Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 530
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Photochem Photobiol ; 100(1): 225-232, 2024.
Article in English | MEDLINE | ID: mdl-37254280

ABSTRACT

There are limited data on comparison of pulsed and continuous wave in photobiomodulation therapy (PBM). This study aimed to investigate the effect of PBM with 980 nm laser in pulsed and continuous wave on the proliferation and migration of human gingival fibroblasts (HGF) cells. Cultured HGF were divided into three main groups: (1) irradiated in pulsed mode (frequencies of 50 and 25 KHz; energy densities of 3 and 5 J/cm2 ), (2) irradiated in continuous mode (energy densities of 3.2 and 5.2 J/cm2 ), and (3) no irradiation as control group. HGF proliferation rate was measured by MTT assay at 24, 48, and 72 h post irradiation. In addition, HGF migration rate was measured by scratch test at 24 h post PBM. At 24 h, the group received continuous irradiation at 5.2 J/cm2 showed significantly higher proliferation compared with the control group (p = 0.012). At 48 and 72 h, the groups received continuous, and 50 Hz pulsed irradiation at energy densities of 5.2 and 5 J/cm2 respectively, had significantly higher HGF proliferation rates compared to the control (p < 0.05). Only the continuous irradiations were effective in significant increase of the cell migration. In conclusion, continuous PBM at energy density of 5.2 J/cm2 showed promising effect on HGF proliferation and migration.


Subject(s)
Low-Level Light Therapy , Humans , Cell Proliferation/radiation effects , Cell Survival , Lasers , Fibroblasts/radiation effects
2.
Arch Oral Biol ; 159: 105874, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38147800

ABSTRACT

OBJECTIVE: Photobiomodulation therapy (PBMT) has proven to reduce inflammation and pain and increase wound healing. Thus, the aim of this study was to analyze the effects of PBMT parameters on migration, proliferation, and gene expression after ionizing radiation and bacterial-induced stress in an in vitro study. DESIGN: Keratinocytes (HaCaT) and Fibroblasts (HGFs) were grown in DMEM with 10 % fetal bovine serum until stressful condition induction with lipopolysaccharide (LPS) of Escherichia coli (1 µg/mL), Porphyromonas gingivalis protein extract (5 µg/mL) and ionizing radiation (8 Gy). Low-laser irradiation (660 nm, 30 mW) was carried out in four sessions, with 6 h intervals, and energy density of 2, 3, 4, and 5 J/cm². Scratch assays, immunofluorescence, and RT-qPCR were performed. RESULTS: Treated fibroblasts and keratinocytes showed significant response in proliferation and migration after scratch assays (p < 0.05). Higher expressions of α-SMA in fibroblasts and F-actin in keratinocytes were observed in cells subjected to 3 J/cm². PI3K-pathway genes expression tended to enhance in fibroblasts, presenting a higher relative expression when compared to keratinocytes. In keratinocytes, PBMT groups demonstrated deregulated expression for all inflammatory cytokines' genes tested while fibroblasts presented a tendency to enhance those genes expression in a dose dependent way. CONCLUSIONS: The present study showed that delivering 660 nm, 30 mW was effective to stimulate cell migration, proliferation and to accelerate wound healing. PBMT can modulate cytokines and pathways involved in wound repair. The different energy densities delivering distinct responses in vitro highlights that understanding laser parameters is fundamental to improve treatment strategies.


Subject(s)
Low-Level Light Therapy , Phosphatidylinositol 3-Kinases , Keratinocytes , Fibroblasts/radiation effects , Cell Proliferation/radiation effects , Radiation, Ionizing
3.
J. oral res. (Impresa) ; 12(1): 86-99, abr. 4, 2023. ilus
Article in English | LILACS | ID: biblio-1512278

