101
|
Bhattacharya M, Dutta A. Computational Modeling of the Photon Transport, Tissue Heating, and Cytochrome C Oxidase Absorption during Transcranial Near-Infrared Stimulation. Brain Sci 2019; 9:brainsci9080179. [PMID: 31357574 PMCID: PMC6721367 DOI: 10.3390/brainsci9080179] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/15/2019] [Accepted: 07/23/2019] [Indexed: 12/16/2022] Open
Abstract
Transcranial near-infrared stimulation (tNIRS) has been proposed as a tool to modulate cortical excitability. However, the underlying mechanisms are not clear where the heating effects on the brain tissue needs investigation due to increased near-infrared (NIR) absorption by water and fat. Moreover, the risk of localized heating of tissues (including the skin) during optical stimulation of the brain tissue is a concern. The challenge in estimating localized tissue heating is due to the light interaction with the tissues' constituents, which is dependent on the combination ratio of the scattering and absorption properties of the constituent. Here, apart from tissue heating that can modulate the cortical excitability ("photothermal effects"); the other mechanism reported in the literature is the stimulation of the mitochondria in the cells which are active in the adenosine triphosphate (ATP) synthesis. In the mitochondrial respiratory chain, Complex IV, also known as the cytochrome c oxidase (CCO), is the unit four with three copper atoms. The absorption peaks of CCO are in the visible (420-450 nm and 600-700 nm) and the near-infrared (760-980 nm) spectral regions, which have been shown to be promising for low-level light therapy (LLLT), also known as "photobiomodulation". While much higher CCO absorption peaks in the visible spectrum can be used for the photobiomodulation of the skin, 810 nm has been proposed for the non-invasive brain stimulation (using tNIRS) due to the optical window in the NIR spectral region. In this article, we applied a computational approach to delineate the "photothermal effects" from the "photobiomodulation", i.e., to estimate the amount of light absorbed individually by each chromophore in the brain tissue (with constant scattering) and the related tissue heating. Photon migration simulations were performed for motor cortex tNIRS based on a prior work that used a 500 mW cm - 2 light source placed on the scalp. We simulated photon migration at 630 nm and 700 nm (red spectral region) and 810 nm (near-infrared spectral region). We found a temperature increase in the scalp below 0.25 °C and a minimal temperature increase in the gray matter less than 0.04 °C at 810 nm. Similar heating was found for 630 nm and 700 nm used for LLLT, so photothermal effects are postulated to be unlikely in the brain tissue.
Collapse
Affiliation(s)
- Mahasweta Bhattacharya
- Department of Biomedical Engineering, University at Buffalo SUNY, Buffalo, NY 14260, USA.
| | - Anirban Dutta
- Department of Biomedical Engineering, University at Buffalo SUNY, Buffalo, NY 14260, USA
| |
Collapse
|
102
|
Chang LY, Fan SMY, Liao YC, Wang WH, Chen YJ, Lin SJ. Proteomic Analysis Reveals Anti-Fibrotic Effects of Blue Light Photobiomodulation on Fibroblasts. Lasers Surg Med 2019; 52:358-372. [PMID: 31321797 DOI: 10.1002/lsm.23137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND OBJECTIVES This study was aimed at determining the effects of blue light photobiomodulation on primary adult mouse dermal fibroblasts (AMDFs) and the associated signaling pathways. STUDY DESIGN/MATERIALS AND METHODS Cultured AMDFs from adult C57BL/6 mice were irradiated by blue light from a light-emitting diode (wavelength = 463 ± 50 nm; irradiance = 5 mW/cm2 ; energy density = 4-8 J/cm2 ). The cells were analyzed using mass spectrometry for proteomics/phosphoproteomics, AlamarBlue assay for mitochondrial activity, time-lapse video for cell migration, quantitative polymerase chain reaction for gene expression, and immunofluorescence for protein expression. RESULTS Proteomic/phosphoproteomic analysis showed inhibition of extracellular signal-regulated kinases/mammalian target of rapamycin and casein kinase 2 pathways, cell motility-related networks, and multiple metabolic processes, including carbon metabolism, biosynthesis of amino acid, glycolysis/gluconeogenesis, and the pentose phosphate pathway. Functional analysis demonstrated inhibition of mitochondrial activities, cell migration, and mitosis. Expression of growth promoting insulin-like growth factor 1 and fibrosis-related genes, including transforming growth factor β1 (TGFβ1) and collagen type 1 ɑ2 chain diminished. Protein expression of α-smooth muscle actin, an important regulator of myofibroblast functions, was also suppressed. CONCLUSIONS Low-level blue light exerted suppressive effects on AMDFs, including suppression of mitochondrial activity, metabolism, cell motility, proliferation, TGFβ1 levels, and collagen I production. Low-level blue light can be a potential treatment for the prevention and reduction of tissue fibrosis, such as hypertrophic scar and keloids. Lasers Surg. Med. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lo-Yu Chang
- School of Medicine, College of Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Sabrina Mai-Yi Fan
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Yen-Chen Liao
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan.,Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd, Taipei 115, Taiwan
| | - Wei-Hung Wang
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Yu-Ju Chen
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan.,Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd, Taipei 115, Taiwan
| | - Sung-Jan Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan.,Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, No. 7, Chung-Shan South Road, Taipei 100, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, No. 7, Chung-Shan South Road, Taipei 100, Taiwan
| |
Collapse
|
103
|
Facchin F, Canaider S, Tassinari R, Zannini C, Bianconi E, Taglioli V, Olivi E, Cavallini C, Tausel M, Ventura C. Physical energies to the rescue of damaged tissues. World J Stem Cells 2019; 11:297-321. [PMID: 31293714 PMCID: PMC6600852 DOI: 10.4252/wjsc.v11.i6.297] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/24/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023] Open
Abstract
Rhythmic oscillatory patterns sustain cellular dynamics, driving the concerted action of regulatory molecules, microtubules, and molecular motors. We describe cellular microtubules as oscillators capable of synchronization and swarming, generating mechanical and electric patterns that impact biomolecular recognition. We consider the biological relevance of seeing the inside of cells populated by a network of molecules that behave as bioelectronic circuits and chromophores. We discuss the novel perspectives disclosed by mechanobiology, bioelectromagnetism, and photobiomodulation, both in term of fundamental basic science and in light of the biomedical implication of using physical energies to govern (stem) cell fate. We focus on the feasibility of exploiting atomic force microscopy and hyperspectral imaging to detect signatures of nanomotions and electromagnetic radiation (light), respectively, generated by the stem cells across the specification of their multilineage repertoire. The chance is reported of using these signatures and the diffusive features of physical waves to direct specifically the differentiation program of stem cells in situ, where they already are resident in all the tissues of the human body. We discuss how this strategy may pave the way to a regenerative and precision medicine without the needs for (stem) cell or tissue transplantation. We describe a novel paradigm based upon boosting our inherent ability for self-healing.
