1
|
da Silva TG, Ribeiro RS, Mencalha AL, de Souza Fonseca A. Photobiomodulation at molecular, cellular, and systemic levels. Lasers Med Sci 2023; 38:136. [PMID: 37310556 DOI: 10.1007/s10103-023-03801-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/05/2023] [Indexed: 06/14/2023]
Abstract
Since the reporting of Endre Mester's results, researchers have investigated the biological effects induced by non-ionizing radiation emitted from low-power lasers. Recently, owing to the use of light-emitting diodes (LEDs), the term photobiomodulation (PBM) has been used. However, the molecular, cellular, and systemic effects involved in PBM are still under investigation, and a better understanding of these effects could improve clinical safety and efficacy. Our aim was to review the molecular, cellular, and systemic effects involved in PBM to elucidate the levels of biological complexity. PBM occurs as a consequence of photon-photoacceptor interactions, which lead to the production of trigger molecules capable of inducing signaling, effector molecules, and transcription factors, which feature it at the molecular level. These molecules and factors are responsible for cellular effects, such as cell proliferation, migration, differentiation, and apoptosis, which feature PBM at the cellular level. Finally, molecular and cellular effects are responsible for systemic effects, such as modulation of the inflammatory process, promotion of tissue repair and wound healing, reduction of edema and pain, and improvement of muscle performance, which features PBM at the systemic level.
Collapse
Affiliation(s)
- Thayssa Gomes da Silva
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Vila Isabel, Boulevard 28 de Setembro, 87, Rio de Janeiro, 20551030, Brazil.
| | - Rickson Souza Ribeiro
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Vila Isabel, Boulevard 28 de Setembro, 87, Rio de Janeiro, 20551030, Brazil
| | - Andre Luiz Mencalha
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Vila Isabel, Boulevard 28 de Setembro, 87, Rio de Janeiro, 20551030, Brazil
| | - Adenilson de Souza Fonseca
- Departamento de Biofísica e Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Vila Isabel, Boulevard 28 de Setembro, 87, Rio de Janeiro, 20551030, Brazil
- Departamento de Ciências Fisiológicas, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rua Frei Caneca, 94, Rio de Janeiro, 20211040, Brazil
| |
Collapse
|
2
|
Ahmadi F, Dalirsani Z, Tayarani-Najaran Z, Ebrahimzadeh-Bideskan A, Shafieian R. A Comparative Analysis of Photobiomodulation-Mediated Biological Effects of Single Versus Double Irradiation on Dental Pulp Stem Cells: An In Vitro Study. Photobiomodul Photomed Laser Surg 2022; 40:334-342. [PMID: 35559714 DOI: 10.1089/photob.2021.0134] [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] Open
Abstract
Objective: In recent years, fractionated irradiation protocols, rather than a simple plan of exposure, have been proposed as a more effective method in the field of tissue regeneration. Thus, this study aimed at a comparative analysis of single versus double irradiation of an 808-nm diode laser, in terms of dental pulp stem cells' (DPSCs) viability and proliferation in vitro. Methods: Subcultured DPSCs were either irradiated, or not (control group), with energy densities of 3, 7, and 12 J·cm-2 in a single- or double-session manner (24 h apart). On 0, 12, 24, 48, and 72 h postirradiation, cell viability and proliferation were evaluated through Trypan Blue and alamarBlue assays, respectively. Results: During the first 48 h postirradiation, the highest rates of DPSC proliferation were assigned to double irradiation at 3 or single exposure to 7 J⋅cm-2, with no cytotoxic effects on cell viability. Inversely, single irradiation at 12, or a double session of exposure to 7 or 12 J⋅cm-2, led to a significant descent in the rates of proliferation and cell viability. Conclusions: Within the limitations of this study, evidence suggests a positive impact on the biological responses of DPSCs following double session of exposure to lower energy densities as well as a single irradiation at a higher energy dosage.
Collapse
Affiliation(s)
- Farahnaz Ahmadi
- Pharmaceutical Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zohreh Dalirsani
- Oral and Maxillofacial Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Tayarani-Najaran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Reyhaneh Shafieian
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Stem Cells and Regenerative Medicine Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
The Effects of Photobiomodulation on Bone Defect Repairing in a Diabetic Rat Model. Int J Mol Sci 2021; 22:ijms222011026. [PMID: 34681687 PMCID: PMC8541159 DOI: 10.3390/ijms222011026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/09/2021] [Accepted: 10/10/2021] [Indexed: 11/17/2022] Open
Abstract
The purpose of this study is to examine the prospective therapeutic effects of photobiomodulation on the healing of bone defects in diabetic mellitus (DM) using rat models to provide basic knowledge of photobiomodulation therapy (PBMT) during bone defect repair. For in vitro study, an Alizzarin red stain assay was used to evaluate the effect of PBMT on osteogenic differentiation. For in vivo study, micro-computed tomography (microCT) scan, H&E and IHC stain analysis were used to investigate the effect of PBMT on the healing of the experimental calvarial defect (3 mm in diameter) of a diabetic rat model. For in vitro study, the high glucose groups showed lower osteogenic differentiation in both irradiated and non-irradiated with PBMT when compared to the control groups. With the PBMT, all groups (control, osmotic control and high glucose) showed higher osteogenic differentiation when compared to the non-irradiated groups. For in vivo study, the hyperglycemic group showed significantly lower bone regeneration when compared to the control group. With the PBMT, the volume of bone regeneration was increasing and back to the similar level of the control group. The treatment of PBMT in 660 nm could improve the bone defect healing on a diabetic rat calvarial defect model.