ABSTRACT

Objective: The objective of this study was to investigate the morphology, proliferation, and differentiation of gingival mesenchymal stem cells (GMSCs) irradiated with a 970 nm Diode Laser (LLLT). It is essential to validate the efficacy of treatment, optimize irradiation conditions and guarantee the safety and quality of stem cells for future use in dental applications. Materials and Methods: GMSCs were cultured in standard conditions and irradiated with a Diode laser (970 nm, 0.5W) with an energy density of 9J/cm2. Cell proliferation was assessed with the WST-1 proliferation kit. GMSCs were differentiated into chondrogenic and osteogenic lineages. Cell morphology was performed with Hematoxylin/eosin staining, and quantitative nuclear analysis was done. Cell viability was monitored with trypan blue testing. Results: GMSCs subjected to irradiation demonstrated a significant increase in proliferation at 72 hours compared to the non-irradiated controls (p=0.027). This indicates that the 970 nm diode laser has a stimulatory effect on the proliferation of GMSCs. LLLT-stimulated GMSCs exhibited the ability to differentiate into chondrogenic and osteogenic lineages. A substantial decrease in cell viability was observed 24 hours after irradiation (p=0.024). However, after 48 hours, the cell viability recovered without any significant differences. This indicates that there might be a temporary negative impact on cell viability immediately following irradiation, but the cells were able to recover and regain their viability over time. Conclusions: This study support that irradiation with a 970 nm diode laser could stimulate the proliferation of GMSCs, maintain their ability to differentiate into chondrogenic and osteogenic lineages, and has minimal impact on the mor- phological characteristics of the cells. These results support the potential use of NIR Lasers in combination with GMSCs as a promising strategy for dental treatments.


Objetivo: El objetivo de este estudio fue investigar la morfología, proliferación y diferenciación de las células madre mesenquimatosas (GMSC) irradiadas con un láser de diodo de 970 nm (LLLT). Es fundamental validar la eficacia del tratamiento, optimizar las condiciones de irradiación y garantizar la seguridad y calidad de las células madre para su uso futuro en aplicaciones dentales.Materiales y Métodos: Las GMSC se cultivaron en condiciones estándar y se irradiaron con un láser de diodo (970 nm, 0,5 W) con una densidad de energía de 9 J/cm2. La proliferación celular se evaluó con el kit de proliferación WST-1. Las GMSC se diferenciaron en linajes condrogénicos y osteogénicos. La morfología celular se realizó con tinción de hematoxilina/eosina y se realizó un análisis nuclear cuantitativo. La viabilidad celular se controló con prueba de azul de tripano. Resultados: Las GMSC sometidas a irradiación demostraron un aumento significativo en la proliferación a las 72 horas en comparación con los controles no irradiados (p=0,027). Esto indica que el láser de diodo de 970 nm tiene un efecto estimulante sobre la proliferación de GMSC. Las GMSC estimuladas con LLLT exhibieron la capacidad de diferenciarse en linajes condrogénicos y osteogénicos. Se observó una disminución sustancial de la viabilidad celular 24 horas después de la irradiación (p=0,024). Sin embargo, después de 48 horas, la viabilidad celular se recuperó sin diferencias significativas. Esto indica que podría haber un impacto negativo temporal en la viabilidad de las células inmediatamente después de la irradiación, pero las células pudieron recuperarse y recuperar su viabilidad con el tiempo. Conclusión: En conclusión, este estudio respalda que la irradiación con un láser de diodo de 970 nm podría estimular la proliferación de GMSC, mantener su capacidad para diferenciarse en linajes condrogénicos y osteogénicos y tiene un impacto mínimo en las características morfológicas de las células. Estos resultados respaldan el uso potencial de láseres NIR en combinación con GMSC como una estrategia prometedora para tratamientos dentales.


Subject(s)
Humans , Low-Level Light Therapy , Cell Proliferation/radiation effects , Lasers, Semiconductor , Mesenchymal Stem Cells/radiation effects , In Vitro Techniques , Gingiva/radiation effects
4.
J Photochem Photobiol B ; 240: 112665, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36736031

ABSTRACT

Worldwide, the most frequently diagnosed cancer is female breast cancer, and it poses a serious global health threat. Traditional cancer therapies are associated with various side effects, so developing better therapies for breast cancer is necessary, such as laser therapy which could be a promising treatment option. The aim of the current study was to investigate the femtosecond laser irradiation effect on breast cancer using T47D cell line as an in vitro model. Cells were seeded at a density of 5 × 104 cells/well in 96-well plates and incubated overnight. After that, the cells were exposed to femtosecond laser irradiation at various wavelengths falling in the UV, visible, and IR ranges for 3, 5, or 10 min and at a constant power of 100 mW. Cell viability was measured directly and 24 h after femtosecond laser irradiation using MTT assay. When using different femtosecond laser irradiation parameters, especially the 380 and 400 nm femtosecond laser irradiation, there was significant inhibition of breast cancer cell growth, either directly or 24 h after femtosecond laser exposure. Also, 420 and 440 nm significantly affected the viability of the cells. It was also observed that increasing exposure time enhances the observed effect, so 10 min exposure time was the best time of exposure. However, 700, 720, 750, and 780 nm did not significantly affect the cells viability with different exposure times. It was possible to conclude from the aforementioned results that femtosecond laser irradiation exerted a significant anticancer effect against T47D cells. Consequently, the femtosecond laser could be used successfully for breast cancer management.