Collapse
Affiliation(s)
- Federica Facchin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), School of Medicine, University of Bologna, Bologna 40100, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Silvia Canaider
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), School of Medicine, University of Bologna, Bologna 40100, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Riccardo Tassinari
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Chiara Zannini
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Eva Bianconi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Valentina Taglioli
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Elena Olivi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | - Claudia Cavallini
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| | | | - Carlo Ventura
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), School of Medicine, University of Bologna, Bologna 40100, Italy
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems, CNR, Bologna 40100, Italy
| |
Collapse
|
104
|
Vakhrushev IV, Yusupov VI, Raeva OS, Pyatnitskiy MA, Bagratashvili VN. Effect of Low-Level Laser Irradiation on Proliferative Activity of Wharton's Jelly Mesenchymal Stromal Cells. Bull Exp Biol Med 2019; 167:136-139. [PMID: 31183648 DOI: 10.1007/s10517-019-04477-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Indexed: 12/15/2022]
Abstract
We studied the effect of low-level laser irradiation on proliferative activity of cultured human Wharton's jelly mesenchymal stromal sells. Cells were irradiated with a solid-state laser emitting at 650 nm; irradiation doses were 0.04, 0.4, or 4 J/cm2. Laser irradiation was performed once at the start of the cell proliferation experiment or daily throughout the experiment. Cells were cultured for 7 days. The number of viable cells was assessed using the MTT test. An increase in cell proliferative activity was detected after daily laser irradiations; the maximum stimulating effect was achieved at a dose of 0.04 J/cm2. These results substantiate medical use of lasers for expansion of cells intended for transplantation.
Collapse
Affiliation(s)
- I V Vakhrushev
- V. N. Orekhovich Research Institute of Biomedical Chemistry, Moscow, Russia. .,Private Institution Laboratory for Biotechnological Research "3D Bioprinting Solutions", Moscow, Russia.
| | - V I Yusupov
- Institute of Photonics Technologies, Federal Research Center for Crystallography and Photonics, Russian Academy of Sciences, Troitsk, Russia
| | - O S Raeva
- V. N. Orekhovich Research Institute of Biomedical Chemistry, Moscow, Russia
| | - M A Pyatnitskiy
- V. N. Orekhovich Research Institute of Biomedical Chemistry, Moscow, Russia
| | - V N Bagratashvili
- Institute of Photonics Technologies, Federal Research Center for Crystallography and Photonics, Russian Academy of Sciences, Troitsk, Russia
| |
Collapse
|
105
|
Explanation of Osteoblastic Differentiation of Stem Cells by Photo Biomodulation Using the Resonant Recognition Model. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9101979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Differentiation of stem cells into different tissues is a promising approach to treat a large number of diseases, as well as for tissue transplantation and repair. It has been shown that parathyroid hormone, similarly to stromal self-derived factor, and the radiation of specific electromagnetic frequencies of blue and green light, can encourage stem cell differentiation into osteoblasts. Here, we analysed parathyroid hormone, its receptor and stromal self-derived factor using the Resonant Recognition Model, which proposes that protein function is based on specific frequencies of electromagnetic radiation within ultra-violet, visible, infra-red and far infra-red light. The purpose of this research is to predict the characteristic frequencies related to parathyroid hormone activities, particularly differentiation of stem cells into osteoblasts. We have found that the most effective wavelength for stem cell differentiation would be 502 nm, which is between 420 nm and 540 nm, already experimentally proven to be effective in stimulating osteoblast differentiation. Thus, we propose that wavelength radiation of 502 nm will be even more efficient for differentiation of stem cells into osteoblasts.
Collapse
|
106
|
Zheng M, Kim DY, Sung JH. Ion channels and transporters in adipose-derived stem cells. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2019. [DOI: 10.1007/s40005-018-00413-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
107
|
Hosseinpour S, Fekrazad R, Arany PR, Ye Q. Molecular impacts of photobiomodulation on bone regeneration: A systematic review. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 149:147-159. [PMID: 31002851 DOI: 10.1016/j.pbiomolbio.2019.04.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/16/2019] [Indexed: 12/13/2022]
Abstract
Photobiomodulation (PBM) encompasses a light application aimed to increase healing process, tissue regeneration, and reducing inflammation and pain. PBM is specifically aimed to modify the expression of cellular molecules; however, PBM impacts on cellular and molecular pathways especially in bone regenerative medicine have been investigated in scattered different studies. The purpose of the current study is to systematically review evidence on molecular impact of PBM on bone regeneration. A comprehensive electronic search in Medline, Scopus, EMBASE, EBSCO, Cochrane library, web of science, and google scholar was conducted from January 1975 to October 2018 limited to English language publications on administrations of photobiomodulation for bone regeneration which evaluated biological factors. In addition, hand search of selected journals was done to retrieve all articles. This systematic review was performed based on PRISMA guideline. Among these studies, five articles reported in vitro results, twelve articles were in vivo, and three of them were clinical trials. The data tabulated according to the type of markers (osteogenic markers, angiogenic markers, growth factors, and inflammation mediators). PBM's effects depend on many parameters which energy density is more important than the others. PBM can significantly enhance expression of osteocalcin, collagen, RUNX-2, vascular endothelial growth factor, bone morphogenic proteins, and COX-2. Although since the heterogeneity of the studies and their limitations, an evidence-based decision for definite therapeutic application of PBM is still unattainable, the findings of our review can help other researchers to ameliorate their study design and elect more efficient approach for their investigation.