Collapse
|
4
|
Souza C, Jayme CC, Rezende N, Tedesco AC. Synergistic effect of photobiomodulation and phthalocyanine photosensitizer on fibroblast signaling responses in an in vitro three-dimensional microenvironment. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 222:112256. [PMID: 34330080 DOI: 10.1016/j.jphotobiol.2021.112256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/12/2021] [Accepted: 07/04/2021] [Indexed: 12/22/2022]
Abstract
Photobiomodulation (PBM) is a promising medical treatment modality in the area of photodynamic therapy (PDT). In this study, we investigated the effect of combined therapy in a 3D microenvironment using aluminum chloride phthalocyanines (AlClPc) as the photosensitizing agent. Normal human fibroblast-containing collagen biomatrix was prepared and treated with an oil-in-water (o/a) AlClPc-loaded nanoemulsion (from 0.5 to 3.0 μM) and irradiated at a range of fluences (from 0.1 to 3.0 J/cm2) using a continuous-wave light-emitting diode (LED) irradiation system (660 nm). PBM at 1.2 J/cm2 and AlClPc/NE at 0.5 μM modified the fibroblast signaling response under 3D conditions, promoting collagen synthesis, ROS production, MMP-9 secretion, proliferation of the actin network, and facile myofibroblastic differentiation. PBM alone (at 1.2 J/cm2 and 0.3 J/cm2) had no significant effect on any of these parameters. The combined therapy affected myofibroblastic differentiation, inflammatory response, and extracellular matrix pliability, and should thus be examined further in subsequent studies considering that no side effects of PBM have been reported. Even though significant progress has been made in the field of phototherapy in recent years, it is necessary to further elucidate the detailed mechanisms underlying its effects already shown in 2D conditions to increase the acceptance of this beneficial and non-invasive therapeutic approach.
Collapse
Affiliation(s)
- Carla Souza
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering -Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto-FFCLRP, University of São Paulo, Ribeirão Preto, São Paulo 14040-901, Brazil
| | - Cristiano Ceron Jayme
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering -Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto-FFCLRP, University of São Paulo, Ribeirão Preto, São Paulo 14040-901, Brazil
| | - Nayara Rezende
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering -Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto-FFCLRP, University of São Paulo, Ribeirão Preto, São Paulo 14040-901, Brazil
| | - Antonio Claudio Tedesco
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering -Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto-FFCLRP, University of São Paulo, Ribeirão Preto, São Paulo 14040-901, Brazil.
| |
Collapse
|
5
|
Chaudary S, Karner L, Weidinger A, Meixner B, Rieger S, Metzger M, Zipperle J, Dungel P. In vitro effects of 635 nm photobiomodulation under hypoxia/reoxygenation culture conditions. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 209:111935. [PMID: 32622295 DOI: 10.1016/j.jphotobiol.2020.111935] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/15/2020] [Accepted: 06/10/2020] [Indexed: 12/18/2022]
Abstract
Photobiomodulation (PBM), especially in the red wavelength range, has been demonstrated to be an effective treatment option for superficial and chronic wounds. However, ischemia and subsequent reperfusion can further challenge wound healing. Therefore, we investigated the effect of pulsed red LED light at 635 nm on cellular function in an in-vitro model of hypoxia/reoxygenation (H/R) challenge. Mouse myoblasts and fibroblasts were incubated in oxygen-deprived starvation medium (hypoxia) for 3 h after which the media was changed to oxygenated, fully supplemented media to simulate reperfusion. Cells were then treated with pulsed red LED light at a wavelength of 635 nm at 40 mW/cm2. Mitochondrial respiratory activity, ATP production and ROS levels were analysed immediately post-illumination. The effects on cellular metabolic activity and proliferation were measured at 6 h and 24 h and apoptosis/necrosis was measured at 24 h post-illumination. Our results show that both cell types reacted differently to H/R challenge and PBM. PBM of H/R-challenged cells enhanced mitochondrial activity and rescued decreased ATP levels, with significant effects in fibroblasts. This was associated with increased cell proliferation rates in both cell types. The increase was again more pronounced in fibroblasts. Our study concluded that PBM with red LED light significantly restored ATP levels during H/R and effectively promoted cell growth under both normoxic and H/R conditions. In clinical applications, PBM has been repeatedly reported to resolve difficult clinical situations in which ischemia/reperfusion injuries are a major issue. Our study confirms the beneficial effects of PBM especially in H/R-challenged cells.