Subject(s)
Breast Neoplasms , Laser Therapy , Low-Level Light Therapy , Female , Humans , Breast Neoplasms/radiotherapy , Lasers , Cell Proliferation/radiation effects
5.
Int J Mol Sci ; 24(4)2023 Feb 13.
Article in English | MEDLINE | ID: mdl-36835175

ABSTRACT

Glioblastoma is one of the most malignant and lethal forms of primary brain tumors in adults. Linearol, a kaurane diterpene isolated from different medicinal plants, including those of the genus Sideritis, has been found to possess significant anti-oxidant, anti-inflammatory and anti-microbial properties. In this study, we aimed to determine whether linearol could exhibit anti-glioma effects when given alone or in combination with radiotherapy in two human glioma cell lines, U87 and T98. Cell viability was examined with the Trypan Blue Exclusion assay, cell cycle distribution was tested with flow cytometry, and the synergistic effects of the combination treatment were analyzed with CompuSyn software. Linearol significantly suppressed cell proliferation and blocked cell cycle at the S phase. Furthermore, pretreatment of T98 cells with increasing linearol concentrations before exposure to 2 Gy irradiation decreased cell viability to a higher extent than linearol or radiation treatment alone, whereas in the U87 cells, an antagonistic relationship was observed between radiation and linearol. Moreover, linearol inhibited cell migration in both tested cell lines. Our results demonstrate for the first time that linearol is a promising anti-glioma agent and further studies are needed to fully understand the underlying mechanism of this effect.


Subject(s)
Brain Neoplasms , Diterpenes , Glioblastoma , Glioma , Humans , Glioblastoma/metabolism , Glioma/pathology , Diterpenes/therapeutic use , Cell Line , Cell Line, Tumor , Cell Proliferation/radiation effects , Brain Neoplasms/metabolism
6.
Lasers Med Sci ; 38(1): 27, 2022 Dec 27.
Article in English | MEDLINE | ID: mdl-36574084

ABSTRACT

Wound treatment, especially for chronic and infected wounds, has been a permanent socio-economical challenge. This study aimed to investigate the ability of red light at 661 nm to accelerate wound healing an in vitro wound model using 3T3 fibroblasts. The purpose is further specified in clarifying the mechanisms of wound closure by means of intracellular ROS production, proliferation and migration of cells, and cellular orientation. Illumination effects of red light from a diode laser (661 nm) at different doses on 3T3 cell viability was assessed via MTT assay and tested in a scratch wound model. Wound closure rates were calculated by image analysis at 0, 24, and 48 h after laser treatment. ROS production was monitored and quantified immediately and 24 h after the treatment by fluorescence microscopy. Cellular orientation was quantified by image analysis. No phototoxic energy doses used and increased cell viability in most of the groups. Scratch assay revealed an energy interval of 3 - 4.5 J/cm2 that promote higher wound healing rate 24 h post treatment. An increase in ROS production was also observed 24 h post irradiation higher in the group with the highest wound healing rate. Also, cellular orientation toward the margin of the wound was observed and quantified after irradiation. Low power laser light at 661 nm activated both the migration and proliferation in the in vitro model used, providing evidence that it could also accelerate wound healing in vivo. Also, ROS production and cellular orientation seem to play an important role in wound healing process.


Subject(s)
Low-Level Light Therapy , Reactive Oxygen Species , Low-Level Light Therapy/methods , Cell Proliferation/radiation effects , Wound Healing/radiation effects , Fibroblasts/radiation effects , Lasers, Semiconductor/therapeutic use
7.
Sci Rep ; 12(1): 18907, 2022 11 07.
Article in English | MEDLINE | ID: mdl-36344673

ABSTRACT

Photobiomodulation (PBM) refers to the use of light to modulate cellular processes, and has demonstrated utility in improving wound healing outcomes, and reducing pain and inflammation. Despite the potential benefits of PBM, the precise molecular mechanisms through which it influences cell behavior are not yet well understood. Inconsistent reporting of key light parameters has created uncertainty around optimal exposure profiles. In addition, very low intensities of light, < 0.1 J/cm2, have not been thoroughly examined for their use in PBM. Here, we present a custom-made compact, and modular LED-based exposure system for studying the effects of very low-intensity visible light (cell proliferation, migration, ROS production, and mitochondrial membrane potential) of three different wavelengths in a parallel manner. The device allows for six repeats of three different exposure conditions plus a non-irradiated control on a single 24-well plate. The immortalised human keratinocyte cell line, HaCaT, was selected as a major cellular component of the skin epidermal barrier. Furthermore, an in vitro wound model was developed by allowing the HaCaT to form a confluent monolayer, then scratching the cells with a pipette tip to form a wound. Cells were exposed to yellow (585 nm, 0.09 mW, ~ 3.7 mJ/cm2), orange (610 nm, 0.8 mW, ~ 31 mJ/cm2), and red (660 nm, 0.8 mW, ~ 31 mJ/cm2) light for 10 min. 48 h post-irradiation, immunohistochemistry was performed to evaluate cell viability, proliferation, ROS production, and mitochondrial membrane potential. The results demonstrate increased proliferation and decreased scratch area for all exposure conditions, however only red light increased the mitochondrial activity. Oxidative stress levels did not increase for any of the exposures. The present exposure system provides opportunities to better understand the complex cellular mechanisms driven by the irradiation of skin cells with visible light.