Collapse
Affiliation(s)
- Sepanta Hosseinpour
- School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia; Department of Periodontology, Dental Faculty - Laser Research Center in Medical Sciences, AJA University of Medical Sciences, 19839, Fatemi, Tehran, Iran.
| | - Reza Fekrazad
- Department of Periodontology, Dental Faculty - Laser Research Center in Medical Sciences, AJA University of Medical Sciences, 19839, Fatemi, Tehran, Iran; International Network for Photo Medicine and Photo Dynamic Therapy (INPMPDT), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Praveen R Arany
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, NY, 14214, USA.
| | - Qingsong Ye
- School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia.
| |
Collapse
|
108
|
Hanna R, Agas D, Benedicenti S, Ferrando S, Laus F, Cuteri V, Lacava G, Sabbieti MG, Amaroli A. A Comparative Study Between the Effectiveness of 980 nm Photobiomodulation Delivered by Hand-Piece With Gaussian vs. Flat-Top Profiles on Osteoblasts Maturation. Front Endocrinol (Lausanne) 2019; 10:92. [PMID: 30842754 PMCID: PMC6391326 DOI: 10.3389/fendo.2019.00092] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/01/2019] [Indexed: 12/19/2022] Open
Abstract
Photobiomodulation (PBM) is a clinically accepted tool in regenerative medicine and dentistry to improve tissue healing and repair and to restore the functional disability. The current in vitro study aimed to investigate the photobiomodulatory effects of 980 nm wavelength (the real energy at the target: ~0.9 W, ~0.9 W/cm2, 60 s, ~55 J/cm2 and a single energy ~55 J in CW) on MC3T3-E1 pre-osteoblast, delivered with flattop profile in comparison to the standard profile. The laser groupings and their associated energies were: Group 1 - once per week (total energy 110 J); Group 2 - three times per week (alternate day) (total energy 330 J); Group 3 - five times per week (total energy 550 J). The metabolic activity and the osteoblasts maturation were analyzed by alkaline phosphatase assay, alizarin red S histological staining, immunoblot and/or double immunolabeling analysis for Bcl2, Bax, Runx-2, Osx, Dlx5, osteocalcin, and collagen Type 1. Our data, for the first time, prove that laser irradiation of 980 nm wavelength with flat-top beam profile delivery system, compared to standard-Gaussian profile, has improved photobiomodulatory efficacy on pre-osteoblastic cells differentiation. Mechanistically, the irradiation enhances the pre-osteoblast differentiation through activation of Wnt signaling and activation of Smads 2/3-βcatenin pathway.
Collapse
Affiliation(s)
- Reem Hanna
- Department of Surgical and Diagnostic Sciences, Laser Therapy Centre, University of Genoa, Genoa, Italy
- Department of Oral Surgery, Dental Institute, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Stefano Benedicenti
- Department of Surgical and Diagnostic Sciences, Laser Therapy Centre, University of Genoa, Genoa, Italy
| | - Sara Ferrando
- Laboratory of New Model Organism (NeMo LAB), Department of Earth, Environmental and Life Sciences, University of Genoa, Genoa, Italy
| | - Fulvio Laus
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Vincenzo Cuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Giovanna Lacava
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | | | - Andrea Amaroli
- Department of Surgical and Diagnostic Sciences, Laser Therapy Centre, University of Genoa, Genoa, Italy
- Laboratory of New Model Organism (NeMo LAB), Department of Earth, Environmental and Life Sciences, University of Genoa, Genoa, Italy
| |
Collapse
|
109
|
Fekrazad R, Asefi S, Eslaminejad MB, Taghiar L, Bordbar S, Hamblin MR. Photobiomodulation with single and combination laser wavelengths on bone marrow mesenchymal stem cells: proliferation and differentiation to bone or cartilage. Lasers Med Sci 2019; 34:115-126. [PMID: 30264177 PMCID: PMC6344244 DOI: 10.1007/s10103-018-2620-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/15/2018] [Indexed: 12/21/2022]
Abstract
Tissue engineering aims to take advantage of the ability of undifferentiated stem cells to differentiate into multiple cell types to repair damaged tissue. Photobiomodulation uses either lasers or light-emitting diodes to promote stem cell proliferation and differentiation. The present study aimed to investigate single and dual combinations of laser wavelengths on mesenchymal stem cells (MSCs). MSCs were derived from rabbit iliac bone marrow. One control and eight laser irradiated groups were designated as Infrared (IR, 810 nm), Red (R, 660 nm), Green (G, 532 nm), Blue (B, 485 nm), IR-R, IR-B, R-G, and B-G. Irradiation was repeated daily for 21 days and cell proliferation, osseous, or cartilaginous differentiation was then measured. RT-PCR biomarkers were SOX9, aggrecan, COL 2, and COL 10 expression for cartilage and ALP, COL 1, and osteocalcin expression for bone. Cellular proliferation was increased in all irradiated groups except G. All cartilage markers were significantly increased by IR and IR-B except COL 10 which was suppressed by IR-B combination. ALP expression was highest in R and IR groups during osseous differentiation. ALP was decreased by combinations of IR with B and with R, and also by G alone. R and B-G groups showed stimulated COL 1 expression; however, COL 1 was suppressed in IR-B, IR-R, and G groups. IR significantly increased osteocalcin expression, but in B, B-G, and G groups it was reduced. Cartilage differentiation was stimulated by IR and IR-B laser irradiation. The effects of single or combined laser irradiation were not clear-cut on osseous differentiation. Stimulatory effects on osteogenesis were seen for R and IR lasers, while G laser had inhibitory effects.
Collapse
Affiliation(s)
- Reza Fekrazad
- Periodontics Department, Dentistry School, Laser Research Center in Medical Sciences, AJA University of Medical Sciences, Tehran, Iran.
- International Network for Photo Medicine and Photo Dynamic Therapy (INPMPDT), Universal Scientific Education and ResearchNetwork (USERN), Tehran, Iran.
| | - Sohrab Asefi
- Orthodontic Department, Dentistry School, International Campus of Tehran University of Medical Sciences, Tehran, Iran
| | | | - Leila Taghiar
- Department of Stem Cells and Developmental Biology, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sima Bordbar
- Department of Stem Cells and Developmental Biology, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
110
|
Krassovka J, Borgschulze A, Sahlender B, Lögters T, Windolf J, Grotheer V. Blue light irradiation and its beneficial effect on Dupuytren's fibroblasts. PLoS One 2019; 14:e0209833. [PMID: 30633751 PMCID: PMC6329497 DOI: 10.1371/journal.pone.0209833] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 12/12/2018] [Indexed: 02/06/2023] Open
Abstract
Dupuytren's contracture is a fibroproliferative disorder affecting the palmar fascia of the hand. Most affected are the ring fingers, and little fingers of middle-aged men. Symptomatic for this disease is the increased proliferation and differentiation of fibroblasts to myofibroblasts, which is accompanied by an elevated α-SMA expression. The present study evaluated the therapeutic benefit of blue light (λ = 453 nm, 38 mW/cm2, continuous radiance, spot size 10-12 cm2) as well as the molecular mechanism mediating this effect. It could be determined that blue light significantly diminished the induced α-SMA protein expression in both normal palmar fibroblasts and Duypuytren's fibroblasts. The beneficial effect mediated by this irradiance, radiant exposure and wavelength was associated with an elevated reactive oxygen species generation. Furthermore, the data underlines the potential usefulness of blue light irradiation as a promising therapy option for Dupuytren's disease, especially for relapse prevention, and may represent a useful strategy to treat further fibrotic diseases, such as keloids, hypertrophic scarring, and scleroderma.
