.0235 in the "high-dose" group only compared to the "sham" group. There was also a significant improvement in visual search and processing speed (p = 0.012) in the "high-dose" group. Replication of these findings in an adequately powered randomized sham-controlled study in healthy older adults can pave the way for clinical application of NIRL as a therapeutic modality in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Himanshu Joshi
- Multimodal Brain Image Analysis Laboratory, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
- Geriatric Clinic and Services, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Preeti Sinha
- Geriatric Clinic and Services, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, India
- Non-invasive Brain Stimulation Services, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Dawn Bowers
- Department of Clinical & Health Psychology, College of Public Health and Health Professions, University of Florida Health Science Center, Gainesville, Florida, USA
- Cognitive Neuroscience Laboratory, Department of Neurology, Fixel Center of Neurological Diseases, University of Florida Health Science Center, Gainesville, Florida, USA
| | - John P John
- Multimodal Brain Image Analysis Laboratory, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| |
Collapse
|
11
|
Shen Q, Guo H, Yan Y. Photobiomodulation for Neurodegenerative Diseases: A Scoping Review. Int J Mol Sci 2024; 25:1625. [PMID: 38338901 PMCID: PMC10855709 DOI: 10.3390/ijms25031625] [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: 11/23/2023] [Revised: 12/27/2023] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Neurodegenerative diseases involve the progressive dysfunction and loss of neurons in the central nervous system and thus present a significant challenge due to the absence of effective therapies for halting or reversing their progression. Based on the characteristics of neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD), which have prolonged incubation periods and protracted courses, exploring non-invasive physical therapy methods is essential for alleviating such diseases and ensuring that patients have an improved quality of life. Photobiomodulation (PBM) uses red and infrared light for therapeutic benefits and functions by stimulating, healing, regenerating, and protecting organizations at risk of injury, degradation, or death. Over the last two decades, PBM has gained widespread recognition as a non-invasive physical therapy method, showing efficacy in pain relief, anti-inflammatory responses, and tissue regeneration. Its application has expanded into the fields of neurology and psychiatry, where extensive research has been conducted. This paper presents a review and evaluation of studies investigating PBM in neurodegenerative diseases, with a specific emphasis on recent applications in AD and PD treatment for both animal and human subjects. Molecular mechanisms related to neuron damage and cognitive impairment are scrutinized, offering valuable insights into PBM's potential as a non-invasive therapeutic strategy.
Collapse
Affiliation(s)
- Qi Shen
- MOE Key Laboratory of Laser Life Science, Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China; (H.G.); (Y.Y.)
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Haoyun Guo
- MOE Key Laboratory of Laser Life Science, Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China; (H.G.); (Y.Y.)
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Yihua Yan
- MOE Key Laboratory of Laser Life Science, Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China; (H.G.); (Y.Y.)
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
12
|
Zorzo C, Rodríguez-Fernández L, Martínez JA, Arias JL. Photobiomodulation increases brain metabolic activity through a combination of 810 and 660 wavelengths: a comparative study in male and female rats. Lasers Med Sci 2024; 39:26. [PMID: 38214813 PMCID: PMC10786747 DOI: 10.1007/s10103-023-03966-0] [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: 08/11/2023] [Accepted: 12/27/2023] [Indexed: 01/13/2024]
Abstract
Photobiomodulation (PBM), an emerging and non-invasive intervention, has been shown to benefit the nervous system by modifying the mitochondrial cytochrome c-oxidase (CCO) enzyme, which has red (620-680 nm) or infrared (760-825 nm) spectral absorption peaks. The effect of a single 810-nm wavelength with a combination of 810 nm and 660 nm lights in the brain metabolic activity of male and female rats was compared. PBM, with a wavelength of 810 nm and a combination of 810 nm and 660 nm, was applied for 5 days on the prefrontal cortex. Then, brain metabolic activity in the prefrontal area, hippocampus, retrosplenial, and parietal cortex was explored. Sex differences were found in cortical and subcortical regions, indicating higher male brain oxidative metabolism, regardless of treatment. CCO activity in the cingulate and prelimbic area, dentate gyrus, retrosplenial and parietal cortex was enhanced in both treatments (810 + 660 nm and 810 nm). Moreover, using the combination of waves, CCO increased in the infralimbic area, and in CA1 and CA3 of the hippocampus. Thus, employment of a single NIR treatment or a combination of red to NIR treatment led to slight differences in CCO activity across the limbic system, suggesting that a combination of lights of the spectrum may be relevant.
Collapse
Affiliation(s)
- Candela Zorzo
- Neuroscience Laboratory, Department of Psychology, University of Oviedo, Oviedo, Spain.
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain.
- ISPA, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.
| | - Lucía Rodríguez-Fernández
- Neuroscience Laboratory, Department of Psychology, University of Oviedo, Oviedo, Spain
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
- ISPA, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Juan A Martínez
- ISPA, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Electronic Technology Area, University of Oviedo, Gijón, Spain
| | - Jorge L Arias
- Neuroscience Laboratory, Department of Psychology, University of Oviedo, Oviedo, Spain
- INEUROPA, Instituto de Neurociencias del Principado de Asturias, Oviedo, Spain
- ISPA, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| |
Collapse
|
13
|
Kashiwagi S, Yokomizo S, Bragin DE, Perle SJ, Kastanenka KV, Gerashchenko D, Atochin DN. Therapeutic Potentials of Near-Infrared II Photobiomodulation to Treat Cerebrovascular Diseases via Nitric Oxide Signalling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1463:195-200. [PMID: 39400823 DOI: 10.1007/978-3-031-67458-7_33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Endothelial dysfunction featuring insufficient endothelial nitric oxide synthase (eNOS) and accompanying nitric oxide (NO) deficiency is implicated in the pathogenesis of cardiovascular diseases. Restoring endothelial NO represents a promising approach to treating cerebrovascular diseases, including stroke. Low-power near-infrared (NIR) light shows diverse beneficial effects, broadly defined as photobiomodulation (PBM). The literature reports that PBM increases bioavailable NO. These lines of evidence indicate that PBM could be used to treat cerebrovascular diseases. Recent investigations revealed that PBM improved stroke outcomes in animal models via augmenting NO signalling and other pathways. However, clinical trials of PBM using NIR light in the NIR-I window (630-900 nm) have yet to demonstrate the beneficial effect of PBM on ischaemic stroke. Since NIR light in the NIR-II window (1000-1700 nm) with the largest penetration depth into tissues compared to NIR I has also been reported to augment NO bioavailability and cerebral blood flow ameliorating stroke injury, PBM using NIR-II light may be suitable for therapeutic use. This new non-pharmacological modality using a physical parameter of NIR-II laser could provide a new avenue for therapeutic strategies for cerebrovascular diseases. Since impaired NO production has been associated with neurological abnormalities, this novel therapeutic approach could be broadly explored to treat various disease conditions such as traumatic brain injury, stroke, and Alzheimer's disease. This review summarises recent findings on PBM in treating stroke and discusses its potential to treat other neurological diseases.
Collapse
Affiliation(s)
- Satoshi Kashiwagi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| | - Shinya Yokomizo
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Denis E Bragin
- Lovelace Biomedical Research Institute, Albuquerque, NM, USA
- Department of Neurology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Stephen J Perle
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ksenia V Kastanenka
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Dmitry Gerashchenko
- Department of Psychiatry, Boston VA Medical Center West Roxbury, Veterans Affairs Boston Healthcare System and Harvard Medical School, West Roxbury, MA, USA
| | - Dmitriy N Atochin
- Department of Psychiatry, Boston VA Medical Center West Roxbury, Veterans Affairs Boston Healthcare System and Harvard Medical School, West Roxbury, MA, USA.
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
O’Donnell CM, Barrett DW, O’Connor P, Gonzalez-Lima F. Prefrontal photobiomodulation produces beneficial mitochondrial and oxygenation effects in older adults with bipolar disorder. Front Neurosci 2023; 17:1268955. [PMID: 38027522 PMCID: PMC10644301 DOI: 10.3389/fnins.2023.1268955] [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: 07/28/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
There is growing evidence of mitochondrial dysfunction and prefrontal cortex (PFC) hypometabolism in bipolar disorder (BD). Older adults with BD exhibit greater decline in PFC-related neurocognitive functions than is expected for age-matched controls, and clinical interventions intended for mood stabilization are not targeted to prevent or ameliorate mitochondrial deficits and neurocognitive decline in this population. Transcranial infrared laser stimulation (TILS) is a non-invasive form of photobiomodulation, in which photons delivered to the PFC photo-oxidize the mitochondrial respiratory enzyme, cytochrome-c-oxidase (CCO), a major intracellular photon acceptor in photobiomodulation. TILS at 1064-nm can significantly upregulate oxidized CCO concentrations to promote differential levels of oxygenated vs. deoxygenated hemoglobin (HbD), an index of cerebral oxygenation. The objective of this controlled study was to use non-invasive broadband near-infrared spectroscopy to assess if TILS to bilateral PFC (Brodmann area 10) produces beneficial effects on mitochondrial oxidative energy metabolism (oxidized CCO) and cerebral oxygenation (HbD) in older (≥50 years old) euthymic adults with BD (N = 15). As compared to sham, TILS to the PFC in adults with BD increased oxidized CCO both during and after TILS, and increased HbD concentrations after TILS. By significantly increasing oxidized CCO and HbD concentrations above sham levels, TILS has the potential ability to stabilize mitochondrial oxidative energy production and prevent oxidative damage in the PFC of adults with BD. In conclusion, TILS was both safe and effective in enhancing metabolic function and subsequent hemodynamic responses in the PFC, which might help alleviate the accelerated neurocognitive decline and dysfunctional mitochondria present in BD.