Subject(s)
Low-Level Light Therapy , Humans , Low-Level Light Therapy/methods , Reactive Oxygen Species/metabolism , Keratinocytes/metabolism , Wound Healing/radiation effects , Cell Proliferation/radiation effects , Light
8.
Lasers Med Sci ; 37(9): 3681-3692, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36227520

ABSTRACT

The effect of near infrared (NIR) laser irradiation on proliferation and osteogenic differentiation of buccal fat pad-derived stem cells and the role of transient receptor potential (TRP) channels was investigated in the current research. After stem cell isolation, a 940 nm laser with 0.1 W, 3 J/cm2 was used in pulsed and continuous mode for irradiation in 3 sessions once every 48 h. The cells were cultured in the following groups: non-osteogenic differentiation medium/primary medium (PM) and osteogenic medium (OM) groups with laser-irradiated (L +), without irradiation (L -), laser treated + Capsazepine inhibitor (L + Cap), and laser treated + Skf96365 inhibitor (L + Skf). Alizarin Red staining and RT-PCR were used to assess osteogenic differentiation and evaluate RUNX2, Osterix, and ALP gene expression levels. The pulsed setting showed the best viability results (P < 0.05) and was used for osteogenic differentiation evaluations. The results of Alizarin red staining were not statistically different between the four groups. Osterix and ALP expression increased in the (L +) group. This upregulation abrogated in the presence of Capsazepine, TRPV1 inhibitor (L + Cap); however, no significant effect was observed with Skf96365 (L + Skf).


Subject(s)
Adipose Tissue , Stem Cells , Transient Receptor Potential Channels , Humans , Adipose Tissue/radiation effects , Cell Differentiation/genetics , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Cells, Cultured , Osteogenesis/genetics , Osteogenesis/radiation effects , Stem Cells/radiation effects , Transient Receptor Potential Channels/metabolism , Infrared Rays
9.
Lasers Med Sci ; 37(9): 3509-3516, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36066778

ABSTRACT

Low-level laser therapy (LLLT) also known as photobiomodulation is a treatment to change cellular biological activity. The exact effects of LLLT remain unclear due to the different irradiation protocols. The purpose of this study was to investigate the effects of LLLT by three different irradiation methods on the proliferation and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in vitro. BMSCs were inoculated in 24-well plates and then irradiated or not (control) with a laser using three different irradiation methods. The irradiation methods were spot irradiation, covering irradiation, and scanning irradiation according to different spot areas (0.07 cm2 or 1.96 cm2) and irradiation areas (0.35 cm2 or 1.96 cm2), respectively. The laser was applied three times at energy densities of 4 J/cm2. The cell proliferation by CCK-8. ALP activity assay, alizarin red, and quantitative real-time polymerase chain reaction (RT-PCR) were performed to assess osteogenic differentiation and mineralization. Increases in cell proliferation was obvious following irradiation, especially for covering irradiation. The ALP activity was significantly increased in irradiated groups compared with non-irradiated control. The level of mineralization was obviously improved following irradiation, particularly for covering irradiation. RT-PCR detected significantly higher expression of ALP, OPN, OCN, and RUNX-2 in the group covering than in the others, and control is the lowest. The presented results indicate that the biostimulative effects of LLLT on BMSCs was influenced by t he irradiation method, and the covering irradiation is more favorable method to promote the proliferation and osteogenic differentiation of BMSCs.