Collapse
Affiliation(s)
- Julia Krassovka
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Annika Borgschulze
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Benita Sahlender
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Tim Lögters
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Joachim Windolf
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Vera Grotheer
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
111
|
Mussttaf RA, Jenkins DFL, Jha AN. Assessing the impact of low level laser therapy (LLLT) on biological systems: a review. Int J Radiat Biol 2019; 95:120-143. [DOI: 10.1080/09553002.2019.1524944] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Ruwaidah A. Mussttaf
- School of Computing, Electronics and Mathematics, University of Plymouth, Plymouth, UK
| | - David F. L. Jenkins
- School of Computing, Electronics and Mathematics, University of Plymouth, Plymouth, UK
| | - Awadhesh N. Jha
- School of Biological and Marine Sciences, University of Plymouth, Plymouth, UK
| |
Collapse
|
112
|
|
113
|
Hamblin MR, Huang YY, Heiskanen V. Non-mammalian Hosts and Photobiomodulation: Do All Life-forms Respond to Light? Photochem Photobiol 2019; 95:126-139. [PMID: 29882348 PMCID: PMC6286699 DOI: 10.1111/php.12951] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 06/04/2018] [Indexed: 12/28/2022]
Abstract
Photobiomodulation (PBM), also known as low-level laser (light) therapy, was discovered over 50 years ago, but only recently has it been making progress toward wide acceptance. PBM originally used red and near-infrared (NIR) lasers, but now other wavelengths and non-coherent light-emitting diodes (LEDs) are being explored. The almost complete lack of side effects makes the conduction of controlled clinical trials relatively easy. Laboratory research has mainly concentrated on mammalian cells (normal or cancer) in culture, and small rodents (mice and rats) as models of different diseases. A sizeable body of work was carried out in the 1970s and 1980s in Russia looking at various bacterial and fungal cells. The present review covers some of these studies and a recent number of papers that have applied PBM to so-called "model organisms." These models include flies (Drosophila), worms (Caenorhabditis elegans), fish (zebrafish) and caterpillars (Galleria). Much knowledge about the genomics and proteomics, and many reagents for these organisms already exist. They are inexpensive to work with and have lower regulatory barriers compared to vertebrate animals. Other researchers have studied different models (snails, sea urchins, Paramecium, toads, frogs and chickens). Plants may respond to NIR light differently from visible light (photosynthesis and photomorphogenesis) but PBM in plants has not been much studied. Veterinarians routinely use PBM to treat non-mammalian patients. The conclusion is that red or NIR light does indeed have significant biologic effects conserved over many different kingdoms, and perhaps it is true that "all life-forms respond to light."
Collapse
Affiliation(s)
- Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Ying-Ying Huang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, 02115, USA
| | | |
Collapse
|
114
|
Zein R, Selting W, Hamblin MR. Review of light parameters and photobiomodulation efficacy: dive into complexity. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-17. [PMID: 30550048 PMCID: PMC8355782 DOI: 10.1117/1.jbo.23.12.120901] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 11/14/2018] [Indexed: 05/09/2023]
Abstract
Photobiomodulation (PBM) therapy, previously known as low-level laser therapy, was discovered more than 50 years ago, yet there is still no agreement on the parameters and protocols for its clinical application. Some groups have recommended the use of a power density less than 100 mW/cm2 and an energy density of 4 to 10 J/cm2 at the level of the target tissue. Others recommend as much as 50 J/cm2 at the tissue surface. The wide range of parameters that can be applied (wavelength, energy, fluence, power, irradiance, pulse mode, treatment duration, and repetition) in some cases has led to contradictory results. In our review, we attempt to evaluate the range of effective and ineffective parameters in PBM. Studies in vitro with cultured cells or in vivo with different tissues were divided into those with higher numbers of mitochondria (muscle, brain, heart, nerve) or lower numbers of mitochondria (skin, tendon, cartilage). Graphs were plotted of energy density against power density. Although the results showed a high degree of variability, cells/tissues with high numbers of mitochondria tended to respond to lower doses of light than those with lower number of mitochondria. Ineffective studies in cells with high mitochondrial activity appeared to be more often due to over-dosing than to under-dosing.
Collapse
Affiliation(s)
- Randa Zein
- University of Genoa, Department of Surgical Science and Integrated Diagnostics, Genoa, Italy
| | - Wayne Selting
- University of Genoa, Department of Surgical Science and Integrated Diagnostics, Genoa, Italy
| | - Michael R. Hamblin
- Massachusetts General Hospital, Wellman Center for Photomedicine, Boston, Massachusetts, United States
- Harvard Medical School, Department of Dermatology, Boston, Massachusetts, United States
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, United States
| |
Collapse
|
115
|
Dalesio NM, Barreto Ortiz SF, Pluznick JL, Berkowitz DE. Olfactory, Taste, and Photo Sensory Receptors in Non-sensory Organs: It Just Makes Sense. Front Physiol 2018; 9:1673. [PMID: 30542293 PMCID: PMC6278613 DOI: 10.3389/fphys.2018.01673] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 11/07/2018] [Indexed: 01/28/2023] Open
Abstract
Sensory receptors that detect and respond to light, taste, and smell primarily belong to the G-protein-coupled receptor (GPCR) superfamily. In addition to their established roles in the nose, tongue, and eyes, these sensory GPCRs have been found in many ‘non-sensory' organs where they respond to different physicochemical stimuli, initiating signaling cascades in these extrasensory systems. For example, taste receptors in the airway, and photoreceptors in vascular smooth muscle cells, both cause smooth muscle relaxation when activated. In addition, olfactory receptors are present within the vascular system, where they play roles in angiogenesis as well as in modulating vascular tone. By better understanding the physiological and pathophysiological roles of sensory receptors in non-sensory organs, novel therapeutic agents can be developed targeting these receptors, ultimately leading to treatments for pathological conditions and potential cures for various disease states.