Collapse
Affiliation(s)
- Courtney M. O’Donnell
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
| | - Douglas W. Barrett
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
| | - Patrick O’Connor
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
| | - F. Gonzalez-Lima
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
- Institute for Neuroscience, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
17
|
Truong NCD, Wang X, Liu H. Temporal and spectral analyses of EEG microstate reveals neural effects of transcranial photobiomodulation on the resting brain. Front Neurosci 2023; 17:1247290. [PMID: 37916179 PMCID: PMC10616257 DOI: 10.3389/fnins.2023.1247290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/25/2023] [Indexed: 11/03/2023] Open
Abstract
Introduction The quantification of electroencephalography (EEG) microstates is an effective method for analyzing synchronous neural firing and assessing the temporal dynamics of the resting state of the human brain. Transcranial photobiomodulation (tPBM) is a safe and effective modality to improve human cognition. However, it is unclear how prefrontal tPBM neuromodulates EEG microstates both temporally and spectrally. Methods 64-channel EEG was recorded from 45 healthy subjects in both 8-min active and sham tPBM sessions, using a 1064-nm laser applied to the right forehead of the subjects. After EEG data preprocessing, time-domain EEG microstate analysis was performed to obtain four microstate classes for both tPBM and sham sessions throughout the pre-, during-, and post-stimulation periods, followed by extraction of the respective microstate parameters. Moreover, frequency-domain analysis was performed by combining multivariate empirical mode decomposition with the Hilbert-Huang transform. Results Statistical analyses revealed that tPBM resulted in (1) a significant increase in the occurrence of microstates A and D and a significant decrease in the contribution of microstate C, (2) a substantial increase in the transition probabilities between microstates A and D, and (3) a substantial increase in the alpha power of microstate D. Discussion These findings confirm the neurophysiological effects of tPBM on EEG microstates of the resting brain, particularly in class D, which represents brain activation across the frontal and parietal regions. This study helps to better understand tPBM-induced dynamic alterations in EEG microstates that may be linked to the tPBM mechanism of action for the enhancement of human cognition.
Collapse
Affiliation(s)
| | | | - Hanli Liu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| |
Collapse
|
18
|
Chaudhari A, Wang X, Wu A, Liu H. Repeated Transcranial Photobiomodulation with Light-Emitting Diodes Improves Psychomotor Vigilance and EEG Networks of the Human Brain. Bioengineering (Basel) 2023; 10:1043. [PMID: 37760145 PMCID: PMC10525861 DOI: 10.3390/bioengineering10091043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/16/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Transcranial photobiomodulation (tPBM) has been suggested as a non-invasive neuromodulation tool. The repetitive administration of light-emitting diode (LED)-based tPBM for several weeks significantly improves human cognition. To understand the electrophysiological effects of LED-tPBM on the human brain, we investigated alterations by repeated tPBM in vigilance performance and brain networks using electroencephalography (EEG) in healthy participants. Active and sham LED-based tPBM were administered to the right forehead of young participants twice a week for four weeks. The participants performed a psychomotor vigilance task (PVT) during each tPBM/sham experiment. A 64-electrode EEG system recorded electrophysiological signals from each participant during the first and last visits in a 4-week study. Topographical maps of the EEG power enhanced by tPBM were statistically compared for the repeated tPBM effect. A new data processing framework combining the group's singular value decomposition (gSVD) with eLORETA was implemented to identify EEG brain networks. The reaction time of the PVT in the tPBM-treated group was significantly improved over four weeks compared to that in the sham group. We observed acute increases in EEG delta and alpha powers during a 10 min LED-tPBM while the participants performed the PVT task. We also found that the theta, beta, and gamma EEG powers significantly increased overall after four weeks of LED-tPBM. Combining gSVD with eLORETA enabled us to identify EEG brain networks and the corresponding network power changes by repeated 4-week tPBM. This study clearly demonstrated that a 4-week prefrontal LED-tPBM can neuromodulate several key EEG networks, implying a possible causal effect between modulated brain networks and improved psychomotor vigilance outcomes.
Collapse
Affiliation(s)
| | | | | | - Hanli Liu
- Department of Bioengineering, University of Texas at Arlington, 500 UTA Blvd, Arlington, TX 76019, USA; (A.C.); (X.W.); (A.W.)
| |
Collapse
|
19
|
Wade ZS, Barrett DW, Davis RE, Nguyen A, Venkat S, Gonzalez-Lima F. Histochemical mapping of the duration of action of photobiomodulation on cytochrome c oxidase in the rat brain. Front Neurosci 2023; 17:1243527. [PMID: 37700747 PMCID: PMC10493319 DOI: 10.3389/fnins.2023.1243527] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/16/2023] [Indexed: 09/14/2023] Open
Abstract
Introduction This is the first study mapping the duration of action of in vivo photobiomodulation (PBM) on cytochrome-c-oxidase (CCO). In cellular bioenergetics, CCO is the terminal rate-limiting enzyme in the mitochondrial electron transport chain, which catalyzes oxygen utilization for aerobic energy production. PBM using transcranial infrared laser stimulation (TILS) is a promising intervention for non-invasively modulating CCO in the brain. TILS of the human prefrontal cortex directly causes CCO photo-oxidation, which is associated with increased cerebral oxygenation and improved cognition. Methods This experiment aimed to map the duration of action of in vivo PBM on CCO activity in discrete neuroanatomic locations within rat brains up to 4 weeks after a single TILS session (50 s, 1064 nm CW, 250 mW/cm2). Control brains from rats treated with a sham session without TILS (laser off) were compared to brains from TILS-treated rats that were collected 1 day, 2 weeks, or 4 weeks post-TILS. Cryostat sections of the 36 collected brains were processed using quantitative enzyme histochemistry and digitally imaged. Densitometric readings of 28 regions of interest were recorded and converted to CCO activity units of oxygen utilization using calibration standards. Data analysis (ANCOVA) compared each laser-treated group to sham with whole-brain average as a covariate. Results The prefrontal infralimbic cortex showed the earliest significant increase in CCO activity between 1-day post-TILS and sham groups, which continued elevated for 2-4 weeks post-TILS. Significant differences in CCO activity between 2-weeks and sham groups were also found in the lateral septum, accumbens core, CA3 of the hippocampus, and the molecular layer of the hippocampus. The medial amygdala showed a significant decrease in CCO activity between 4-weeks and sham. Further analyses showed significant inter-regional CCO activity correlations among the brain regions as the result of TILS, with the most pronounced changes at 4-weeks post-stimulation. Discussion The time course of changes in CCO activity and network connectivity suggested that TILS caused different neuroplasticity types of bioenergetic changes at different time scales, depending on brain region and its depth from the cortex. In conclusion, this controlled CCO histochemical study demonstrated a long-lasting duration of action of PBM in the rat brain.
Collapse
Affiliation(s)
| | | | | | | | | | - F. Gonzalez-Lima
- Department of Psychology and Institute for Neuroscience, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
20
|
Bowen R, Arany PR. Use of either transcranial or whole-body photobiomodulation treatments improves COVID-19 brain fog. JOURNAL OF BIOPHOTONICS 2023; 16:e202200391. [PMID: 37018063 DOI: 10.1002/jbio.202200391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 06/19/2023]
Abstract
There is increasing recognition of post-COVID-19 sequelae involving chronic fatigue and brain fog, for which photobiomodulation (PBM) therapy has been utilized. This open-label, pilot, human clinical study examined the efficacy of two PBM devices, for example, a helmet (1070 nm) for transcranial (tPBM) and a light bed (660 and 850 nm) for whole body (wbPBM), over a 4-week period, with 12 treatments for two separate groups (n = 7 per group). Subjects were evaluated with a neuropsychological test battery, including the Montreal Cognitive Assessment (MoCA), the digit symbol substitution test (DSST), the trail-making tests A and B, the physical reaction time (PRT), and a quantitative electroencephalography system (WAVi), both pre- and post- the treatment series. Each device for PBM delivery was associated with significant improvements in cognitive tests (p < 0.05 and beyond). Changes in WAVi supported the findings. This study outlines the benefits of utilizing PBM therapy (transcranial or whole-body) to help treat long-COVID brain fog.
Collapse
Affiliation(s)
- Robert Bowen
- Shepherd University, Shepherdstown, West Virginia, USA
- West Virginia University, Martinsburg, West Virginia, USA
| | - Praveen R Arany
- Shepherd University, Shepherdstown, West Virginia, USA
- University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
21
|
Torres-Martinez N, Chabardes S, Mitrofanis J. Lights for epilepsy: can photobiomodulation reduce seizures and offer neuroprotection? Neural Regen Res 2023; 18:1423-1426. [PMID: 36571337 PMCID: PMC10075120 DOI: 10.4103/1673-5374.360288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Epilepsy is synonymous with individuals suffering repeated "fits" or seizures. The seizures are triggered by bursts of abnormal neuronal activity, across either the cerebral cortex and/or the hippocampus. In addition, the seizure sites are characterized by considerable neuronal death. Although the factors that generate this abnormal activity and death are not entirely clear, recent evidence indicates that mitochondrial dysfunction plays a central role. Current treatment options include drug therapy, which aims to suppress the abnormal neuronal activity, or surgical intervention, which involves the removal of the brain region generating the seizure activity. However, ~30% of patients are unresponsive to the drugs, while the surgery option is invasive and has a morbidity risk. Hence, there is a need for the development of an effective non-pharmacological and non-invasive treatment for this disorder, one that has few side effects. In this review, we consider the effectiveness of a potential new treatment for epilepsy, known as photobiomodulation, the use of red to near-infrared light on body tissues. Recent studies in animal models have shown that photobiomodulation reduces seizure-like activity and improves neuronal survival. Further, it has an excellent safety record, with little or no evidence of side effects, and it is non-invasive. Taken all together, this treatment appears to be an ideal treatment option for patients suffering from epilepsy, which is certainly worthy of further consideration.