Subject(s)
Low-Level Light Therapy , Mesenchymal Stem Cells , Osteogenesis/genetics , Osteogenesis/radiation effects , Bone Marrow Cells , Mesenchymal Stem Cells/radiation effects , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Cells, Cultured
10.
J Photochem Photobiol B ; 234: 112527, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35914464

ABSTRACT

In recent decades, the laser treatment of cancer has been introduced as a promising treatment option. Because of the maldistribution of optical energy and an ambiguous boundary between the normal and tumor tissues, laser irradiation can stimulate residual cancer cells, leading to a cancer regrowth. As photobiomodulation (PBM) is involved in an extensive range of cellular responses, profound comprehension of photo-stimulated mechanisms against the cancer cells is required to establish a safety margin for PBM. Therefore, we aimed to identify the stimulant effects of PBM at various wavelengths against the tumor cells to establish a safety margin for the laser treatment. CT26 murine colon cancer cells were exposed to either 405 (BL), 635 (VIS), or 808 (NIR) nm laser lights at the fluences of 0, 10, 30, and 50 J/cm2. In addition, CT26 tumor-bearing mice were irradiated with BL, VIS, or NIR at a fluence of 30 J/cm2. Both the proliferation and angiogenesis potential of the CT26 cells and tumors were evaluated using the MTT assay, western blot, and immunohistochemistry (IHC) staining analyses. Although cell viability was not statistically significant, BL significantly induced p-ERK upregulation in the CT26 cells, indicating that PBM with BL can stimulate proliferation. In vivo tests showed that the NIR group exhibited the maximum relative tumor volume, and BL yielded a slight increase compared to the control. In the IHC staining and western blot analyses, both BL and NIR increased the expression of EGFR, VEGF, MMP-9, and HIF-1α, which are related to the proliferation and angiogenesis-related factors. Further investigations will be pursued to clarify the molecular pathways that depend on the cancer cell types and laser wavelengths for the establishment of safety guidelines in clinical environments.


Subject(s)
Colonic Neoplasms , Low-Level Light Therapy , Animals , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Colonic Neoplasms/radiotherapy , Light , Mice
11.
Lasers Med Sci ; 37(8): 3031-3049, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35751706

ABSTRACT

Low-level laser therapy (LLLT)-induced photobiomodulation (PBM) stimulates bone tissue regeneration by inducing osteoblast differentiation and mitochondrial activation. However, the role of reactive oxygen species (ROS) in this process remains controversial. The aim of this systematic review was to collect and analyze the available literature on the cellular and molecular effects of LLLT on osteoblasts and the role of ROS in this process. A search was conducted in PubMed, ScienceDirect, Scopus, and Web of Science. Studies published in English over the past 15 years were selected. Fourteen articles were included with moderate (n = 9) and low risk of bias (n = 5). Thirteen studies reported the use of diode lasers with wavelengths (λ) between 635 and 980 nm. One study used an Nd:YAG laser (λ1064 nm). The most commonly used λ values were 808 and 635 nm. The energy densities ranged from 0.378 to 78.75 J/cm2, and irradiation times from 1.5 to 300 s. Most studies found increases in proliferation, ATP synthesis, mitochondrial activity, and osteoblastic differentiation related to moderate and dose-dependent increases in intracellular ROS levels. Only two studies reported no significant changes. The data presented heterogeneity owing to the variety of LLLT protocols. Although several studies have shown a positive role of ROS in the induction of proliferation, migration, and differentiation of different cell types, further research is required to determine the specific role of ROS in the osteoblastic cell response and the molecular mechanisms involved in triggering previously reported cellular events.


Subject(s)
Low-Level Light Therapy , Osteoblasts , Adenosine Triphosphate/metabolism , Cell Proliferation/radiation effects , Lasers, Semiconductor/therapeutic use , Reactive Oxygen Species/metabolism
12.
Photobiomodul Photomed Laser Surg ; 40(5): 334-342, 2022 May.
Article in English | MEDLINE | ID: mdl-35559714

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.


Subject(s)
Low-Level Light Therapy , Cell Proliferation/radiation effects , Dental Pulp , Lasers, Semiconductor/therapeutic use , Stem Cells/radiation effects
13.
Lasers Med Sci ; 37(7): 2855-2863, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35394552