Collapse
Affiliation(s)
- Nicholas M Dalesio
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States.,Department of Otolaryngology/Head & Neck Surgery, Johns Hopkins University, Baltimore, MD, United States
| | - Sebastian F Barreto Ortiz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Jennifer L Pluznick
- Department of Physiology, Johns Hopkins University, Baltimore, MD, United States
| | - Dan E Berkowitz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
116
|
Amaroli A, Ferrando S, Benedicenti S. Photobiomodulation Affects Key Cellular Pathways of all Life-Forms: Considerations on Old and New Laser Light Targets and the Calcium Issue. Photochem Photobiol 2018; 95:455-459. [PMID: 30281800 DOI: 10.1111/php.13032] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/26/2018] [Indexed: 12/22/2022]
Abstract
After 50 years of studies on photobiomodulation (PBM), there is still so much to investigate to understand the laser light-nonplant cells interactions. The current scientific knowledge allows to say that the phenomena induced by PBM are based on cellular pathways that are the key points of cell life. The mitochondria chromophores, also present on the bacterial membrane, the calcium channels, ion that regulates the life-and-death cellular processes, as well as the TRP family, whose genes have been found in protozoa and suggest that its basic mechanism evolved long before the appearance of animals, seem to be elective targets in photobiomodulatory events by wavelengths from 600 up to 980 nm. The ambiguous resulting cellular communication way, mediated by ATP, ROS and/or calcium, leads to cell manipulation, which modifies its metabolism and whose response connects all life-forms from bacteria to vertebrates. Because of the Giano-Bifronte features of ROS and calcium, as well as the fine balance of energetic mitochondrial processes, whose alteration is responsible for several diseases, the PBM can show unpredictable results and it requires scrupulous approach to avoid cellular damages. However, when carefully applied, PBM is able to improve nonhealthy cell's responses and represents a reliable support in human and veterinary medicine.
Collapse
Affiliation(s)
- Andrea Amaroli
- Department of Surgical and Diagnostic Sciences (D.I.S.C), Laser Therapy Center, University of Genoa, Genoa, Italy
| | - Sara Ferrando
- Department of Earth, Environmental and Life Sciences, Laboratory of New Model Organism (NeMo LAB), University of Genoa, Genoa, Italy
| | - Stefano Benedicenti
- Department of Surgical and Diagnostic Sciences (D.I.S.C), Laser Therapy Center, University of Genoa, Genoa, Italy
| |
Collapse
|
117
|
Castellano-Pellicena I, Uzunbajakava NE, Mignon C, Raafs B, Botchkarev VA, Thornton MJ. Does blue light restore human epidermal barrier function via activation of Opsin during cutaneous wound healing? Lasers Surg Med 2018; 51:370-382. [PMID: 30168605 DOI: 10.1002/lsm.23015] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND OBJECTIVE Visible light has beneficial effects on cutaneous wound healing, but the role of potential photoreceptors in human skin is unknown. In addition, inconsistency in the parameters of blue and red light-based therapies for skin conditions makes interpretation difficult. Red light can activate cytochrome c oxidase and has been proposed as a wound healing therapy. UV-blue light can activate Opsin 1-SW, Opsin 2, Opsin 3, Opsin 4, and Opsin 5 receptors, triggering biological responses, but their role in human skin physiology is unclear. MATERIALS AND METHODS Localization of Opsins was analyzed in situ in human skin derived from face and abdomen by immunohistochemistry. An ex vivo human skin wound healing model was established and expression of Opsins confirmed by immunohistochemistry. The rate of wound closure was quantitated after irradiation with blue and red light and mRNA was extracted from the regenerating epithelial tongue by laser micro-dissection to detect changes in Opsin 3 (OPN3) expression. Retention of the expression of Opsins in primary cultures of human epidermal keratinocytes and dermal fibroblasts was confirmed by qRT-PCR and immunocytochemistry. Modulation of metabolic activity by visible light was studied. Furthermore, migration in a scratch-wound assay, DNA synthesis and differentiation of epidermal keratinocytes was established following irradiation with blue light. A role for OPN3 in keratinocytes was investigated by gene silencing. RESULTS Opsin receptors (OPN1-SW, 3 and 5) were similarly localized in the epidermis of human facial and abdominal skin in situ. Corresponding expression was confirmed in the regenerating epithelial tongue of ex vivo wounds after 2 days in culture, and irradiation with blue light stimulated wound closure, with a corresponding increase in OPN3 expression. Expression of Opsins was retained in primary cultures of epidermal keratinocytes and dermal fibroblasts. Both blue and red light stimulated the metabolic activity of cultured keratinocytes. Low levels of blue light reduced DNA synthesis and stimulated differentiation of keratinocytes. While low levels of blue light did not alter keratinocyte migration in a scratch wound assay, higher levels inhibited migration. Gene silencing of OPN3 in keratinocytes was effective (87% reduction). The rate of DNA synthesis in OPN3 knockdown keratinocytes did not change following irradiation with blue light, however, the level of differentiation was decreased. CONCLUSIONS Opsins are expressed in the epidermis and dermis of human skin and in the newly regenerating epidermis following wounding. An increase in OPN3 expression in the epithelial tongue may be a potential mechanism for the stimulation of wound closure by blue light. Since keratinocytes and fibroblasts retain their expression of Opsins in culture, they provide a good model to investigate the mechanism of blue light in wound healing responses. Knockdown of OPN3 led to a reduction in early differentiation of keratinocytes following irradiation with blue light, suggesting OPN3 is required for restoration of the barrier function. Understanding the function and relationship of different photoreceptors and their response to specific light parameters will lead to the development of reliable light-based therapies for cutaneous wound healing. Lasers Surg. Med. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Irene Castellano-Pellicena
- Centre for Skin Sciences, Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom.,Philips Research, High Tech Campus, Eindhoven, The Netherlands
| | | | - Charles Mignon
- Centre for Skin Sciences, Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom.,Philips Research, High Tech Campus, Eindhoven, The Netherlands
| | - Bianca Raafs
- Philips Research, High Tech Campus, Eindhoven, The Netherlands
| | - Vladimir A Botchkarev
- Centre for Skin Sciences, Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
| | - M Julie Thornton
- Centre for Skin Sciences, Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
| |
Collapse
|
118
|
Photobiomodulation with 808-nm diode laser light promotes wound healing of human endothelial cells through increased reactive oxygen species production stimulating mitochondrial oxidative phosphorylation. Lasers Med Sci 2018; 34:495-504. [DOI: 10.1007/s10103-018-2623-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022]
|
119
|
Abstract
Photobiomodulation (PBM) is a treatment method based on research findings showing that irradiation with certain wavelengths of red or near-infrared light has been shown to produce a range of physiological effects in cells, tissues, animals and humans. Scientific research into PBM was initially started in the late 1960s by utilizing the newly invented (1960) lasers, and the therapy rapidly became known as "low-level laser therapy". It was mainly used for wound healing and reduction of pain and inflammation. Despite other light sources being available during the first 40 years of PBM research, lasers remained by far the most commonly employed device, and in fact, some authors insisted that lasers were essential to the therapeutic benefit. Collimated, coherent, highly monochromatic beams with the possibility of high power densities were considered preferable. However in recent years, non-coherent light sources such as light-emitting diodes (LEDs) and broad-band lamps have become common. Advantages of LEDs include no laser safety considerations, ease of home use, ability to irradiate a large area of tissue at once, possibility of wearable devices, and much lower cost per mW. LED photobiomodulation is here to stay.