Collapse
Affiliation(s)
| | - Stephan Chabardes
- University of Grenoble Alpes, FDD and CEA-LETI, Clinatec, Grenoble, France
| | - John Mitrofanis
- University of Grenoble Alpes, FDD and CEA-LETI, Clinatec, Grenoble, France
| |
Collapse
|
22
|
Tian T, Wang Z, Chen L, Xu W, Wu B. Photobiomodulation activates undifferentiated macrophages and promotes M1/M2 macrophage polarization via PI3K/AKT/mTOR signaling pathway. Lasers Med Sci 2023; 38:86. [PMID: 36932298 DOI: 10.1007/s10103-023-03753-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 03/13/2023] [Indexed: 03/19/2023]
Abstract
Macrophages are the main mediators of the inflammatory response and play a major role in the onset and maintenance of periodontitis. Studies revealed that photobiomodulation (PBM) can change the polarization state of macrophages and inflammation reduction, although the cellular mechanisms are not fully elucidated. Here, the present study explored the effect of PBM (980 nm) on undifferentiated and M1-type macrophages and the underlying mechanism. RAW264.7 cells were exposed to laser irradiation under different laser parameters (0.5, 5.0, and 10.0 J/cm2) with or without LY294002 (an inhibitor of PI3K pathway). Then, confocal laser microscopy was used to observe cell differentiation; qPCR was performed to examine the gene expression and western blotting was used to detect the protein in the PI3K/AKT/mTOR pathway and activated macrophage markers. The obtained results revealed that 980 nm PBM increased the mRNA expression of iNOS, Il-10, Arg1, and Il-12 along with the inflammatory cytokines Tnfα, IL-1β, and Il-6 in M0-type macrophages in dose-dependent manner. More interestingly, PBM at 5 J/cm2 decreased the mRNA expression of iNOS, Il-12, Tnfα, IL-1β, and Il-6 and increased the expression of Arg1 and Il-10 by M1-type macrophages, along with the elevated expression of phosphorylation of AKT and mTOR. Moreover, PBM-induced M1-type macrophage polarization was significantly attenuated via LY294002 treatment. These suggest that 980 nm PBM could activate M0-type macrophages and increase M2/M1 ratio via the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Tian Tian
- Department of Endodontics, Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, 143 Dongzong Road, Pingshan District, Shenzhen, 518118, China
| | - Ziting Wang
- Department of pediatric dentistry, Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, 143 Dongzong Road, Pingshan District, Shenzhen, 518118, China
| | - Leyi Chen
- Department of Endodontics, Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, 143 Dongzong Road, Pingshan District, Shenzhen, 518118, China
- School of Stomatology, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Wenan Xu
- Department of pediatric dentistry, Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, 143 Dongzong Road, Pingshan District, Shenzhen, 518118, China.
- School of Stomatology, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| | - Buling Wu
- Department of Endodontics, Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, 143 Dongzong Road, Pingshan District, Shenzhen, 518118, China.
- School of Stomatology, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| |
Collapse
|
23
|
Pan WT, Liu PM, Ma D, Yang JJ. Advances in photobiomodulation for cognitive improvement by near-infrared derived multiple strategies. J Transl Med 2023; 21:135. [PMID: 36814278 PMCID: PMC9945713 DOI: 10.1186/s12967-023-03988-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
Cognitive function is an important ability of the brain, but cognitive dysfunction can easily develop once the brain is injured in various neuropathological conditions or diseases. Photobiomodulation therapy is a type of noninvasive physical therapy that is gradually emerging in the field of neuroscience. Transcranial photobiomodulation has been commonly used to regulate neural activity in the superficial cortex. To stimulate deeper brain activity, advanced photobiomodulation techniques in conjunction with photosensitive nanoparticles have been developed. This review addresses the mechanisms of photobiomodulation on neurons and neural networks and discusses the advantages, disadvantages and potential applications of photobiomodulation alone or in combination with photosensitive nanoparticles. Photobiomodulation and its associated strategies may provide new breakthrough treatments for cognitive improvement.
Collapse
Affiliation(s)
- Wei-tong Pan
- grid.412633.10000 0004 1799 0733Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China ,grid.207374.50000 0001 2189 3846Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, 450052 China ,Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, 450000 People’s Republic of China
| | - Pan-miao Liu
- grid.412633.10000 0004 1799 0733Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China ,grid.207374.50000 0001 2189 3846Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, 450052 China ,Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, 450000 People’s Republic of China
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine & Intensive Care, Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK. .,National Clinical Research Center for Child Health, Hangzhou, 310052, China.
| | - Jian-jun Yang
- grid.412633.10000 0004 1799 0733Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China ,grid.207374.50000 0001 2189 3846Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, 450052 China ,Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, 450000 People’s Republic of China
| |
Collapse
|
24
|
Brain Waste Removal System and Sleep: Photobiomodulation as an Innovative Strategy for Night Therapy of Brain Diseases. Int J Mol Sci 2023; 24:ijms24043221. [PMID: 36834631 PMCID: PMC9965491 DOI: 10.3390/ijms24043221] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Emerging evidence suggests that an important function of the sleeping brain is the removal of wastes and toxins from the central nervous system (CNS) due to the activation of the brain waste removal system (BWRS). The meningeal lymphatic vessels (MLVs) are an important part of the BWRS. A decrease in MLV function is associated with Alzheimer's and Parkinson's diseases, intracranial hemorrhages, brain tumors and trauma. Since the BWRS is activated during sleep, a new idea is now being actively discussed in the scientific community: night stimulation of the BWRS might be an innovative and promising strategy for neurorehabilitation medicine. This review highlights new trends in photobiomodulation of the BWRS/MLVs during deep sleep as a breakthrough technology for the effective removal of wastes and unnecessary compounds from the brain in order to increase the neuroprotection of the CNS as well as to prevent or delay various brain diseases.
Collapse
|
25
|
Zaizar ED, Papini S, Gonzalez-Lima F, Telch MJ. Singular and combined effects of transcranial infrared laser stimulation and exposure therapy on pathological fear: a randomized clinical trial. Psychol Med 2023; 53:908-917. [PMID: 34284836 PMCID: PMC9976021 DOI: 10.1017/s0033291721002270] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/16/2021] [Accepted: 05/21/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Preclinical findings suggest that transcranial infrared laser stimulation (TILS) improves fear extinction learning and cognitive function by enhancing prefrontal cortex (PFC) oxygen metabolism. These findings prompted our investigation of treating pathological fear using this non-invasive stimulation approach either alone to the dorsolateral PFC (dlPFC), or to the ventromedial PFC (vmPFC) in combination with exposure therapy. METHODS Volunteers with pathological fear of either enclosed spaces, contamination, public speaking, or anxiety-related bodily sensations were recruited for this randomized, single-blind, sham-controlled trial with four arms: (a) Exposure + TILS_vmPFC (n = 29), (b) Exposure + sham TILS_vmPFC (n = 29), (c) TILS_dlPFC alone (n = 26), or (d) Sham TILS _dlPFC alone (n = 28). Post-treatment assessments occurred immediately following treatment. Follow-up assessments occurred 2 weeks after treatment. RESULTS A total of 112 participants were randomized [age range: 18-63 years; 96 females (85.71%)]. Significant interactions of Group × Time and Group × Context indicated differential treatment effects on retention (i.e. between time-points, averaged across contexts) and on generalization (i.e. between contexts, averaged across time-points), respectively. Among the monotherapies, TILS_dlPFC outperformed SHAM_dlPFC in the initial context, b = -13.44, 95% CI (-25.73 to -1.15), p = 0.03. Among the combined treatments, differences between EX + TILS_vmPFC and EX + SHAM_vmPFC were non-significant across all contrasts. CONCLUSIONS TILS to the dlPFC, one of the PFC regions implicated in emotion regulation, resulted in a context-specific benefit as a monotherapy for reducing fear. Contrary to prediction, TILS to the vmPFC, a region implicated in fear extinction memory consolidation, did not enhance exposure therapy outcome.
Collapse
Affiliation(s)
- Eric D. Zaizar
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
- Institute for Mental Health Research, The University of Texas at Austin, Austin, TX, USA
| | - Santiago Papini
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
- Institute for Mental Health Research, The University of Texas at Austin, Austin, TX, USA
| | - F. Gonzalez-Lima
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
- Institute for Neuroscience, The University of Texas at Austin, Austin, TX, USA
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Michael J. Telch
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
- Institute for Mental Health Research, The University of Texas at Austin, Austin, TX, USA
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
26
|
Chen L, Xue J, Zhao Q, Liang X, Zheng L, Fan Z, Souare ISJ, Suo Y, Wei X, Ding D, Mao Y. A Pilot Study of Near-Infrared Light Treatment for Alzheimer's Disease. J Alzheimers Dis 2023; 91:191-201. [PMID: 36373323 DOI: 10.3233/jad-220866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Laboratory investigations have demonstrated that near-infrared (NIR) light treatment can reduce amyloid-β burden in models of Alzheimer's disease (AD). However, previous clinical studies are rather insufficient. OBJECTIVE Before starting a large-scale clinical trial, we performed a pilot study to characterize the efficacy of NIR light for AD patients. METHODS Twenty participants with mild to moderate AD were assigned randomly to the intervention (1060-1080 nm and 800-820 nm NIR light treatment for 12 weeks) or control group (without sham treatment). Safety and efficacy were evaluated at baseline, week 4, 8, and 12, and 4 weeks after treatment. RESULTS In the intervention and control groups at week 12, mean changes from baseline on the Alzheimer's Disease Assessment Scale-Cognitive (ADAS-Cog) were -3.1 and -1.3 (p = 0.5689). Mean changes from baseline on the Activities of Daily Living (ADL) were -3.6 versus 3.1 (p = 0.0437). Mean changes from baseline on the Mini-Mental State Examination (MMSE) were 4.4 versus 1.0 (p = 0.0253). The percentage of participants who exhibited a change larger than 4 points from baseline to week 12 was determined for the intervention and control groups on the ADAS-Cog (57% versus 29%), ADL (29% versus 0%), and MMSE (57% versus 14%). Treatment with NIR light did not increase the incidence of adverse events in participants. CONCLUSION NIR light treatment appears to be safe and potentially beneficial for AD patients. It improved cognitive function and activities of daily living. The preliminary data encouraged us to launch a large-sample, multicenter, double-blind clinical trial.