ABSTRACT

Photobiomodulation (PBM) therapy utilizes low-power lasers to modulate the viability of living human cells and leads to changes in proliferation, differentiation, adhesion and gene expression, even though the rearrangement of cytoskeleton was not previously studied. The present study aims to evaluate the photobiological effects on the elastic behavior of human osteosarcoma cells (MG-63) and their morphological changes. Fluorescence staining, confocal imaging and atomic force microscopy (AFM) topography were performed to study the effects of PBM therapy with the exposure of 532 nm-25mW, 650 nm-3mW, 650 nm-150mW and 780 nm-70mW beams following the 5-min continuous irradiation. The area of each beam was 3.14cm2 with a source-surface distance of 20 cm. Besides the cell proliferation assessment, the migratory potential of MG-63 was determined with the wound healing technique. The results indicated an increase in stiffness and shape index of radiation-induced cells 24 h after exposure along with the obvious F-actins changes. But, cell stiffening was not observed 72 h after 532 nm laser irradiation. Also, a decrease in the migration rate was seen in all of the groups after 72 h of irradiation except cells treated with 532 nm wavelength. However, 532 nm laser beams increase the migratory potential 24 h after exposure. Within 72 h after irradiation, the cell proliferation was only affected by applying 532 nm and 650 nm-150mW laser beams. It was concluded that applying photobiomodulation with wavelengths of 650 nm (at both utilized powers) and 780 nm alters the migration capability and provides a quantitative description of cytoskeletal changes. Moreover, membrane stiffening can be considered as the biological marker of PBM treatments.


Subject(s)
Low-Level Light Therapy , Osteosarcoma , Cell Proliferation/radiation effects , Cytoskeleton , Elastic Modulus , Humans , Low-Level Light Therapy/methods , Osteosarcoma/radiotherapy
14.
Int J Mol Sci ; 23(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35162958

ABSTRACT

The development of nerve conduits with a three-dimensional porous structure has attracted great attention as they closely mimic the major features of the natural extracellular matrix of the nerve tissue. As low levels of reactive oxygen species (ROS) function as signaling molecules to promote cell proliferation and growth, this study aimed to fabricate protoporphyrin IX (PpIX)-immobilized cellulose (CEPP) monoliths as a means to both guide and stimulate nerve regeneration. CEPP monoliths can be fabricated via a simple thermally induced phase separation method and surface modification. The improved nerve tissue regeneration of CEPP monoliths was achieved by the activation of mitogen-activated protein kinases, such as extracellular signal-regulated kinases (ERKs). The resulting CEPP monoliths exhibited interconnected microporous structures and uniform morphology. The results of in vitro bioactivity assays demonstrated that the CEPP monoliths with under 0.54 ± 0.07 µmol/g PpIX exhibited enhanced photodynamic activity on Schwann cells via the generation of low levels of ROS. This photodynamic activation of the CEPP monoliths is a cell-safe process to stimulate cell proliferation without cytotoxic side effects. In addition, the protein expression of phospho-ERK increased considerably after the laser irradiation on the CEPP monoliths with low content of PpIX. Therefore, the CEPP monoliths have a potential application in nerve tissue regeneration as new nerve conduits.


Subject(s)
Cellulose/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Protoporphyrins/pharmacology , Schwann Cells/cytology , Animals , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Low-Level Light Therapy , Nerve Regeneration , Nerve Tissue/chemistry , Phosphorylation , Protoporphyrins/chemistry , Rats , Reactive Oxygen Species/metabolism , Schwann Cells/drug effects , Schwann Cells/metabolism , Schwann Cells/radiation effects
15.
Lasers Med Sci ; 37(3): 1993-2003, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34787763

ABSTRACT

This study aims to evaluate the impact of red LED irradiation on the viability, proliferation, colonogenic potential, markers expression along with osteogenic and chondrogenic differentiation of dental pulp stem cells. DPSCs were isolated from sound human permanent teeth using enzymatic digestion method and seeded with regular culture media. Cells at P4 were irradiated using red LED Light (627 nm, 2 J/cm2) and examined for growth kinetics, and multilineage differentiation using the appropriate differentiation media. The irradiated groups showed an increase in cellular growth rates, cell viability, clonogenic potential, and decrease in population doubling time compared to the control group. Cells of the irradiated groups showed enhanced differentiation towards osteogenic and chondrogenic lineages as revealed by histochemical staining using alizarin red and alcian blue stains. Photobiomodulation is an emerging promising element of tissue engineering triad besides stem cells, scaffolds, and growth factors.


Subject(s)
Low-Level Light Therapy , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Cells, Cultured , Dental Pulp , Humans , Kinetics , Osteogenesis/radiation effects , Stem Cells
16.
Photochem Photobiol ; 98(4): 969-973, 2022 07.
Article in English | MEDLINE | ID: mdl-34932837