Collapse
Affiliation(s)
- Sidrah Chaudary
- Ludwig Boltzmann institute for experimental and clinical traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Lisa Karner
- Ludwig Boltzmann institute for experimental and clinical traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Adelheid Weidinger
- Ludwig Boltzmann institute for experimental and clinical traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Barbara Meixner
- Ludwig Boltzmann institute for experimental and clinical traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Stefan Rieger
- Ludwig Boltzmann institute for experimental and clinical traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Magdalena Metzger
- Ludwig Boltzmann institute for experimental and clinical traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Johannes Zipperle
- Ludwig Boltzmann institute for experimental and clinical traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Peter Dungel
- Ludwig Boltzmann institute for experimental and clinical traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria.
| |
Collapse
|
6
|
Saghaei Bagheri H, Rasta SH, Mohammadi SM, Rahimi AAR, Movassaghpour A, Nozad Charoudeh H. Low-Level Laser Irradiation Modulated Viability of Normal and Tumor Human Lymphocytes In Vitro. J Lasers Med Sci 2020; 11:174-180. [PMID: 32273959 DOI: 10.34172/jlms.2020.29] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Introduction: Laser radiation is a promising strategy against various malignancies. Recent studies have shown that the application of low-power laser therapy (LPLT) at different doses and exposure times could modulate the growth dynamic of tumor cells. Based on the type of laser, LPLT could potentially trigger cell proliferation, differentiation, and apoptosis in different cell lines. Methods: In this study, MTT assay was used to monitor the effect of low and high laser intensities on the viability of normal and cancer lymphocytes. The protein levels of Ki-67 (a proliferation marker) and Caspase-3 (an apoptosis factor) were measured in human peripheral mononuclear cells (PBMCs) and the B-lymphoblastic cell line (Nalm-6) using flow cytometry after being-exposed to 630-nm LPLT at low (2, 4, 6, and 10 J/cm2 ) and high (15, 30, 60, and 120 J/cm2) energy densities in a continuous mode for 48 and 72 hours. Results: By using higher energy densities, 60 and 120 J/cm2 , a significant decrease was shown in the viability of Nalm-6 cells, which reached 6.6 and 10.1% after 48 hours compared to the control cells (P<0.05). Notably, Cell exposure to doses 30, 60, and 120 J/cm2 yielded 7.5, 12.9, and 21.6 cell viability reduction after 72 hours. The collected data showed that the high-intensity parameters of LPLT (15 to 120 J/cm2) promoted significant apoptotic changes in the exposed cells coincided with the activation of Caspase-3 compared to the none-treated control cells (P<0.05). The data further showed the stimulation of the Ki-67 factor both in primary PBMCs and the lymphoblastic cell line treated with LPLT at energy densities of 4 and 6 J/cm2 (P<0.05), indicating enhanced cell proliferation. Similar to Nalm-6 cells, primary PBMCs showed apoptosis after 48 hours of being exposed to doses 60, and 120 J/cm2 , indicated by increased Caspase-3 levels (P<0.05). As expected, the Nalm-6 cells were resistant to cytotoxic effects of laser irradiation in the first 48 hours (P>0.05) compared to normal PBMCs. The exposure of Nalm-6 cells to low-intensity laser intensities increased a proliferation rate compared to the PBMCs treated with the same doses. Conclusion: We showed the potency of LPLT in the induction of apoptosis and proliferation in human primary PBMCs and Nalm-6 cells in a dose and time-dependent manner after 72 hours.
Collapse
Affiliation(s)
- Hesam Saghaei Bagheri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Physics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran 3
| | - Seyed Hossein Rasta
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Physics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran 3.,Department of Medical Bioengineering, Tabriz University of Medical Sciences, Tabriz, Iran.,School of Biomedical Sciences, University of Aberdeen, Aberdeen, UK
| | | | - Ali Akbar Rahim Rahimi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | |
Collapse
|
7
|
Wang YH, Wu JY, Kong SC, Chiang MH, Ho ML, Yeh ML, Chen CH. Low power laser irradiation and human adipose-derived stem cell treatments promote bone regeneration in critical-sized calvarial defects in rats. PLoS One 2018; 13:e0195337. [PMID: 29621288 PMCID: PMC5886537 DOI: 10.1371/journal.pone.0195337] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/20/2018] [Indexed: 01/18/2023] Open
Abstract
Both stem cell therapy and physical treatments have been shown to be beneficial in accelerating bone healing. However, the efficacy of combined treatment with stem cells and physical stimuli for large bone defects remains uncertain. The aim of this study was to evaluate the bone regeneration effects of low-power laser irradiation (LPLI) and human adipose-derived stem cell (ADSC) treatments during fracture repair using a comparative rat calvarial defect model. We evaluated the viability of human ADSCs, which were cultured on a porous PLGA scaffold using an MTS assay. The critical-sized calvarial bone defect rats were divided into 4 groups: control group, LPLI group, ADSC group, and ADSC+LPLI group. Bone formation was evaluated using micro-CT. New bone formation areas and osteogenic factor expression levels were then examined by histomorphological analysis and immunohistochemical staining. Our data showed that PLGA had no cytotoxic effect on human ADSCs. Micro-CT analyses revealed that both the LPLI and ADSC groups showed improved calvarial bone defect healing compared to the control group. In addition, the ADSC+LPLI group showed significantly increased bone volume at 16 weeks after surgery. The area of new bone formation ranked as follows: control group < LPLI group < ADSC group < ADSC+LPLI group. There were significant differences between the groups. In addition, both ADSC and ADSC+LPLI groups showed strong signals of vWF expression. ADSC and LPLI treatments improved fracture repair in critical-sized calvarial defects in rats. Importantly, the combined treatment of ADSCs and LPLI further enhances the bone healing process.