Collapse
Affiliation(s)
- Liang Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Jun Xue
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Qianhua Zhao
- Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoniu Liang
- Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Li Zheng
- Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhen Fan
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Ibrahima Sory Jnr Souare
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Yuanzhen Suo
- Biomedical Pioneering Innovation Center, Peking University, Beijing, China
| | - Xunbin Wei
- Department of Biomedical Engineering, Peking University, Beijing, China
| | - Ding Ding
- Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| |
Collapse
|
27
|
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
|
28
|
Lee TL, Ding Z, Chan AS. Can transcranial photobiomodulation improve cognitive function? A systematic review of human studies. Ageing Res Rev 2023; 83:101786. [PMID: 36371017 DOI: 10.1016/j.arr.2022.101786] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/12/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Transcranial photobiomodulation (tPBM) has been studied for over a decade as a possible cognitive intervention. OBJECTIVE To evaluate the effect of tPBM for enhancing human cognitive function in healthy adults and remediating impaired cognitive function in adults with cognitive disorders. METHODS A systematic literature search from three electronic databases (PubMed, Scopus, Web of Science) was conducted from 1987 to May 2022. The cognitive function being evaluated included learning and memory, attention, executive function, language, and global cognitive function. RESULTS Of the 35 studies identified, 29 (82.9 %) studies reported positive improvement in cognitive functions after tPBM. All nine studies on participants with subjective memory complaints, mild cognitive impairment, and dementia, showed positive outcomes. Seven (87.5 %) studies on traumatic brain injury (TBI) patients also showed positive results. A series of clinical trials on stroke patients showed positive trends on improved neurological deficit at first, but was prematurely terminated later at phase III due to the lack of statistical significance. One of the most common protocols for clinical populations employed devices delivering near-infrared light (810 nm), the irradiance of 20-25 mW/cm2, and fluence of 1-10 J/cm2. While this was common, the reviewed protocols also included other wavelengths of light ranging from visible, red (630-635 nm) to invisible near-infrared maximum wavelengths of 1060-1068 nm. CONCLUSIONS tPBM seems to improve cognitive function. However, only half of the reviewed clinical trials were randomized control trials, further investigation is warranted.
Collapse
Affiliation(s)
- Tsz-Lok Lee
- Neuropsychology Laboratory, Department of Psychology, The Chinese University of Hong Kong, Hong Kong, China
| | - Zihan Ding
- Neuropsychology Laboratory, Department of Psychology, The Chinese University of Hong Kong, Hong Kong, China
| | - Agnes S Chan
- Neuropsychology Laboratory, Department of Psychology, The Chinese University of Hong Kong, Hong Kong, China; Research Centre for Neuropsychological Well-Being, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
29
|
Pope NJ, Denton ML. Differential effects of 808-nm light on electron transport chain enzymes in isolated mitochondria: Implications for photobiomodulation initiation. Mitochondrion 2023; 68:15-24. [PMID: 36371074 DOI: 10.1016/j.mito.2022.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 10/06/2022] [Accepted: 11/04/2022] [Indexed: 11/10/2022]
Abstract
Photobiomodulation is a term for using low-power red to near-infrared light to stimulate a variety of positive biological effects. Though the scientific and clinical acceptance of PBM as a therapeutic intervention has increased dramatically in recent years, the molecular underpinnings of the effect remain poorly understood. The putative chromophore for PBM effects is cytochrome c oxidase. It is postulated that light absorption at cytochrome c oxidase initiates a signaling cascade involving ATP and generation of reactive oxygen species (ROS), which subsequently results in improved cellular robustness. However, this hypothesis is largely based on inference and indirect evidence, and the precise molecular mechanisms that govern how photon absorption leads to these downstream effects remain poorly understood. We conducted low-power PBM-type light exposures of isolated mitochondria to 808 nm NIR light, at a number of irradiances. NIR exposure was found to enhance the activity of complex IV, depress the activity of complex III, and had no effect on the activity of complex II. Further, examining the dose-response of complex IV we found NIR enhancement did not exhibit irradiance reciprocity, indicating the effect on complex IV may not have direct photochemical basis. In summary, this research presents a novel method to interrogate the earliest stages of PBM in the mitochondria, and a unique window into the corresponding molecular mechanism(s) of induction.
Collapse
Affiliation(s)
| | - Michael L Denton
- Air Force Research Laboratory, Bioeffects Division, JBSA Fort Sam Houston, TX 78234, United States.
| |
Collapse
|
30
|
Abstract
This review deals with an unwelcome reality about several forms of dementia, including Alzheimer's disease- that these dementias are caused, in part or whole, by the aging of the vasculature. Since the vasculature ages in us all, dementia is our fate, sealed by the realit!ies of the circulation; it is not a disease with a cure pending. Empirically, cognitive impairment before our 7th decade is uncommon and considered early, while a diagnosis in our 11th decade is late but common in that cohort (>40%). Projections from earlier ages suggest that the prevalence of dementia in people surviving into their 12th decade exceeds 80%. We address the question why so few of many interventions known to delay dementia are recognized as therapy; and we try to resolve this few-and-many paradox, identifying opportunities for better treatment, especially pre-diagnosis. The idea of dementia as a fate is resisted, we argue, because it negates the hope of a cure. But the price of that hope is lost opportunity. An approach more in line with the evidence, and more likely to limit suffering, is to understand the damage that accumulates with age in the cerebral vasculature and therefore in the brain, and which eventually gives rise to cognitive symptoms in late life, too often leading to dementia. We argue that hope should be redirected to delaying that damage and with it the onset of cognitive loss; and, for each individual, it should be redirected to a life-long defense of their brain.
Collapse
Affiliation(s)
- Marcus J Andersson
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Jonathan Stone
- School of Medical Sciences and Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
31
|
Zhao C, Li D, Kong Y, Liu H, Hu Y, Niu H, Jensen O, Li X, Liu H, Song Y. Transcranial photobiomodulation enhances visual working memory capacity in humans. SCIENCE ADVANCES 2022; 8:eabq3211. [PMID: 36459562 PMCID: PMC10936045 DOI: 10.1126/sciadv.abq3211] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 10/18/2022] [Indexed: 06/17/2023]
Abstract
Transcranial photobiomodulation (tPBM) is a safe and noninvasive intervention that has shown promise for improving cognitive performance. Whether tPBM can modulate brain activity and thereby enhance working memory (WM) capacity in humans remains unclear. In this study, we found that 1064-nm tPBM applied to the right prefrontal cortex (PFC) improves visual working memory capacity and increases occipitoparietal contralateral delay activity (CDA). The CDA set-size effect during retention mediated the effect between the 1064-nm tPBM and subsequent WM capacity. The behavioral benefits and the corresponding changes in the CDA set-size effect were absent with tPBM at a wavelength of 852 nm or with stimulation of the left PFC. Our findings provide converging evidence that 1064-nm tPBM applied to the right PFC can improve WM capacity.
Collapse
Affiliation(s)
- Chenguang Zhao
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- Center for Cognition and Neuroergonomics, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University at Zhuhai, Guangdong, China
- School of Systems Science, Beijing Normal University, Beijing, China
| | - Dongwei Li
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- Centre for Human Brain Health, School of Psychology, University of Birmingham, Birmingham, UK
| | - Yuanjun Kong
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Hongyu Liu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Yiqing Hu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Haijing Niu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Ole Jensen
- Centre for Human Brain Health, School of Psychology, University of Birmingham, Birmingham, UK
| | - Xiaoli Li
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- Center for Cognition and Neuroergonomics, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University at Zhuhai, Guangdong, China
| | - Hanli Liu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA
| | - Yan Song
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| |
Collapse
|
32
|
Monteiro F, Carvalho Ó, Sousa N, Silva FS, Sotiropoulos I. Photobiomodulation and visual stimulation against cognitive decline and Alzheimer's disease pathology: A systematic review. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12249. [PMID: 36447479 PMCID: PMC9695760 DOI: 10.1002/trc2.12249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 12/01/2021] [Accepted: 12/15/2021] [Indexed: 11/27/2022]
Abstract
Introduction Given the ineffectiveness of the available drug treatment against Alzheimer disease (AD), light-based therapeutic modalities have been increasingly receiving attention with photobiomodulation (PBM) and, more recently, visual stimulation (VS) being among the most promising approaches. However, the PBM and VS light parameters tested so far, as well as their outcomes, vary a lot with conflicting results being reported. Methods Based on Scopus, PubMed, and Web of Science databases search, this systematic review summarizes, compares, and discusses 43 cell, animal, and human studies of PBM and VS related to cognitive decline and AD pathology. Results Preclinical work suggests that PBM with 640±30-nm light and VS at 40 Hz attenuates Aβ and Tau pathology and improves neuronal and synaptic plasticity with most studies pointing towards enhancement of degradation/clearance mechanisms in the brain of AD animal models. Despite the gap of the translational evidence for both modalities, the few human studies performed so far support the use of PBM at 810-870 nm light pulsing at 40 Hz for improving brain network connectivity and memory in older subjects and AD patients, while 40 Hz VS in humans seems to improve cognition; further clinical investigation is urgently required to clarify the beneficial impact of PBM and VS in AD patients. Discussion This review highlights PBM and VS as promising light-based therapeutic approaches against AD brain neuropathology and related cognitive decline, clarifying the most effective light parameters for further preclinical and clinical testing and use. Highlights Light-based brain stimulation produces neural entrainment and reverts neuronal damageBrain PBM and VS attenuate AD neuropathologyPMB and VS are suggested to improve cognitive performance in AD patients and animal modelsLight stimulation represents a promising therapeutic strategy against neurodegeneration.