ABSTRACT

Photobiomodulation is recognized as an effective method for adjunct therapy in periodontal treatments. Our purpose in this study was to investigate the effects of different energy densities of 915 nm diode laser on the viability and viability capacity of human gingival fibroblast cells. Cell samples were examined in five groups, including four irradiation groups with low-level diode laser 915 nm, 1, 2, 3, 4 J cm-2 and a control group (no Laser irradiation). Cell viability and viability were measured 1, 3 and 5 days after irradiation by MTT and DAPI assay. Statistical differences between groups at any time were analyzed by one-way ANOVA and a post hoc Turkey's test. The cell viability and viability capacity increased on the third day at an energy density of 3 J cm-2 ; (P-value = 0.007) and the fifth day at energy densities of 2, 3 and 4 J cm-2 was recorded compared with the control group (P-value = 0.000). Also, a significant decrease in the viability and viability of irradiated cells with an energy density of 1 J cm-2 was found (P-value = 0.033). According to our results, Photobiomodulation with 915 nm diode laser has a positive stimulating effect on the viability and viability capacity of human gingival fibroblast cells.


Subject(s)
Low-Level Light Therapy , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Fibroblasts/radiation effects , Gingiva , Humans , Lasers, Semiconductor , Low-Level Light Therapy/methods
17.
Bioengineered ; 13(1): 917-929, 2022 01.
Article in English | MEDLINE | ID: mdl-34968160

ABSTRACT

Radiation therapy (RT) is widely applied in cancer treatment. The sensitivity of tumor cells to RT is the key to the treatment. This study probes the role and mechanism of miR-20b-5p in Pembrolizumab's affecting the radiosensitivity of tumor cells. After Pembrolizumab treatment or cell transfection (miR-20b-5p mimics and miR-20b-5p inhibitors), tumor cells (NCI-H460 and ZR-75-30) were exposed to RT. The sensitivity of NCI-H460 and ZR-75-30 to RT was evaluated by monitoring cell proliferation and apoptosis. The dual-luciferase reporter assay and RNA immunoprecipitation (RIP) were adopted to evaluate the binding relationship between miR-20b-5p and CD274 (PD-L1). The xenograft model was established in nude mice to examine the mechanism of action of Pembrolizumab in vivo. Our outcomes exhibited that either Pembrolizumab treatment or miR-20b-5p overexpression potentiated radiosensitivity of tumor cells. Overexpressing miR-20b-5p enhanced radiosensitization of Pembrolizumab in vivo and in vitro by targeting PD-L1 and inactivating PD-L1/PD1. Overall, miR-20b-5p overexpression combined with Pembrolizumab potentiated cancer cells' sensitivity to RT by repressing PD-L1/PD1.Abbreviations Akt: serine/threonine kinase 1; cDNA: complementary DNA; CO2: carbon dioxide; EDTA: Ethylene Diamine Tetraacetic Acid; ENCORI: The Encyclopedia of RNA Interactomes; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; IGF2BP2: insulin like growth factor 2 mRNA binding protein 2; IHC: Immunohistochemistry; LncRNA MALAT1: Long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1; miRNAs: MicroRNAs; Mt: Mutant type; MTT: 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide; NC: negative control; NR2F2: nuclear receptor subfamily 2 group F member 2; NSCLC: non-small cell lung cancer; OD: optical density; PBS: phosphate-buffered saline; PD-L1: Programmed death-ligand 1; PD-1: programmed death 1; PI3K: phosphatidylinositol 3-kinase; qRT-PCR: Quantitative reverse transcription-polymerase chain reaction; RIP: RNA immunoprecipitation; RIPA: Radio Immunoprecipitation Assay; RRM2: ribonucleotide reductase regulatory subunit M2; RT: Radiation therapy; U6: U6 small nuclear RNA; V: volume; WB: Western blot; Wt: wild type; x ± sd: mean ± standard deviation.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , B7-H1 Antigen/genetics , Breast Neoplasms/therapy , Carcinoma, Non-Small-Cell Lung/therapy , Down-Regulation , Lung Neoplasms/therapy , MicroRNAs/genetics , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Chemoradiotherapy , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Lung Neoplasms/genetics , Mice , Mice, Nude , Transfection , Xenograft Model Antitumor Assays
18.
Photochem Photobiol ; 98(5): 1182-1189, 2022 09.
Article in English | MEDLINE | ID: mdl-34970994