Collapse
Affiliation(s)
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA and Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA and Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
120
|
Heiskanen V, Hamblin MR. Photobiomodulation: lasers vs. light emitting diodes? Photochem Photobiol Sci 2018; 17:1003-1017. [PMID: 30044464 PMCID: PMC6091542 DOI: 10.1039/c8pp90049c] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Photobiomodulation (PBM) is a treatment method based on research findings showing that irradiation with certain wavelengths of red or near-infrared light has been shown to produce a range of physiological effects in cells, tissues, animals and humans. Scientific research into PBM was initially started in the late 1960s by utilizing the newly invented (1960) lasers, and the therapy rapidly became known as "low-level laser therapy". It was mainly used for wound healing and reduction of pain and inflammation. Despite other light sources being available during the first 40 years of PBM research, lasers remained by far the most commonly employed device, and in fact, some authors insisted that lasers were essential to the therapeutic benefit. Collimated, coherent, highly monochromatic beams with the possibility of high power densities were considered preferable. However in recent years, non-coherent light sources such as light-emitting diodes (LEDs) and broad-band lamps have become common. Advantages of LEDs include no laser safety considerations, ease of home use, ability to irradiate a large area of tissue at once, possibility of wearable devices, and much lower cost per mW. LED photobiomodulation is here to stay.
Collapse
Affiliation(s)
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA and Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA and Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
121
|
Photobiomodulation of extracellular matrix enzymes in human nucleus pulposus cells as a potential treatment for intervertebral disk degeneration. Sci Rep 2018; 8:11654. [PMID: 30076336 PMCID: PMC6076240 DOI: 10.1038/s41598-018-30185-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 07/24/2018] [Indexed: 12/18/2022] Open
Abstract
Intervertebral disc (IVD) degeneration is associated with imbalances between catabolic and anabolic responses, regulated by extracellular matrix (ECM)-modifying enzymes such as matrix metalloproteinases (MMPs) and their endogenous tissue inhibitors of metalloproteinases (TIMPs). Potential contributing factors, such as interleukin (IL)-1β and tumor necrosis factor (TNF)-α, derived from infiltrated, activated macrophages within IVD tissues, can trigger abnormal production of ECM-modifying enzymes and progression of IVD degeneration. Novel therapies for regulating ECM-modifying enzymes can prevent or ameliorate IVD degeneration. Photobiomodulation (PBM), known to regulate wound repair, exhibits regenerative potential by modulating biological molecules. This study examined the effects of PBM, administered at various wavelengths (630, 525, and 465 nm) and energy densities (16, 32, and 64 J/cm2), on the production of ECM-modifying enzymes in replicated degenerative IVD. Our results showed that PBM selectively inhibited the production of ECM-modifying enzymes in a dose- and wavelength-dependent manner, suggesting that it could be a novel tool for treating symptomatic IVD degeneration.
Collapse
|
122
|
Odinokov D, Hamblin MR. Aging of lymphoid organs: Can photobiomodulation reverse age-associated thymic involution via stimulation of extrapineal melatonin synthesis and bone marrow stem cells? JOURNAL OF BIOPHOTONICS 2018; 11:e201700282. [PMID: 29227581 PMCID: PMC5995606 DOI: 10.1002/jbio.201700282] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 12/07/2017] [Indexed: 05/27/2023]
Abstract
Thymic atrophy and the subsequent reduction in T-cell production are the most noticeable age-related changes affecting lymphoid organs in the immune system. In fact, thymic involution has been described as "programmed aging." New therapeutic approaches, such as photobiomodulation (PBM), may reduce or reverse these changes. PBM (also known as low-level laser therapy) involves the delivery of non-thermal levels of red or near-infrared light that are absorbed by mitochondrial chromophores, in order to prevent tissue death and stimulate healing and regeneration. PBM may reverse or prevent thymic involution due to its ability to induce extrapineal melatonin biosynthesis via cyclic adenosine monophosphate (AMP) or NF-kB activation, or alternatively by stimulating bone marrow stem cells that can regenerate the thymus. This perspective puts forward a hypothesis that PBM can alter thymic involution, improve immune functioning in aged people and even extend lifespan.