Collapse
Affiliation(s)
- Yan-Hsiung Wang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jyun-Yi Wu
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Fresenius Kabi Taiwan Ltd, Taipei, Taiwan
| | - Su Chii Kong
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Min-Hsuan Chiang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Ling Ho
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ming-Long Yeh
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Hsin Chen
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail:
| |
Collapse
|
8
|
Abstract
Despite diverse methods being applied to induce wound healing, many wounds remain recalcitrant to all treatments. Photobiomodulation involves inducing wound healing by illuminating wounds with light emitting diodes or lasers. While used on different animal models, in vitro, and clinically, wound healing is induced by many different wavelengths and powers with no optimal set of parameters yet being identified. While data suggest that simultaneous multiple wavelength illumination is more efficacious than single wavelengths, the optimal single and multiple wavelengths must be better defined to induce more reliable and extensive healing of different wound types. This review focuses on studies in which specific wavelengths induce wound healing and on their mechanisms of action.
Collapse
Affiliation(s)
- Damien P Kuffler
- Institute of Neurobiology, University of Puerto Rico, 201 Calle Norzagaray, San Juan 00901, Puerto Rico
| |
Collapse
|
9
|
Liang WZ, Liu PF, Fu E, Chung HS, Jan CR, Wu CH, Shu CW, Hsieh YD. Selective cytotoxic effects of low-power laser irradiation on human oral cancer cells. Lasers Surg Med 2015; 47:756-64. [PMID: 26395333 DOI: 10.1002/lsm.22419] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2015] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND OBJECTIVES Low-power laser irradiation (LPLI) is known to regulate cell proliferation and migration in clinical use. Recent studies have shown that LPLI induces cell death in some certain types of cancer cell lines. However, the cytotoxic selectivity of LPLI for cancer cells is not fully understood. The aim of this study was to compare the cytotoxic effects of LPLI in both human oral cancer OC2 cells and normal human gingival fibroblast (HGF) cells. MATERIALS AND METHODS LPLI at 810 nm with an energy density from 10 to 60 J/cm(2) was used to irradiate human oral cancer OC2 cells and normal HGF cells. RESULTS We found that LPLI significantly diminished cell viability of human oral cancer OC2 cells due to cell cycle arrest at the G1 phase and the induction of cell death but that it had no or little effects on cell cycle progression and death in normal HGF cells. Moreover, the production of reactive oxygen species (ROS) and the loss of mitochondrial membrane potential (MMP) were elevated in human oral cancer OC2 cells compared with the un-irradiated cells. In contrast, these effects remained unchanged in normal HGF cells after exposure to LPLI. LPLI also induced apoptosis in caspase-3 dependent manner in human oral cancer OC2 cells, a mode of action that could be mediated by ROS and mitochondrial damage. CONCLUSION Our findings imply LPLI might be a potential therapy for oral cancers.