ABSTRACT

Several methods have been proposed to enhance the regeneration and healing time in periodontal therapy. Photobiomodulation therapy (PBMT) is a recently suggested novel technique for this purpose. This study aimed to compare the efficacy of PBMT with various laser wavelengths and energy densities on proliferation of human periodontal ligament mesenchymal stem cells (PDLMSCs). The wells containing PDLMSCs were subjected to laser irradiation at 635, 660, 808 and 980 nm wavelengths with 1, 1.5, 2.5 and 4 J cm-2 energy densities. Cell proliferation and viability were evaluated after 1, 3 and 5 days with the methyl thiazolyl tetrazolium (MTT) assay and 4,6-diamidino-2-phenylindole (DAPI) staining. No significant difference was observed among the experimental and the control groups on day 1 (P > 0.05). On day 3, 808 nm laser at 4 J cm-2 energy density and 980 nm laser at all densities had significant differences with control group. On day 5, the control group had significant differences in cell proliferation with 808 nm laser at 2.5 and 4 J cm-2 energy densities, and 980 nm laser at all densities. PBMT with 635, 660, 808 and 980 nm wavelengths increased the proliferation of PDLMSCs but the maximum cell viability was prominent after irradiation by 980 nm laser with energy density of 4 J cm-2 on day 3.


Subject(s)
Low-Level Light Therapy , Mesenchymal Stem Cells , Cell Proliferation/radiation effects , Humans , Lasers , Low-Level Light Therapy/methods , Periodontal Ligament
19.
J Photochem Photobiol B ; 225: 112349, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34742031

ABSTRACT

PURPOSE: To investigate the safety of photobiomodulation therapy (PBM) in tumors and its potential as a radiosensitizer when combined with radiotherapy. METHODS: We have performed in vitro experiments in A431 cells to assess proliferation and cell cycle after PBM, as well as clonogenic assay and H2AX-gamma immunolabeling to quantify double strand breaks after the combination of PBM and radiation. In vivo experiments in xenografts included Kaplan-Meier survival analysis, optical coherence tomography (OCT) and histological analysis. RESULTS: PBM did not induce proliferation in vitro, but increased the G2/M fraction by 27% 24h after illumination, resulting in an enhancement of 30% in radiation effect in the clonogenic assay. The median survival of the PBM-RT group increased by 4 days and the hazard ratio was 0.417 (CI 95%: 0.173-1.006) when compared to radiation alone. OCT analysis over time demonstrated that PBM increases tumor necrosis due to radiation, and histological analysis showed that illumination increased cell differentiation and angiogenesis, which may play a role in the synergetic effect of PBM and radiation. CONCLUSION: PBM technique may be one of the most appropriate approaches for radiosensitizing tumors while protecting normal tissue because of its low cost and low training requirements for staff.


Subject(s)
Low-Level Light Therapy/methods , Neoplasms/therapy , Radiation-Sensitizing Agents/administration & dosage , Animals , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Humans , Mice , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/therapy
20.
Int J Mol Sci ; 22(21)2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34769035

ABSTRACT

Background: The invention of non-ionizing emission devices revolutionized science, medicine, industry, and the military. Currently, different laser systems are commonly used, generating the potential threat of excessive radiation exposure, which can lead to adverse health effects. Skin is the organ most exposed to laser irradiation; therefore, this study aims to evaluate the effects of 445 nm, 520 nm, and 638 nm non-ionizing irradiation on keratinocytes and fibroblasts. Methods: Keratinocytes and fibroblasts were exposed to a different fluency of 445 nm, 520 nm, and 638 nm laser irradiation. In addition, viability, type of cell death, cell cycle distribution, and proliferation rates were investigated. Results: The 445 nm irradiation was cytotoxic to BJ-5ta (≥58.7 J/cm2) but not to Ker-CT cells. Exposure influenced the cell cycle distribution of Ker-CT (≥61.2 J/cm2) and BJ-5ta (≥27.6 J/cm2) cells, as well as the Bj-5ta proliferation rate (≥50.5 J/cm2). The 520 nm irradiation was cytotoxic to BJ-5ta (≥468.4 J/cm2) and Ker-CT (≥385.7 J/cm2) cells. Cell cycle distribution (≥27.6 J/cm2) of Ker-CT cells was also affected. The 638 nm irradiation was cytotoxic to BJ-5ta and Ker-CT cells (≥151.5 J/cm2). The proliferation rate and cell cycle distribution of BJ-5ta (≥192.9 J/cm2) and Ker-CT (13.8 and 41.3 J/cm2) cells were also affected. Conclusions: At high fluences, 455 nm, 520 nm, and 638 nm irradiation, representing blue, green, and red light spectra, are hazardous to keratinocytes and fibroblasts. However, laser irradiation may benefit the cells at low fluences by modulating the cell cycle and proliferation rate.


Subject(s)
Fibroblasts/radiation effects , Skin/radiation effects , Cell Cycle/radiation effects , Cell Death/radiation effects , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Cells, Cultured , Humans , Lasers , Light , Low-Level Light Therapy/methods
SELECTION OF CITATIONS
SEARCH DETAIL