Collapse
Affiliation(s)
- Denis Odinokov
- Department of Biomedical Engineering, Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Dermatology, Harvard Medical School, Boston, Massachusetts
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts
| |
Collapse
|
123
|
Tani A, Chellini F, Giannelli M, Nosi D, Zecchi-Orlandini S, Sassoli C. Red (635 nm), Near-Infrared (808 nm) and Violet-Blue (405 nm) Photobiomodulation Potentiality on Human Osteoblasts and Mesenchymal Stromal Cells: A Morphological and Molecular In Vitro Study. Int J Mol Sci 2018; 19:ijms19071946. [PMID: 29970828 PMCID: PMC6073131 DOI: 10.3390/ijms19071946] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/15/2018] [Accepted: 06/29/2018] [Indexed: 01/08/2023] Open
Abstract
Photobiomodulation (PBM) has been used for bone regenerative purposes in different fields of medicine and dentistry, but contradictory results demand a skeptical look for its potential benefits. This in vitro study compared PBM potentiality by red (635 ± 5 nm) or near-infrared (NIR, 808 ± 10 nm) diode lasers and violet-blue (405 ± 5 nm) light-emitting diode operating in a continuous wave with a 0.4 J/cm2 energy density, on human osteoblast and mesenchymal stromal cell (hMSC) viability, proliferation, adhesion and osteogenic differentiation. PBM treatments did not alter viability (PI/Syto16 and MTS assays). Confocal immunofluorescence and RT-PCR analyses indicated that red PBM (i) on both cell types increased vinculin-rich clusters, osteogenic markers expression (Runx-2, alkaline phosphatase, osteopontin) and mineralized bone-like nodule structure deposition and (ii) on hMSCs induced stress fiber formation and upregulated the expression of proliferation marker Ki67. Interestingly, osteoblast responses to red light were mediated by Akt signaling activation, which seems to positively modulate reactive oxygen species levels. Violet-blue light-irradiated cells behaved essentially as untreated ones and NIR irradiated ones displayed modifications of cytoskeleton assembly, Runx-2 expression and mineralization pattern. Although within the limitations of an in vitro experimentation, this study may suggest PBM with 635 nm laser as potential effective option for promoting/improving bone regeneration.
Collapse
Affiliation(s)
- Alessia Tani
- Department of Experimental and Clinical Medicine-Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| | - Flaminia Chellini
- Department of Experimental and Clinical Medicine-Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| | - Marco Giannelli
- Odontostomatologic Laser Therapy Center, via dell' Olivuzzo 162, 50143 Florence, Italy.
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine-Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| | - Sandra Zecchi-Orlandini
- Department of Experimental and Clinical Medicine-Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| | - Chiara Sassoli
- Department of Experimental and Clinical Medicine-Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| |
Collapse
|
124
|
Liao X, Li SH, Xie GH, Xie S, Xiao LL, Song JX, Liu HW. Preconditioning With Low-Level Laser Irradiation Enhances the Therapeutic Potential of Human Adipose-derived Stem Cells in a Mouse Model of Photoaged Skin. Photochem Photobiol 2018; 94:780-790. [PMID: 29457847 DOI: 10.1111/php.12912] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 02/09/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Xuan Liao
- Department of Plastic Surgery; The First Affiliated Hospital of Jinan University; Innovative Technology Research Institute of Tissue Repair and Regeneration; Key Laboratory of Regenerative Medicine; Ministry of Education; Guangzhou Guangdong China
| | - Sheng-Hong Li
- Department of Plastic Surgery; The First Affiliated Hospital of Jinan University; Innovative Technology Research Institute of Tissue Repair and Regeneration; Key Laboratory of Regenerative Medicine; Ministry of Education; Guangzhou Guangdong China
| | - Guang-Hui Xie
- Department of Plastic Surgery; The First Affiliated Hospital of Jinan University; Innovative Technology Research Institute of Tissue Repair and Regeneration; Key Laboratory of Regenerative Medicine; Ministry of Education; Guangzhou Guangdong China
| | - Shan Xie
- Department of Plastic Surgery; The First Affiliated Hospital of Jinan University; Innovative Technology Research Institute of Tissue Repair and Regeneration; Key Laboratory of Regenerative Medicine; Ministry of Education; Guangzhou Guangdong China
| | - Li-Ling Xiao
- Department of Plastic Surgery; The First Affiliated Hospital of Jinan University; Innovative Technology Research Institute of Tissue Repair and Regeneration; Key Laboratory of Regenerative Medicine; Ministry of Education; Guangzhou Guangdong China
| | - Jian-Xing Song
- Department of Plastic Surgery; Changhai Hospital; The Second Military Medical University; Shanghai China
| | - Hong-Wei Liu
- Department of Plastic Surgery; The First Affiliated Hospital of Jinan University; Innovative Technology Research Institute of Tissue Repair and Regeneration; Key Laboratory of Regenerative Medicine; Ministry of Education; Guangzhou Guangdong China
| |
Collapse
|
125
|
Mignon C, Uzunbajakava NE, Castellano-Pellicena I, Botchkareva NV, Tobin DJ. Differential response of human dermal fibroblast subpopulations to visible and near-infrared light: Potential of photobiomodulation for addressing cutaneous conditions. Lasers Surg Med 2018; 50:859-882. [PMID: 29665018 DOI: 10.1002/lsm.22823] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND OBJECTIVES The past decade has witnessed a rapid expansion of photobiomodulation (PBM), demonstrating encouraging results for the treatment of cutaneous disorders. Confidence in this approach, however, is impaired not only by a lack of understanding of the light-triggered molecular cascades but also by the significant inconsistency in published experimental outcomes, design of the studies and applied optical parameters. This study aimed at characterizing the response of human dermal fibroblast subpopulations to visible and near-infrared (NIR) light in an attempt to identify the optical treatment parameters with high potential to address deficits in aging skin and non-healing chronic wounds. MATERIALS AND METHODS Primary human reticular and papillary dermal fibroblasts (DF) were isolated from the surplus of post-surgery human facial skin. An in-house developed LED-based device was used to irradiate cell cultures using six discrete wavelengths (450, 490, 550, 590, 650, and 850 nm). Light dose-response at a standard oxygen concentration (20%) at all six wavelengths was evaluated in terms of cell metabolic activity. This was followed by an analysis of the transcriptome and procollagen I production at a protein level, where cells were cultured in conditions closer to in vivo at 2% environmental oxygen and 2% serum. Furthermore, the production of reactive oxygen species (ROS) was accessed using real-time fluorescence confocal microscopy imaging. Here, production of ROS in the presence or absence of antioxidants, as well as the cellular localization of ROS, was evaluated. RESULTS In terms of metabolic activity, consecutive irradiation with short-wavelength light (⇐530 nm) exerted an inhibitory effect on DF, while longer wavelengths (>=590 nm) had essentially a neutral effect. Cell behavior following treatment with 450 nm was biphasic with two distinct states: inhibitory at low- to mid- dose levels (<=30 J/cm2 ), and cytotoxic at higher dose levels (>30 J/cm2 ). Cell response to blue light was accompanied by a dose-dependent release of ROS that was localized in the perinuclear area close to mitochondria, which was attenuated by an antioxidant. Overall, reticular DFs exhibited a greater sensitivity to light treatment at the level of gene expression