Collapse
Affiliation(s)
- Wei-Zhe Liang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
| | - Pei-Feng Liu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan.,Department of Biotechnology, Fooyin University, Kaohsiung, 831, Taiwan
| | - Earl Fu
- Department of Periodontology, School of Dentistry, National Defense Medical Center and Tri-Service General Hospital, Taipei, 114, Taiwan
| | - Hao-Sheng Chung
- Department of Stomatology, Kaohsiung Veteran General Hospital, Kaohsiung, 813, Taiwan
| | - Chung-Ren Jan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
| | - Chih-Hsuan Wu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
| | - Chih-Wen Shu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
| | - Yao-Dung Hsieh
- Department of Periodontology, School of Dentistry, National Defense Medical Center and Tri-Service General Hospital, Taipei, 114, Taiwan.,Department of Stomatology, Kaohsiung Veteran General Hospital, Kaohsiung, 813, Taiwan.,Department of Dentistry, Kaohsiung Veterans General Hospital, Pingtung, 912, Taiwan
| |
Collapse
|
10
|
In vitro study on the safety of near infrared laser therapy in its potential application as postmastectomy lymphedema treatment. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2015; 151:285-96. [PMID: 26355716 DOI: 10.1016/j.jphotobiol.2015.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 07/29/2015] [Accepted: 08/11/2015] [Indexed: 12/18/2022]
Abstract
Clinical studies demonstrated the effectiveness of laser therapy in the management of postmastectomy lymphedema, a discomforting disease that can arise after surgery/radiotherapy and gets progressively worse and chronic. However, safety issues restrict the possibility to treat cancer patients with laser therapy, since the effects of laser radiation on cancer cell behavior are not completely known and the possibility of activating postmastectomy residual cancer cells must be considered. This paper reports the results of an in vitro study aimed to investigate the effect of a class IV, dual-wavelength (808 nm and 905 nm), NIR laser system on the behavior of two human breast adenocarcinoma cell lines (namely, MCF7 and MDA-MB361 cell lines), using human dermal fibroblasts as normal control. Cell viability, proliferation, apoptosis, cell cycle and ability to form colonies were analyzed in order to perform a cell-based safety testing of the laser treatment in view of its potential application in the management of postmastectomy lymphedema. The results showed that, limited to the laser source, treatment conditions and experimental models used, laser radiation did not significantly affect the behavior of human breast adenocarcinoma cells, including their clonogenic efficiency. Although these results do not show any significant laser-induced modification of cancer cell behavior, further studies are needed to assess the possibility of safely applying NIR laser therapy for the management of postmastectomy lymphedema.
Collapse
|
11
|
Yang Y, Yu B, Sun D, Wu Y, Xiao Y. The dose-dependence biological effect of laser fluence on rabbit fibroblasts derived from urethral scar. Lasers Med Sci 2014; 30:1019-29. [PMID: 25388915 DOI: 10.1007/s10103-014-1683-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 10/20/2014] [Indexed: 11/28/2022]
Abstract
Two-micrometer laser vaporization resection has been used in clinic for years, but some patients received the treatment are still faced with excessive and abnormal wound repair which leads to the recurrent of urethral stricture eventually. Fibroblasts play a key role in the processes of "narrow-expansion/operation-restenosis" recurring problems. Here, we investigated the effect of laser fluence biomodulation on urethral scar fibroblasts as well as the underlying mechanism. Urethral scar fibroblasts were isolated and cultured, and laser irradiation (2 μm) was applied at different laser fluence or doses (0, 0.125, 0.5, 2, 8, 32 J/cm(2)) with a single exposure in 1 day. The effect of 2-μm laser irradiation on cell proliferation, viability, and expression of scar formation related genes were investigated. Two-micrometer laser irradiation with intermediate dose (8 J/cm(2)) promoted scar fibroblasts proliferation and reactive oxygen species (ROS) production, while higher doses of 32 J/cm(2) are suppressive as it decreased the survival rate, viability, and proliferation of fibroblasts. In addition, qRT-PCR and Western blotting results both proven that collagen type I, collagen IV, MMP9, and CTGF display significant increase, yet the TGF-β1 expression was severely reduced at intermediate dose (8 J/cm(2)) group when compared with the others groups. Our findings suggest the scar formation-related genes are sensitive to intermediate laser irradiation dose, the most in scar fibroblasts. We revealed the bioeffect and molecular mechanism of 2-μm laser irradiation on rabbit urethral scar fibroblasts. Our study provides new insights into the mechanisms which involved in the excessive and abnormal wound repair of 2-μm laser vaporization resection. These results could potentially contribute to further study on biological effects and application of 2-μm laser irradiation in urethral stricture therapy.
Collapse
Affiliation(s)
- Yong Yang
- Department of Urology, First Hospital Affiliated to General Hospital of PLA, No. 51 Fucheng Road, Haidian District, Beijing, 100048, China,
| | | | | | | | | |
Collapse
|
12
|
Dungel P, Hartinger J, Chaudary S, Slezak P, Hofmann A, Hausner T, Strassl M, Wintner E, Redl H, Mittermayr R. Low level light therapy by LED of different wavelength induces angiogenesis and improves ischemic wound healing. Lasers Surg Med 2014; 46:773-80. [PMID: 25363448 DOI: 10.1002/lsm.22299] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVE Low-level light therapy (LLLT) has been revealed as a potential means to improve wound healing. So far, most studies are being performed with irradiation in the red to near-infrared spectra. Recently, we showed that blue light (470 nm) can significantly influence biological systems such as nitric oxide (NO) metabolism and is able to release NO from nitrosyl-hemoglobin or mitochondrial protein complexes. Therefore, the aim of this study was to evaluate and compare the therapeutic value of blue or red light emitting diodes (LEDs) on wound healing in an ischemia disturbed rodent flap model. STUDY DESIGN/MATERIALS AND METHODS An abdominal flap was rendered ischemic by ligation of one epigastric bundle and subjected to LED illumination with a wavelength of 470 nm (blue, n = 8) or 629 nm (red, n = 8) each at 50 mW/cm(2) and compared to a non-treated control group (n = 8). Illumination was performed for 10 minutes on five consecutive days. RESULTS LED therapy with both wavelengths significantly increased angiogenesis in the sub-epidermal layer and intramuscularly (panniculus carnosus muscle) which was associated with significantly improved tissue perfusion 7 days after the ischemic insult. Accordingly, tissue necrosis was significantly reduced and shrinkage significantly less pronounced in the LED-treated groups of both wavelengths. CONCLUSIONS LED treatment of ischemia challenged tissue improved early wound healing by enhancing angiogenesis irrespective of the wavelength thus delineating this noninvasive means as a potential, cost effective tool in complicated wounds.