Collapse
Affiliation(s)
- Francisca Monteiro
- Center for Microelectromechanical Systems (CMEMS)Campus AzurémUniversity of MinhoGuimarãesPortugal
- ICVS/3B's ‐ PT Government Associate LaboratoryBraga/GuimarãesPortugal
- LABBELS—Associate LaboratoryUniversity of MinhoGuimarãesPortugal
| | - Óscar Carvalho
- Center for Microelectromechanical Systems (CMEMS)Campus AzurémUniversity of MinhoGuimarãesPortugal
- LABBELS—Associate LaboratoryUniversity of MinhoGuimarãesPortugal
| | - Nuno Sousa
- ICVS/3B's ‐ PT Government Associate LaboratoryBraga/GuimarãesPortugal
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of MinhoCampus de GualtarBragaPortugal
| | - Filipe S. Silva
- Center for Microelectromechanical Systems (CMEMS)Campus AzurémUniversity of MinhoGuimarãesPortugal
- LABBELS—Associate LaboratoryUniversity of MinhoGuimarãesPortugal
| | - Ioannis Sotiropoulos
- ICVS/3B's ‐ PT Government Associate LaboratoryBraga/GuimarãesPortugal
- Life and Health Sciences Research Institute (ICVS)School of MedicineUniversity of MinhoCampus de GualtarBragaPortugal
- Institute of Biosciences and ApplicationsNCSR DemokritosAthensGreece
| |
Collapse
|
33
|
Shahdadian S, Wang X, Wanniarachchi H, Chaudhari A, Truong NCD, Liu H. Neuromodulation of brain power topography and network topology by prefrontal transcranial photobiomodulation. J Neural Eng 2022; 19:10.1088/1741-2552/ac9ede. [PMID: 36317341 PMCID: PMC9795815 DOI: 10.1088/1741-2552/ac9ede] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/31/2022] [Indexed: 11/18/2022]
Abstract
Objective.Transcranial photobiomodulation (tPBM) has shown promising benefits, including cognitive improvement, in healthy humans and in patients with Alzheimer's disease. In this study, we aimed to identify key cortical regions that present significant changes caused by tPBM in the electroencephalogram (EEG) oscillation powers and functional connectivity in the healthy human brain.Approach. A 64-channel EEG was recorded from 45 healthy participants during a 13 min period consisting of a 2 min baseline, 8 min tPBM/sham intervention, and 3 min recovery. After pre-processing and normalizing the EEG data at the five EEG rhythms, cluster-based permutation tests were performed for multiple comparisons of spectral power topographies, followed by graph-theory analysis as a topological approach for quantification of brain connectivity metrics at global and nodal/cluster levels.Main results. EEG power enhancement was observed in clusters of channels over the frontoparietal regions in the alpha band and the centroparietal regions in the beta band. The global measures of the network revealed a reduction in synchronization, global efficiency, and small-worldness of beta band connectivity, implying an enhancement of brain network complexity. In addition, in the beta band, nodal graphical analysis demonstrated significant increases in local information integration and centrality over the frontal clusters, accompanied by a decrease in segregation over the bilateral frontal, left parietal, and left occipital regions.Significance.Frontal tPBM increased EEG alpha and beta powers in the frontal-central-parietal regions, enhanced the complexity of the global beta-wave brain network, and augmented local information flow and integration of beta oscillations across prefrontal cortical regions. This study sheds light on the potential link between electrophysiological effects and human cognitive improvement induced by tPBM.
Collapse
Affiliation(s)
| | | | | | | | | | - Hanli Liu
- Authors to whom any correspondence should be addressed,
| |
Collapse
|
34
|
Lights on for Autism: Exploring Photobiomodulation as an Effective Therapeutic Option. Neurol Int 2022; 14:884-893. [PMID: 36412693 PMCID: PMC9680350 DOI: 10.3390/neurolint14040071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 12/14/2022] Open
Abstract
Autism is a neurodevelopmental condition that starts in childhood and continues into adulthood. The core characteristics include difficulties with social interaction and communication, together with restricted and repetitive behaviours. There are a number of key abnormalities of brain structure and function that trigger these behavioural patterns, including an imbalance of functional connectivity and synaptic transmission, neuronal death, gliosis and inflammation. In addition, autism has been linked to alterations in the gut microbiome. Unfortunately, as it stands, there are few treatment options available for patients. In this mini-review, we consider the effectiveness of a potential new treatment for autism, known as photobiomodulation, the therapeutic use of red to near infrared light on body tissues. This treatment has been shown in a range of pathological conditions-to improve the key changes that characterise autism, including the functional connectivity and survival patterns of neurones, the patterns of gliosis and inflammation and the composition of the microbiome. We highlight the idea that photobiomodulation may form an ideal treatment option for autism, one that is certainly worthy of further investigation.
Collapse
|
35
|
Vrankic M, Vlahinić S, Šverko Z, Markovinović I. EEG-Validated Photobiomodulation Treatment of Dementia-Case Study. SENSORS (BASEL, SWITZERLAND) 2022; 22:s22197555. [PMID: 36236654 PMCID: PMC9573554 DOI: 10.3390/s22197555] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 05/15/2023]
Abstract
In this article, we perform a case study of the impact of photobiomodulation (PBM) on brain power spectrum and connectivity in an elderly person with a Self Administered Gerocognitive Exam (SAGE) score indicating probable memory and thinking disorder. First, we designed and realized the prototype of a near-infrared (NIR) device for PBM. Analysing the alpha band of the power spectrum, we found a positive long-term effect in nine out of sixteen electrodes in the eyes-open condition (OE) and in twelve out of sixteen electrodes in the eyes-closed condition (CE), while in the theta band, a positive long-term effect was found in nine out of sixteen electrodes for OE and seven out of sixteen electrodes for CE. When considering the theta-alpha ratio (TAR), the positive long-term effect is found on thirteen of sixteen electrodes for OE and on fourteen of sixteen electrodes for CE. A connectivity analysis using the imaginary component of the complex Pearson correlation coefficient (imCPCC) was also performed, and a global efficiency measure based on connectivity matrices with thresholds was calculated. The global efficiency calculated for the long-term effect was higher than before stimulation by a factor of 5.24 for the OE condition and by a factor of 1.25 for the CE condition. This case study suggests that PBM could have positive effects on improving desired brain activity, measured as improvement in power spectrum and connectivity measures in theta and alpha bands, for elderly people with memory and thinking disorders.
Collapse
|
36
|
Buendía D, Guncay T, Oyanedel M, Lemus M, Weinstein A, Ardiles ÁO, Marcos J, Fernandes A, Zângaro R, Muñoz P. The Transcranial Light Therapy Improves Synaptic Plasticity in the Alzheimer’s Disease Mouse Model. Brain Sci 2022; 12:brainsci12101272. [PMID: 36291206 PMCID: PMC9599908 DOI: 10.3390/brainsci12101272] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/10/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is the main cause of dementia worldwide. Emerging non-invasive treatments such as photobiomodulation target the mitochondria to minimize brain damage, improving cognitive functions. In this work, an experimental design was carried out to evaluate the effect of transcranial light therapy (TLTC) on synaptic plasticity (SP) and cognitive functions in an AD animal model. Twenty-three mice were separated into two general groups: an APP/PS1 (ALZ) transgenic group and a wild-type (WT) group. Each group was randomly subdivided into two subgroups: mice with and without TLTC, depending on whether they would undergo treatment with TLTC. Cognitive function, measured through an object recognition task, showed non-significant improvement after TLTC. SP, on the other hand, was evaluated using four electrophysiological parameters from the Schaffer-CA1 collateral hippocampal synapses: excitatory field potentials (fEPSP), paired pulse facilitation (PPF), long-term depression (LTD), and long-term potentiation (LTP). An improvement was observed in subjects treated with TLTC, showing higher levels of LTP than those transgenic mice that were not exposed to the treatment. Therefore, the results obtained in this work showed that TLTC could be an efficient non-invasive treatment for AD-associated SP deficits.
Collapse
Affiliation(s)
- Débora Buendía
- Programa de Engenharia Biomédica, Instituto de Engenharia Biomédica, Universidade Anhembi Morumbi—UAM, Rua Casa do Ator, 294, Sao Paulo 04546-001, Brazil
- Escuela de Ingeniería Civil Biomédica, Facultad de Ingeniería, Universidad de Valparaíso, General Cruz 222, Valparaíso 2362905, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Centro de Inovação, Tecnología e Educação—CITÉ, Parque Tecnológico de São José dos Campos, Estrada Dr. Altino Bondesan 500, São José dos Campos 12247-016, Brazil
| | - Tatiana Guncay
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Macarena Oyanedel
- Escuela de Ingeniería Civil Biomédica, Facultad de Ingeniería, Universidad de Valparaíso, General Cruz 222, Valparaíso 2362905, Chile
| | - Makarena Lemus
- Escuela de Ingeniería Civil Biomédica, Facultad de Ingeniería, Universidad de Valparaíso, General Cruz 222, Valparaíso 2362905, Chile
| | - Alejandro Weinstein
- Escuela de Ingeniería Civil Biomédica, Facultad de Ingeniería, Universidad de Valparaíso, General Cruz 222, Valparaíso 2362905, Chile
| | - Álvaro O. Ardiles
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Angamos 655, Viña del Mar 2540064, Chile
| | - José Marcos
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Escuela de Ciencias Agrícolas y Veterinarias, Universidad Viña del Mar, Viña del Mar 2572007, Chile
| | - Adriana Fernandes
- Programa de Engenharia Biomédica, Instituto de Engenharia Biomédica, Universidade Anhembi Morumbi—UAM, Rua Casa do Ator, 294, Sao Paulo 04546-001, Brazil
- Centro de Inovação, Tecnología e Educação—CITÉ, Parque Tecnológico de São José dos Campos, Estrada Dr. Altino Bondesan 500, São José dos Campos 12247-016, Brazil
| | - Renato Zângaro
- Programa de Engenharia Biomédica, Instituto de Engenharia Biomédica, Universidade Anhembi Morumbi—UAM, Rua Casa do Ator, 294, Sao Paulo 04546-001, Brazil
- Centro de Inovação, Tecnología e Educação—CITÉ, Parque Tecnológico de São José dos Campos, Estrada Dr. Altino Bondesan 500, São José dos Campos 12247-016, Brazil
- Correspondence: (R.Z.); (P.M.); Tel.: +55-12-997830843 (R.Z.); +56-969028160 (P.M.)