than did papillary DFs, with more genes significantly up- or down- regulated. At the intra-cellular signaling pathway level, the up- or down- regulation of vital pathways was observed only for reticular DF, after treatment with 30 J/cm2 of blue light. At the cellular level, short visible wavelengths exerted a greater inhibitory effect on reticular DF. Several genes involved in the TGF-β signaling pathway were also affected. In addition, procollagen I production was inhibited. By contrast, 850 nm near-infrared (NIR) light (20 J/cm2 ) exerted a stimulatory metabolic effect in these cells, with no detectable intracellular ROS formation. Here too, reticular DF were more responsive than papillary DF. This stimulatory effect was only observed under in vivo-like low oxygen conditions, corresponding to normal dermal tissue oxygen levels (approximately 2%). CONCLUSION This study highlights a differential impact of light on human skin cells with upregulation of metabolic activity with NIR light, and inhibition of pro-collagen production and proliferation in response to blue light. These findings open-up new avenues for developing therapies for different cutaneous conditions (e.g., treatment of keloids and fibrosis) or differential therapy at distinct stages of wound healing. Lasers Surg. Med. 50:859-882, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Charles Mignon
- Centre for Skin Sciences, University of Bradford, BD71DP, Bradford, United-Kingdom.,Philips Research, High Tech Campus, Eindhoven, Netherlands
| | | | - Irene Castellano-Pellicena
- Centre for Skin Sciences, University of Bradford, BD71DP, Bradford, United-Kingdom.,Philips Research, High Tech Campus, Eindhoven, Netherlands
| | | | - Desmond J Tobin
- Centre for Skin Sciences, University of Bradford, BD71DP, Bradford, United-Kingdom
| |
Collapse
|
126
|
Abstract
Photobiomodulation or low-level light therapy (LLLT) has extensive applications based on light-induced effects in biological systems. Photobiomodulation remains controversial because of a poorly understood biochemical mechanism limited by the well-known biphasic dose response or Arndt-Schulz curve. The Arndt-Schulz curve states that an optimal dose of light is a key factor for realizing a therapeutic effect. In this report, we demonstrate a tunable optical system for photobiomodulation to aid physicians in overcoming the constraints of light due to biphasic dose response. The tunable optical system is based on a white light-emitting diode and four liquid crystal (LC) photonic devices: three LC phase retarders, and one LC lens. The output light of the tunable optical system exhibits electrical tunability for the wavelength, energy density and beam size. The operating principle is introduced, and the experimental results are presented. The proposed concept can be further extended to other electrically tunable photonic devices for different clinical purposes for photobiomodulation.
Collapse
|
127
|
Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochem Photobiol 2018; 94:199-212. [PMID: 29164625 PMCID: PMC5844808 DOI: 10.1111/php.12864] [Citation(s) in RCA: 347] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 10/31/2017] [Indexed: 12/23/2022]
Abstract
Photobiomodulation (PBM) involves the use of red or near-infrared light at low power densities to produce a beneficial effect on cells or tissues. PBM therapy is used to reduce pain, inflammation, edema, and to regenerate damaged tissues such as wounds, bones, and tendons. The primary site of light absorption in mammalian cells has been identified as the mitochondria and, more specifically, cytochrome c oxidase (CCO). It is hypothesized that inhibitory nitric oxide can be dissociated from CCO, thus restoring electron transport and increasing mitochondrial membrane potential. Another mechanism involves activation of light or heat-gated ion channels. This review will cover the redox signaling that occurs in PBM and examine the difference between healthy and stressed cells, where PBM can have apparently opposite effects. PBM has a marked effect on stem cells, and this is proposed to operate via mitochondrial redox signaling. PBM can act as a preconditioning regimen and can interact with exercise on muscles.
Collapse
Affiliation(s)
- Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA
- Department of Dermatology, Harvard Medical School, Boston, MA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA
| |
Collapse
|
128
|
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
129
|
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
130
|
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
131
|
Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochem Photobiol 2018. [DOI: 10.1111/php.12864 and make_set(2234=2234,4853)-- tppa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
132
|
Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochem Photobiol 2018. [DOI: 10.1111/php.12864 or updatexml(4295,concat(0x2e,0x717a717671,(select (elt(4295=4295,1))),0x71706a6271),3985)-- bssu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
133
|
Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochem Photobiol 2018. [DOI: 10.1111/php.12864 or not 3194=3194# dgnj] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
134
|
Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochem Photobiol 2018. [DOI: 10.1111/php.12864 and (select (case when (5719=8223) then null else ctxsys.drithsx.sn(1,5719) end) from dual) is null] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
135
|
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
136
|
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
137
|
Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochem Photobiol 2018. [DOI: 10.1111/php.12864 and 8885=3318-- bykq] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
138
|
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
139
|
Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochem Photobiol 2018. [DOI: 10.1111/php.12864 or not 8779=2113# mdth] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
140
|
Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochem Photobiol 2018. [DOI: 10.1111/php.12864 or not 5169=2257-- ejdi] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
141
|
Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochem Photobiol 2018. [DOI: 10.1111/php.12864 and 2019=2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
142
|
Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochem Photobiol 2018. [DOI: 10.1111/php.12864 and 1705=('qzqvq'||(select case 1705 when 1705 then 1 else 0 end from rdb$database)||'qpjbq')-- qsrj] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
143
|
Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochem Photobiol 2018. [DOI: 10.1111/php.12864 and extractvalue(6022,concat(0x5c,0x717a717671,(select (elt(6022=6022,1))),0x71706a6271))# igpm] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
144
|
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
145
|
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
146
|
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
147
|
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
148
|
Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochem Photobiol 2018. [DOI: 10.1111/php.12864 and 2341=9012# mbxq] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
149
|
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|
150
|
Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochem Photobiol 2018. [DOI: 10.1111/php.12864 or not 9689=3416#] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Michael R. Hamblin
- Wellman Center for Photomedicine Massachusetts General Hospital Boston MA
- Department of Dermatology Harvard Medical School Boston MA
- Harvard‐MIT Division of Health Sciences and Technology Cambridge MA
| |
Collapse
|