Collapse
Affiliation(s)
- Peter Dungel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Gomes Henriques ÁC, Ginani F, Oliveira RM, Keesen TSL, Galvão Barboza CA, Oliveira Rocha HA, de Castro JFL, Della Coletta R, de Almeida Freitas R. Low-level laser therapy promotes proliferation and invasion of oral squamous cell carcinoma cells. Lasers Med Sci 2014; 29:1385-95. [PMID: 24526326 DOI: 10.1007/s10103-014-1535-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 01/28/2014] [Indexed: 01/27/2023]
Abstract
Low-level laser therapy (LLLT) has been shown to be effective in promoting cell proliferation. There is speculation that the biostimulatory effect of LLLT causes undesirable enhancement of tumor growth in neoplastic diseases since malignant cells are more susceptible to proliferative stimuli. This study evaluated the effects of LLLT on proliferation, invasion, and expression of cyclin D1, E-cadherin, β-catenin, and MMP-9 in a tongue squamous carcinoma cell line (SCC25). Cells were irradiated with a diode laser (660 nm) using two energy densities (0.5 and 1.0 J/cm(2)). The proliferative potential was assessed by cell growth curves and cell cycle analysis, whereas the invasion of cells was evaluated using a Matrigel cell invasion assay. Expression of cyclin D1, E-cadherin, β-catenin, and MMP-9 was analyzed by immunofluorescence and flow cytometry and associated with the biological activities studied. LLLT induced significantly the proliferation of SCC25 cells at 1.0 J/cm(2), which was accomplished by an increase in the expression of cyclin D1 and nuclear β-catenin. At 1.0 J/cm(2), LLLT significantly reduced E-cadherin and induced MMP-9 expression, promoting SCC25 invasion. The results of this study demonstrated that LLLT exerts a stimulatory effect on proliferation and invasion of SCC25 cells, which was associated with alterations on expression of proteins studied.
Collapse
|
14
|
Shedding light on a new treatment for diabetic wound healing: a review on phototherapy. ScientificWorldJournal 2014; 2014:398412. [PMID: 24511283 PMCID: PMC3913345 DOI: 10.1155/2014/398412] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 10/09/2013] [Indexed: 12/22/2022] Open
Abstract
Impaired wound healing is a common complication associated with diabetes with complex pathophysiological underlying mechanisms and often necessitates amputation. With the advancement in laser technology, irradiation of these wounds with low-intensity laser irradiation (LILI) or phototherapy, has shown a vast improvement in wound healing. At the correct laser parameters, LILI has shown to increase migration, viability, and proliferation of diabetic cells in vitro; there is a stimulatory effect on the mitochondria with a resulting increase in adenosine triphosphate (ATP). In addition, LILI also has an anti-inflammatory and protective effect on these cells. In light of the ever present threat of diabetic foot ulcers, infection, and amputation, new improved therapies and the fortification of wound healing research deserves better prioritization. In this review we look at the complications associated with diabetic wound healing and the effect of laser irradiation both in vitro and in vivo in diabetic wound healing.