| | - Pablo Muñoz
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Angamos 655, Viña del Mar 2540064, Chile
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de Valparaíso, Angamos 655, Viña del Mar 2540064, Chile
- Correspondence: (R.Z.); (P.M.); Tel.: +55-12-997830843 (R.Z.); +56-969028160 (P.M.)
| |
Collapse
|
37
|
McGee C, Liebert A, Herkes G, Bicknell B, Pang V, McLachlan CS, Kiat H. Protocol for randomized controlled trial to evaluate the safety and feasibility of a novel helmet to deliver transcranial light emitting diodes photobiomodulation therapy to patients with Parkinson’s disease. Front Neurosci 2022; 16:945796. [PMID: 36061601 PMCID: PMC9428720 DOI: 10.3389/fnins.2022.945796] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/06/2022] [Indexed: 11/15/2022] Open
Abstract
Introduction Parkinson’s disease (PD) is the second most common, progressive, and debilitating neurodegenerative disease associated with aging and the most common movement disorder. Photobiomodulation (PBM), the use of non-thermal light for therapeutic purposes using laser or light emitting diodes (LED) is an emerging non-invasive treatment for a diverse range of neurological conditions. The main objectives of this clinical trial are to investigate the feasibility, safety, tolerability, and efficacy of a novel transcranial LED helmet device (the “PDNeuro”) in the alleviation of symptoms of PD. Methods and analysis This is a 24-week, two-arm, triple-blinded randomized placebo-controlled clinical trial of a novel transcranial “PDNeuro” LED Helmet, comparing an active helmet to a sham helmet device. In a survey, 40 PD participants with Hoehn and Yahr Stage I–III during ON periods will be enrolled and randomly assigned into two groups. Both groups will be monitored weekly for the safety and tolerability of the “PDNeuro” LED Helmet. Clinical signs and symptoms assessed will include mobility, fine motor skills and cognition, with data collected at baseline, 12 weeks, and 24 weeks. Assessment tools include the TUG, UPDRS, and MoCA all validated for use in PD patients. Patient’s adherence to the device usage and participant drop out will be monitored weekly. At 12 weeks both placebo and treatment groups will crossover and placebo participants offered the treatment. The main indicator for clinical efficacy of the “PDneuro” Helmet is evidence of sustained improvements in motor and non-motor symptoms obtained from participant self-reported changes, carer reporting of changes and objective reassessment by the investigators. The outcomes will assist in a future larger randomized trial design. Clinical Trial Registration [https://www.anzctr.org.au], identifier [12621001722886].
Collapse
Affiliation(s)
- Claire McGee
- Faculty of Health Sciences, Torrens University, Sydney, NSW, Australia
| | - Ann Liebert
- School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
- Department of Research and Governance, San Hospital, Wahroonga, NSW, Australia
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW, Australia
- *Correspondence: Ann Liebert,
| | - Geoffrey Herkes
- Department of Neurology, San Hospital, Wahroonga, NSW, Australia
- Australian National University, Canberra, ACT, Australia
| | - Brian Bicknell
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW, Australia
| | - Vincent Pang
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW, Australia
| | | | - Hosen Kiat
- NICM Health Research Institute, University of Western Sydney, Westmead, NSW, Australia
- Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, NSW, Australia
- College of Health and Medicine, Australian National University, Canberra, ACT, Australia
- Cardiac Health Institute, Sydney, NSW, Australia
| |
Collapse
|
38
|
Moro C, Valverde A, Dole M, Hoh Kam J, Hamilton C, Liebert A, Bicknell B, Benabid AL, Magistretti P, Mitrofanis J. The effect of photobiomodulation on the brain during wakefulness and sleep. Front Neurosci 2022; 16:942536. [PMID: 35968381 PMCID: PMC9366035 DOI: 10.3389/fnins.2022.942536] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/08/2022] [Indexed: 11/26/2022] Open
Abstract
Over the last seventy years or so, many previous studies have shown that photobiomodulation, the use of red to near infrared light on body tissues, can improve central and peripheral neuronal function and survival in both health and in disease. These improvements are thought to arise principally from an impact of photobiomodulation on mitochondrial and non-mitochondrial mechanisms in a range of different cell types, including neurones. This impact has downstream effects on many stimulatory and protective genes. An often-neglected feature of nearly all of these improvements is that they have been induced during the state of wakefulness. Recent studies have shown that when applied during the state of sleep, photobiomodulation can also be of benefit, but in a different way, by improving the flow of cerebrospinal fluid and the clearance of toxic waste-products from the brain. In this review, we consider the potential differential effects of photobiomodulation dependent on the state of arousal. We speculate that the effects of photobiomodulation is on different cells and systems depending on whether it is applied during wakefulness or sleep, that it may follow a circadian rhythm. We speculate further that the arousal-dependent photobiomodulation effects are mediated principally through a biophoton – ultra-weak light emission – network of communication and repair across the brain.
Collapse
Affiliation(s)
- Cecile Moro
- FDD and CEA-LETI, Clinatec, Université Grenoble Alpes, Grenoble, France
| | - Audrey Valverde
- FDD and CEA-LETI, Clinatec, Université Grenoble Alpes, Grenoble, France
| | - Marjorie Dole
- FDD and CEA-LETI, Clinatec, Université Grenoble Alpes, Grenoble, France
| | - Jaimie Hoh Kam
- FDD and CEA-LETI, Clinatec, Université Grenoble Alpes, Grenoble, France
| | | | - Ann Liebert
- Governance and Research Department, Sydney Adventist Hospital, Sydney, NSW, Australia
| | - Brian Bicknell
- Faculty of Health Sciences, Australian Catholic University, Sydney, NSW, Australia
| | | | - Pierre Magistretti
- FDD and CEA-LETI, Clinatec, Université Grenoble Alpes, Grenoble, France
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - John Mitrofanis
- FDD and CEA-LETI, Clinatec, Université Grenoble Alpes, Grenoble, France
- Institute of Ophthalmology, University College London, London, United Kingdom
- *Correspondence: John Mitrofanis,
| |
Collapse
|
39
|
Zhang R, Zhou T, Samanta S, Luo Z, Li S, Xu H, Qu J. Synergistic photobiomodulation with 808-nm and 1064-nm lasers to reduce the β-amyloid neurotoxicity in the in vitro Alzheimer's disease models. FRONTIERS IN NEUROIMAGING 2022; 1:903531. [PMID: 37555169 PMCID: PMC10406259 DOI: 10.3389/fnimg.2022.903531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/11/2022] [Indexed: 08/10/2023]
Abstract
BACKGROUND In Alzheimer's disease (AD), the deposition of β-amyloid (Aβ) plaques is closely associated with the neuronal apoptosis and activation of microglia, which may result in the functional impairment of neurons through pro-inflammation and over-pruning of the neurons. Photobiomodulation (PBM) is a non-invasive therapeutic approach without any conspicuous side effect, which has shown promising attributes in the treatment of chronic brain diseases such as AD by reducing the Aβ burden. However, neither the optimal parameters for PBM treatment nor its exact role in modulating the microglial functions/activities has been conclusively established yet. METHODS An inflammatory stimulation model of Alzheimer's disease (AD) was set up by activating microglia and neuroblastoma with fibrosis β-amyloid (fAβ) in a transwell insert system. SH-SY5Y neuroblastoma cells and BV2 microglial cells were irradiated with the 808- and 1,064-nm lasers, respectively (a power density of 50 mW/cm2 and a dose of 10 J/cm2) to study the PBM activity. The amount of labeled fAβ phagocytosed by microglia was considered to assess the microglial phagocytosis. A PBM-induced neuroprotective study was conducted with the AD model under different laser parameters to realize the optimal condition. Microglial phenotype, microglial secretions of the pro-inflammatory and anti-inflammatory factors, and the intracellular Ca2+ levels in microglia were studied in detail to understand the structural and functional changes occurring in the microglial cells of AD model upon PBM treatment. CONCLUSION A synergistic PBM effect (with the 808- and 1,064-nm lasers) effectively inhibited the fAβ-induced neurotoxicity of neuroblastoma by promoting the viability of neuroblastoma and regulating the intracellular Ca2+ levels of microglia. Moreover, the downregulation of Ca2+ led to microglial polarization with an M2 phenotype, which promotes the fAβ phagocytosis, and resulted in the upregulated expression of anti-inflammatory factors and downregulated expression of inflammatory factors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| |
Collapse
|
40
|
Qu X, Li L, Zhou X, Dong Q, Liu H, Liu H, Yang Q, Han Y, Niu H. Repeated transcranial photobiomodulation improves working memory of healthy older adults: behavioral outcomes of poststimulation including a three-week follow-up. NEUROPHOTONICS 2022; 9:035005. [PMID: 36177151 PMCID: PMC9514540 DOI: 10.1117/1.nph.9.3.035005] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 09/12/2022] [Indexed: 06/16/2023]
Abstract
SIGNIFICANCE Decline in cognitive ability is a significant issue associated with healthy aging. Transcranial photobiomodulation (tPBM) is an emerging non-invasive neuromodulation technique and has shown promise to overcome this challenge. AIM This study aimed to investigate the effects of seven-day repeated tPBM, compared to those of single tPBM and baseline, on improving N -back working memory in healthy older adults and to evaluate the persistent efficacy of repeated tPBM. APPROACH In a sham-controlled and within-subject design, 61 healthy older adults were recruited to participate in a longitudinal study involving an experimental baseline, seven days of tPBM treatment (12 min daily, 1064-nm laser, 250 mW / cm 2 ) in the left dorsolateral prefrontal cortex and three weeks of follow-ups. Behavioral performance in the N -back ( N = 1,2 , 3 ) was recorded poststimulation during the baseline, the first and seventh days of the tPBM session, and the three weekly follow-ups. A control group with 25 participants was included in this study to rule out the practice and placebo effects. The accuracy rate and response time were used in the statistical analysis. RESULTS Repeated and single tPBM significantly improved accuracy rate in 1- and 3-back tasks and decreased response time in 3-back compared to the baseline. Moreover, the repeated tPBM resulted in a significantly higher improvement in accuracy rate than the single tPBM. These improvements in accuracy rate and response time lasted at least three weeks following repeated tPBM. In contrast, the control group showed no significant improvement in behavioral performance. CONCLUSIONS This study demonstrated that seven-day repeated tPBM improved the working memory of healthy older adults more efficiently, with the beneficial effect lasting at least three weeks. These findings provide fundamental evidence that repeated tPBM may be a potential intervention for older individuals with memory decline.