Collapse
|
15
|
Adly AS, Haggag MH, Mostafa MSM. Low Intensity Laser Irradiation Influence Proliferation of Mesenchymal Stem Cells: Comparison of Experimental Data to Intelligent Agent-Based Model Predictions. APPLIED METHODS AND TECHNIQUES FOR MECHATRONIC SYSTEMS 2014. [DOI: 10.1007/978-3-642-36385-6_16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
16
|
Anwer AG, Gosnell ME, Perinchery SM, Inglis DW, Goldys EM. Visible 532 nm laser irradiation of human adipose tissue-derived stem cells: effect on proliferation rates, mitochondria membrane potential and autofluorescence. Lasers Surg Med 2012; 44:769-78. [PMID: 23047589 DOI: 10.1002/lsm.22083] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2012] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND OBJECTIVE The photobiological effect of laser light on cells and tissues originates from light absorption by endogenous chromophores and hence it depends on the wavelength of light source and cell type. Earlier studies regarding the biostimulation effects of green laser light investigated a wide variety of cells but not adipose tissue-derived stem cells (ADSCS). In this study we reported the in vitro effect of 532-nm Nd:YAG laser on proliferation, mitochondrial activity of these mesenchymal stem cells (MSCs) on the autofluorescence emission at wavelengths associated with nicotinamide adenine dinucleotide (NADH) and flavoproteins. MATERIALS AND METHODS ADSCS were exposed to 532 nm second harmonic generation laser light at moderate power density (0.153 W/cm(2)) for periods of 30, 45, 60, 180, and 300 seconds. Mitochondrial membrane potential was measured using JC1 stain and confocal laser scanning microscopy, cell proliferation rates, and cellular autofluorescence emission at 450 and 540 nm wavelengths were measured using micro plate spectrofluorometer 48 hours after irradiation. RESULTS Shorter (30-60 seconds) exposure times led to significantly increased proliferation, attributed to increased mitochondrial activity (P < 0.05). At longer exposures we observed a significant decrease in proliferation and autofluorescence (P < 0.05). Strong correlation was observed between proliferation rates of cells and autofluorescence intensity. CONCLUSION Our results show that autofluorescence of the respiratory chain components and key autofluorescent metabolites offers a non-invasive method to quantify cellular response to laser irradiation.
Collapse
Affiliation(s)
- Ayad G Anwer
- Ewa M Goldys MQ BioFocus Research Centre, Macquarie University, North Ryde, 2109 NSW, Australia
| | | | | | | | | |
Collapse
|
17
|
Wu JY, Wang YH, Wang GJ, Ho ML, Wang CZ, Yeh ML, Chen CH. Low-power GaAlAs laser irradiation promotes the proliferation and osteogenic differentiation of stem cells via IGF1 and BMP2. PLoS One 2012; 7:e44027. [PMID: 22962596 PMCID: PMC3433487 DOI: 10.1371/journal.pone.0044027] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 08/01/2012] [Indexed: 11/18/2022] Open
Abstract
Low-power laser irradiation (LPLI) has been found to induce various biological effects and cellular processes. Also, LPLI has been shown to promote fracture repair. Until now, it has been unclear how LPLI promotes bone formation and fracture healing. The aim of this study was to investigate the potential mechanism of LPLI-mediated enhancement of bone formation using mouse bone marrow mesenchymal stem cells (D1 cells). D1 cells were irradiated daily with a gallium-aluminum-arsenide (GaAlAs) laser at dose of 0, 1, 2, or 4 J/cm(2). The lactate dehydrogenase (LDH) assay showed no cytotoxic effects of LPLI on D1 cells, and instead, LPLI at 4 J/cm(2) significantly promoted D1 cell proliferation. LPLI also enhanced osteogenic differentiation in a dose-dependent manner and moderately increased expression of osteogenic markers. The neutralization experiments indicated that LPLI regulated insulin-like growth factor 1 (IGF1) and bone morphogenetic protein 2 (BMP2) signaling to promote cell proliferation and/or osteogenic differentiation. In conclusion, our study suggests that LPLI may induce IGF1 expression to promote both the proliferation and osteogenic differentiation of D1 cells, whereas it may induce BMP2 expression primarily to enhance osteogenic differentiation.
Collapse
Affiliation(s)
- Jyun-Yi Wu
- Institute of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Yan-Hsiung Wang
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
| | - Gwo-Jaw Wang
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
- Department of Orthopaedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
- Department of Orthopaedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Mei-Ling Ho
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
| | - Chau-Zen Wang
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
| | - Ming-Long Yeh
- Institute of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan, Republic of China
- * E-mail: (MY); (CC)
| | - Chia-Hsin Chen
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, Republic of China
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
- Department of Physical Medicine and Rehabilitation, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan, Republic of China
- * E-mail: (MY); (CC)
| |
Collapse
|
18
|
Influence of three laser wavelengths on human fibroblasts cell culture. Lasers Med Sci 2012; 28:457-63. [DOI: 10.1007/s10103-012-1084-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Accepted: 03/13/2012] [Indexed: 12/14/2022]
|
19
|
Feng J, Zhang Y, Xing D. Low-power laser irradiation (LPLI) promotes VEGF expression and vascular endothelial cell proliferation through the activation of ERK/Sp1 pathway. Cell Signal 2012; 24:1116-25. [PMID: 22326662 DOI: 10.1016/j.cellsig.2012.01.013] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 01/10/2012] [Accepted: 01/26/2012] [Indexed: 12/11/2022]
Abstract
Angiogenesis, the growth of new blood vessels from pre-existing vessels, represents an excellent therapeutic target for the treatment of wound healing and cardiovascular disease. Herein, we report that LPLI (low-power laser irradiation) activates ERK/Sp1 (extracellular signal-regulated kinase/specificity protein 1) pathway to promote VEGF expression and vascular endothelial cell proliferation. We demonstrate for the first time that LPLI enhances DNA-binding and transactivation activity of Sp1 on VEGF promoter in vascular endothelial cells. Moreover, Sp1-regulated transcription is in an ERK-dependent manner. Activated ERK by LPLI translocates from cytoplasm to nuclear and leads to increasing interaction with Sp1, triggering a progressive phosphorylation of Sp1 on Thr453 and Thr739, resulting in the upregulation of VEGF expression. Furthermore, selective inhibition of Sp1 by mithramycin-A or shRNA suppresses the promotion effect of LPLI on cell cycle progression and proliferation, which is also significantly abolished by inhibition of ERK activity. These findings highlight the important roles of ERK/Sp1 pathway in angiogenesis, supplying potential strategy for angiogenesis-related diseases with LPLI treatment.