Collapse
Affiliation(s)
- Xiujuan Qu
- Beijing Normal University, IDG/McGovern Institute for Brain Research, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing, China
| | - Lexuan Li
- Beijing Normal University, IDG/McGovern Institute for Brain Research, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing, China
| | - Xiaohan Zhou
- Beijing Normal University, IDG/McGovern Institute for Brain Research, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing, China
| | - Qi Dong
- Beijing Normal University, IDG/McGovern Institute for Brain Research, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing, China
| | - Hanli Liu
- University of Texas at Arlington, Department of Bioengineering, Arlington, Texas, United States
| | - Hesheng Liu
- Medical University of South Carolina, Department of Neuroscience, Charleston, South Carolina, United States
| | - Qin Yang
- Xuanwu Hospital of Capital Medical University, Department of Neurology, Beijing, China
| | - Ying Han
- Xuanwu Hospital of Capital Medical University, Department of Neurology, Beijing, China
- Hainan University, School of Biomedical Engineering, Haikou, China
- Beijing Institute for Brain Disorders, Center of Alzheimer’s Disease, Beijing, China
- National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Haijing Niu
- Beijing Normal University, IDG/McGovern Institute for Brain Research, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing, China
| |
Collapse
|
41
|
Truong NCD, Wang X, Wanniarachchi H, Liu H. Enhancement of Frequency-Specific Hemodynamic Power and Functional Connectivity by Transcranial Photobiomodulation in Healthy Humans. Front Neurosci 2022; 16:896502. [PMID: 35757526 PMCID: PMC9226485 DOI: 10.3389/fnins.2022.896502] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/09/2022] [Indexed: 12/03/2022] Open
Abstract
Transcranial photobiomodulation (tPBM) has been considered a safe and effective brain stimulation modality being able to enhance cerebral oxygenation and neurocognitive function. To better understand the underlying neurophysiological effects of tPBM in the human brain, we utilized a 111-channel functional near infrared spectroscopy (fNIRS) system to map cerebral hemodynamic responses over the whole head to 8-min tPBM with 1,064-nm laser given on the forehead of 19 healthy participants. Instead of analyzing broad-frequency hemodynamic signals (0–0.2 Hz), we investigated frequency-specific effects of tPBM on three infra-slow oscillation (ISO) components consisting of endogenic, neurogenic, and myogenic vasomotions. Significant changes induced by tPBM in spectral power of oxygenated hemoglobin concentration (Δ[HbO]), functional connectivity (FC), and global network metrics at each of the three ISO frequency bands were identified and mapped topographically for frequency-specific comparisons. Our novel findings revealed that tPBM significantly increased endogenic Δ[HbO] powers over the right frontopolar area near the stimulation site. Also, we demonstrated that tPBM enabled significant enhancements of endogenic and myogenic FC across cortical regions as well as of several global network metrics. These findings were consistent with recent reports and met the expectation that myogenic oscillation is highly associated with endothelial activity, which is stimulated by tPBM-evoked nitric oxide (NO) release.
Collapse
Affiliation(s)
- Nghi Cong Dung Truong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Xinlong Wang
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Hashini Wanniarachchi
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Hanli Liu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| |
Collapse
|
42
|
Cardoso FDS, Gonzalez-Lima F, Coimbra NC. Mitochondrial Photobiomodulation as a Neurotherapeutic Strategy for Epilepsy. Front Neurol 2022; 13:873496. [PMID: 35785362 PMCID: PMC9243228 DOI: 10.3389/fneur.2022.873496] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Fabrízio dos Santos Cardoso
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo, Ribeirão Preto, Brazil
- *Correspondence: Fabrízio dos Santos Cardoso
| | - Francisco Gonzalez-Lima
- Department of Psychology and Institute for Neuroscience, The University of Texas at Austin, Austin, TX, United States
- Francisco Gonzalez-Lima
| | - Norberto Cysne Coimbra
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, Ribeirão Preto Medical School of the University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
43
|
Transcranial Photobiomodulation Therapy for Sexual Dysfunction Associated with Depression or Induced by Antidepressant Medications. PHOTONICS 2022. [DOI: 10.3390/photonics9050330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sexual dysfunction (SD) is frequently encountered in patients suffering from depression. There is a bidirectional relationship between various types of SD and depression, so the presence or treatment of one condition may exacerbate or improve the other condition. The most frequent sexual problem in untreated depressed patients is declining sexual desire, while in treated depressed patients it is difficulties with erection/ejaculation and with orgasm. Numerous classes of neuropsychiatric medications, commonly used in depressed patients—such as antidepressant, antipsychotic, alpha sympathetic, and opioid drugs—may cause SD. Photobiomodulation (PBM) therapy, also called low-level light/laser therapy, is a novel neuromodulation technique for neuropsychiatric conditions, such as depression. Transcranial PBM (tPBM) targets the cellular metabolism—through the mitochondrial respiratory enzyme, cytochrome c oxidase—and has numerous cellular and physiological beneficial effects on the central nervous system. This paper represents a comprehensive review of the application of tPBM to SD, coexisting with depression or induced by antidepressant medications.
Collapse
|
44
|
Wang X, Wanniarachchi H, Wu A, Liu H. Combination of Group Singular Value Decomposition and eLORETA Identifies Human EEG Networks and Responses to Transcranial Photobiomodulation. Front Hum Neurosci 2022; 16:853909. [PMID: 35620152 PMCID: PMC9127055 DOI: 10.3389/fnhum.2022.853909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Transcranial Photobiomodulation (tPBM) has demonstrated its ability to alter electrophysiological activity in the human brain. However, it is unclear how tPBM modulates brain electroencephalogram (EEG) networks and is related to human cognition. In this study, we recorded 64-channel EEG from 44 healthy humans before, during, and after 8-min, right-forehead, 1,064-nm tPBM or sham stimulation with an irradiance of 257 mW/cm2. In data processing, a novel methodology by combining group singular value decomposition (gSVD) with the exact low-resolution brain electromagnetic tomography (eLORETA) was implemented and performed on the 64-channel noise-free EEG time series. The gSVD+eLORETA algorithm produced 11 gSVD-derived principal components (PCs) projected in the 2D sensor and 3D source domain/space. These 11 PCs took more than 70% weight of the entire EEG signals and were justified as 11 EEG brain networks. Finally, baseline-normalized power changes of each EEG brain network in each EEG frequency band (delta, theta, alpha, beta and gamma) were quantified during the first 4-min, second 4-min, and post tPBM/sham periods, followed by comparisons of frequency-specific power changes between tPBM and sham conditions. Our results showed that tPBM-induced increases in alpha powers occurred at default mode network, executive control network, frontal parietal network and lateral visual network. Moreover, the ability to decompose EEG signals into individual, independent brain networks facilitated to better visualize significant decreases in gamma power by tPBM. Many similarities were found between the cortical locations of SVD-revealed EEG networks and fMRI-identified resting-state networks. This consistency may shed light on mechanistic associations between tPBM-modulated brain networks and improved cognition outcomes.
Collapse
|
45
|
O'Donnell CM, Barrett DW, Fink LH, Garcia-Pittman EC, Gonzalez-Lima F. Transcranial Infrared Laser Stimulation Improves Cognition in Older Bipolar Patients: Proof of Concept Study. J Geriatr Psychiatry Neurol 2022; 35:321-332. [PMID: 33525934 DOI: 10.1177/0891988720988906] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This is the first study to examine if transcranial infrared laser stimulation (TILS) improves cognition in older euthymic bipolar patients, who exhibit greater cognitive decline than is expected for age-matched controls. TILS is a non-invasive novel form of photobiomodulation that augments prefrontal oxygenation and improves cognition in young adults by upregulating the mitochondrial respiratory enzyme cytochrome-c-oxidase. We used a crossover sham-controlled design to examine if TILS to bilateral prefrontal cortex produces beneficial effects on cognition in 5 euthymic bipolar patients (ages 60-85). We measured cognitive flexibility, verbal fluency, working memory, sustained attention and impulsivity with tasks that have been shown to differentiate between healthy older adults and older bipolar adults. We found TILS-induced improvements in cognitive performance on the tasks that measure cognitive flexibility and impulsivity, after 5 weekly sessions of TILS. We concluded that TILS appeared both safe and effective in helping alleviate the accelerated cognitive decline present in older bipolar patients.
Collapse
Affiliation(s)
- Courtney M O'Donnell
- Department of Psychology and Institute for Neuroscience, 7067University of Texas at Austin, TX, USA
| | - Douglas W Barrett
- Department of Psychology and Institute for Neuroscience, 7067University of Texas at Austin, TX, USA
| | - Latham H Fink
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, 12330University of Texas at Austin, TX, USA
| | - Erica C Garcia-Pittman
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, 12330University of Texas at Austin, TX, USA
| | - Francisco Gonzalez-Lima
- Department of Psychology and Institute for Neuroscience, 7067University of Texas at Austin, TX, USA.,Department of Psychiatry and Behavioral Sciences, Dell Medical School, 12330University of Texas at Austin, TX, USA
| |
Collapse
|
46
|
Zhao X, Du W, Jiang J, Han Y. Brain Photobiomodulation Improves Sleep Quality in Subjective Cognitive Decline: A Randomized, Sham-Controlled Study. J Alzheimers Dis 2022; 87:1581-1589. [PMID: 35491787 DOI: 10.3233/jad-215715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Sleep appears to be a sensitive biomarker that facilitates early detection and effective intervention for Alzheimer’s disease, while subjective cognitive decline (SCD) is a risk factor for Alzheimer’s disease. Prefrontal cortex atrophy is associated with both sleep disruption and cognitive decline. Transcranial brain photobiomodulation (PBM) therapy can enhance frontal cortex oxygen consumption, increasing frontal cortex mediated memory function. Objective: This study aimed to test whether PBM therapy targeting the frontal cortex could improve sleep and cognitive function in SCD. Methods: Fifty-eight SCDs were divided into the PBM group (N = 32) in which real light therapy was administered and a sham light therapy group (N = 26). All the participants received either real light or sham light therapy for 6 days consecutively, while the sleep data were recorded. The n-back task was employed to measure each participant’s working memory. Results: We found no differences in sleep efficiency change (F = 211, p = 0.279), REM stage percent change (F = 420, p = 0.91), and wake-up time (F = 212, p = 0.277) between the two groups. The sleep efficiency and REM were improved within the true light group on the fifth day. The true light group perform better than the control group in the n-back test, the accuracy was higher in the 2-back test (88.6% versus 79.6%, p = 0.001), and the reaction time in 1-back was shorter (544.80±202.00 versus 592.87±222.05, p = 0.003). Conclusion: After five days of PBM therapy targeting the prefrontal cortex, sleep efficiency and N-back cognitive performance were improved on the fifth day.
Collapse
Affiliation(s)
- Xing Zhao
- Department of NeurologyXuanwu Hospital of Capital Medical University, Beijing, China
| | - Wenying Du
- Department of NeurologyXuanwu Hospital of Capital Medical University, Beijing, China
| | - Jiehui Jiang
- Institute of Biomedical Engineering School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Ying Han
- Department of NeurologyXuanwu Hospital of Capital Medical University, Beijing, China
- Biomedical Engineering Institute, Hainan University, Haikou, China
- Center of Alzheimer’s Disease, Beijing Institute for Brain Diseases, Beijing, China
| |
Collapse
|
47
|
Cardoso FDS, Barrett DW, Wade Z, Gomes da Silva S, Gonzalez-Lima F. Photobiomodulation of Cytochrome c Oxidase by Chronic Transcranial Laser in Young and Aged Brains. Front Neurosci 2022; 16:818005. [PMID: 35368252 PMCID: PMC8971717 DOI: 10.3389/fnins.2022.818005] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/23/2022] [Indexed: 12/15/2022] Open
Abstract
In cellular bioenergetics, cytochrome c oxidase (CCO) is the enzyme responsible for oxygen consumption in the mitochondrial electron transport chain, which drives oxidative phosphorylation for adenosine triphosphate (ATP) production. CCO is also the major intracellular acceptor of photons in the light wavelengths used for photobiomodulation (PBM). Brain function is critically dependent on oxygen consumption by CCO for ATP production. Therefore, our objectives were (1) to conduct the first detailed brain mapping study of the effects of PBM on regional CCO activity, and (2) to compare the chronic effects of PBM on young and aged brains. Specifically, we used quantitative CCO histochemistry to map the differences in CCO activity of brain regions in healthy young (4 months old) and aged (20 months old) rats from control groups with sham stimulation and from treated groups with 58 consecutive days of transcranial laser PBM (810 nm wavelength and 100 mW power). We found that aging predominantly decreased regional brain CCO activity and systems-level functional connectivity, while the chronic laser stimulation predominantly reversed these age-related effects. We concluded that chronic PBM modified the effects of aging by causing the CCO activity on brain regions in laser-treated aged rats to reach levels similar to those found in young rats. Given the crucial role of CCO in bioenergetics, PBM may be used to augment brain and behavioral functions of older individuals by improving oxidative energy metabolism.
Collapse
Affiliation(s)
- Fabrízio dos Santos Cardoso
- Department of Psychology, Institute for Neuroscience, The University of Texas at Austin, Austin, TX, United States
- Núcleo de Pesquisas Tecnológicas, Universidade de Mogi das Cruzes, Mogi das Cruzes, Brazil
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Douglas W. Barrett
- Department of Psychology, Institute for Neuroscience, The University of Texas at Austin, Austin, TX, United States
| | - Zachary Wade
- Department of Psychology, Institute for Neuroscience, The University of Texas at Austin, Austin, TX, United States
| | - Sérgio Gomes da Silva
- Núcleo de Pesquisas Tecnológicas, Universidade de Mogi das Cruzes, Mogi das Cruzes, Brazil
- Centro Universitário UNIFAMINAS, Muriaé, Brazil
- Hospital do Câncer de Muriaé, Fundação Cristiano Varella, Muriaé, Brazil
| | - F. Gonzalez-Lima
- Department of Psychology, Institute for Neuroscience, The University of Texas at Austin, Austin, TX, United States
- *Correspondence: F. Gonzalez-Lima,
| |
Collapse
|
48
|
Jordan S, Zielinski M, Kortylewski M, Kuhn T, Bystritsky A. Noninvasive Delivery of Biologicals to the Brain. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2022; 20:64-70. [PMID: 35746928 PMCID: PMC9063603 DOI: 10.1176/appi.focus.20210028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In the past, psychotherapy and neuropharmacological approaches have been the most common treatments for disordered thoughts, moods, and behaviors. One new path of brain therapeutics is in the deployment of noninvasive approaches designed to reprogram brain function at the cellular level. Treatment at the cellular level may be considered for a wide array of disorders, ranging from mood disorders to neurodegenerative disorders. Brain-targeted biological therapy may provide minimally invasive and accurate delivery of treatment. The present article discusses the hurdles and advances that characterize the pathway to this goal.
Collapse
|
49
|
Huang LD. Brighten the Future: Photobiomodulation and Optogenetics. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2022; 20:36-44. [PMID: 35746943 PMCID: PMC9063588 DOI: 10.1176/appi.focus.20210025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Safe, noninvasive, and effective treatments for brain conditions are everyone's dream. Low-level light therapy (LLLT) based on the photobiomodulation (PBM) phenomenon has recently been adopted in practice, with solid scientific evidence. Optogenetics provides high spatiotemporal resolution to precisely switch on and off a particular circuitry in the brain. However, there are currently no human trials of optogenetics on the human brain. These two approaches-PBM and optogenetics-are promising photonic treatments that target the brain using completely different technologies. PBM is based on the mitochondrial reaction to the photons for up- or downregulation on the cytochrome c oxidase synthase in cellular respiration. It is safe, noninvasive, and good for long-term treatments, with wide applications using light wavelengths ranging from 650 nm to ≈1,100 nm, the red to near-infrared range. Optogenetics is based on the expression of engineered opsins on targeted tissues through viral vectors. The opsins are engineered to be sensors, actuators, or switches and could be precisely controlled by light wavelength ranging from 450 nm to ≈650 nm, the visible light range. The penetration of visible light is limited, and thus the photons cannot be applied directly outside the head without surgical means to create a physical window. PBM using near-infrared light could reach deeper tissues for light directly applied outside the head. Detailed scientific foundations and the state of the art for both technologies are reviewed. Ongoing developments are discussed to provide insight for future research and applications.
Collapse
|
50
|
Belova AN, Israelyan YA, Sushin VO, Shabanova MA, Rezenova AM. [Transcranial photobiomodulation in therapy of neurodegenerative diseases of the brain: theoretical background and clinical effectiveness]. VOPROSY KURORTOLOGII, FIZIOTERAPII, I LECHEBNOI FIZICHESKOI KULTURY 2022; 98:61-67. [PMID: 34965698 DOI: 10.17116/kurort20219806161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Transcranial photobiomodulation (tPBM) is a form of light therapy that uses monochromatic visible and infrared light from non-ionizing radiation sources (lasers, LEDs) placed on the scalp, forehead, or intranasally to project light directly to target areas of the brain. Accumulated experimental and clinical data indicate the safety and potential efficacy of tPBM in some central nervous system diseases.This article briefly reviews the general concepts of tPBM, the results of experimental and clinical studies on the efficacy of tPBM in Alzheimer's disease, Parkinson's disease, and brain stroke. The possible mechanisms of the tPBM therapeutic effect and the need to choose optimal exposure parameters are discussed. Although the evidence base regarding the efficacy of tPBM in neurodegenerative and vascular brain diseases is still insufficient, analysis of the published data justifies considering tPBM as a promising method of adjuvant therapy for some central nervous system diseases.
Collapse
Affiliation(s)
- A N Belova
- Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - Yu A Israelyan
- Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - V O Sushin
- Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - M A Shabanova
- Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - A M Rezenova
- Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| |
Collapse
|