Collapse
Affiliation(s)
- Jie Feng
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | | | | |
Collapse
|
20
|
Saygun I, Nizam N, Ural AU, Serdar MA, Avcu F, Tözüm TF. Low-level laser irradiation affects the release of basic fibroblast growth factor (bFGF), insulin-like growth factor-I (IGF-I), and receptor of IGF-I (IGFBP3) from osteoblasts. Photomed Laser Surg 2012; 30:149-54. [PMID: 22235971 DOI: 10.1089/pho.2011.3079] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE It was the aim of the present study to evaluate whether the laser irradiation of osteoblasts could enhance the release of growth factors including basic fibroblast growth factor (bFGF), insulin-like growth factor-I (IGF-I), and receptor of IGF-I (IGFBP3). BACKGROUND DATA Low-level laser therapy (LLLT) has been shown to have biostimulatory effects on various cell types by enhancing production of some cytokines and growth factors. MATERIALS AND METHODS Human mesenchymal stem cells (MSCs) were seeded in osteogenic medium and differentiated into osteoblasts. Three groups were formed: in the first group (single dose group), osteoblasts were irradiated with laser (685 nm, 25 mW, 14.3 mW/cm(2), 140 sec, 2 J/cm(2)) for one time; and in the second group, energy at the same dose was applied for 2 consecutive days (double dose group). The third group was not irradiated with laser and served as the control group. Proliferation, viability, bFGF, IGF-I, and IGFBP3 levels were compared between groups. RESULTS Both of the irradiated groups revealed higher proliferation, viability, bFGF, IGF-I, and IGFBP3 expressions than did the nonirradiated control group. There was increase in bFGF and IGF-I expressions and decrease in IGFBP3 in the double dose group compared to single dose group. CONCLUSIONS The results of the present study indicate that LLLT increases the proliferation of osteoblast cells and stimulates the release of bFGF, IGF-I, and IGFBP3 from these cells. The biostimulatory effect of LLLT may be related to the enhanced production of the growth factors.
Collapse
Affiliation(s)
- Işıl Saygun
- Department of Periodontology, Gülhane Military Medical Academy, Ankara, Turkey.
| | | | | | | | | | | |
Collapse
|
21
|
Murayama H, Sadakane K, Yamanoha B, Kogure S. Low-power 808-nm laser irradiation inhibits cell proliferation of a human-derived glioblastoma cell line in vitro. Lasers Med Sci 2011; 27:87-93. [DOI: 10.1007/s10103-011-0924-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 04/06/2011] [Indexed: 10/18/2022]
|
22
|
Low intensity light stimulates nitrite-dependent nitric oxide synthesis but not oxygen consumption by cytochrome c oxidase: Implications for phototherapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2011; 102:182-91. [DOI: 10.1016/j.jphotobiol.2010.12.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 11/03/2010] [Accepted: 12/01/2010] [Indexed: 12/14/2022]
|
23
|
Low-level laser therapy: a useful technique for enhancing the proliferation of various cultured cells. Lasers Med Sci 2011; 27:237-49. [DOI: 10.1007/s10103-011-0885-2] [Citation(s) in RCA: 282] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Accepted: 01/05/2011] [Indexed: 12/16/2022]
|
24
|
Peplow PV, Chung TY, Baxter GD. Laser Photobiomodulation of Proliferation of Cells in Culture: A Review of Human and Animal Studies. Photomed Laser Surg 2010; 28 Suppl 1:S3-40. [DOI: 10.1089/pho.2010.2771] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Philip V. Peplow
- Department of Anatomy & Structural Biology, University of Otago, New Zealand
| | - Tzu-Yun Chung
- Department of Anatomy & Structural Biology, University of Otago, New Zealand
| | - G. David Baxter
- Centre for Physiotherapy Research, School of Physiotherapy, University of Otago, New Zealand
| |
Collapse
|
25
|
Argon laser phototherapy could eliminate the damage effects induced by the ionizing radiation “gamma radiation” in irradiated rabbits. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2010; 99:29-35. [DOI: 10.1016/j.jphotobiol.2010.01.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2009] [Revised: 01/09/2010] [Accepted: 01/25/2010] [Indexed: 11/20/2022]
|