Open Access
23 December 2013 Investigation of low-level laser therapy potentiality on proliferation and differentiation of human osteoblast-like cells in the absence/presence of osteogenic factors
Nora Bloise, Gabriele Ceccarelli, Paolo Minzioni, Marco Vercellino, Laura Benedetti, Maria Gabriella C. De Angelis, Marcello Imbriani, Livia Visai
Author Affiliations +
Abstract
Several studies have shown that low-level laser irradiation (LLLI) has beneficial effects on bone regeneration. The objective of this study was to examine the in vitro effects of LLLI on proliferation and differentiation of a human osteoblast-like cell line (Saos-2 cell line). Cultured cells were exposed to different doses of LLLI with a semiconductor diode laser (659 nm; 10 mW power output). The effects of laser on proliferation were assessed daily up to seven days of culture in cells irradiated once or for three consecutive days with laser doses of 1 or 3  J/cm 2 . The obtained results showed that laser stimulation enhances the proliferation potential of Saos-2 cells without changing their telomerase pattern or morphological characteristics. The effects on cell differentiation were assessed after three consecutive laser irradiation treatments in the presence or absence of osteo-inductive factors on day 14. Enhanced secretion of proteins specific for differentiation toward bone as well as calcium deposition and alkaline phosphatase activity were observed in irradiated cells cultured in a medium not supplemented with osteogenic factors. Taken together these findings indicate that laser treatment enhances the in vitro proliferation of Saos-2 cells, and also influences their osteogenic maturation, which suggest it is a helpful application for bone tissue regeneration.

1.

Introduction

The identification of an amplification approach to improve and accelerate the modeling and remodeling of bone tissue is an intriguing challenge in the field of tissue engineering. Over the past 40 years, the importance of physical factors that modulate and accelerate biological processes has been highlighted. Physical factors, as well as biochemical factors, may induce cells to reprogram their functions and dynamically adapt to environmental conditions.13 These factors may be applied in biomedicine and biotechnology in order to drive and modulate cell behavior for therapeutic purposes. In this context, studies focused on physical factors that promote tissue regeneration and that are therefore highly promising. Low-level laser therapy (LLLT) is a well-established clinical tool used to treat pathological tissue conditions, inflammatory processes, and promote wound healing.4 LLLT refers to the use of lasers emitting a wavelength ranging from 600 to 1100 nm with an output power within 1 to 500 mW. A number of different laser light sources, including semiconductor diode lasers, He-Ne, and argon lasers, have been applied in different treatments.5 LLLT has been found to modulate various biological processes, such as collagen production,6 DNA synthesis,7 mitochondrial respiration, and ATP synthesis.8 Various studies have demonstrated that LLLT promotes both repair and regeneration.9,10 It has been shown that LLLT induces cell proliferation,1113 promotes angiogenesis,14 and allows the wound site to close more quickly.10,15 Additionally, it has been demonstrated that LLLT irradiation results in an increase of circulating antioxidants and expression of heat shock protein. LLLT was also shown to stimulate the expression of multiple genes related to cellular migration, proliferation, anti-apoptosis, and prosurvival elements responsive to nuclear factor kappa-light-chain-enhancer of activated B cells,14 besides modulating the production of growth factors and cytokines.1012

Although the biochemical and cellular mechanisms behind the action of LLLT are not fully understood, it has been postulated that laser irradiation at low doses modulates cell activity by different mechanisms. It is proposed that its effect occurs through the absorption of red and near-IR light by chromophores, in particular, cytochrome c oxidase, which is contained in the respiratory chain located within the mitochondria.16 It is supposed that this absorption of energy may cause the photodissociation of inhibitory nitric oxide from cytochrome c oxidase and lead to increased enzyme activity and electron transport.17 Furthermore, it has been demonstrated that LLLT enhances cell proliferation by increasing the release of calcium into the cytoplasm, which triggers mitosis by causing a short-term rise in the intracellular pH.18

Increased proliferation after laser irradiation has been shown in cells of different origin, including fibroblasts, lymphocytes, mesenchymal and cardiac stem cells.19 With respect to bone, LLLT has been applied in several clinical situations, such as orthodontic treatment, alveolar repair after tooth extraction, bone fracture healing, and osseointegration of dental implants as an adjuvant therapy.20 In vitro laser irradiation improves proliferation and differentiation of human osteoblast cells,21 and in vivo irradiation increases the functional attachment of biomaterial implants to bone.22 This finding is also supported by in vitro observations that LLLT enhances the proliferation and osteogenic differentiation of murine mesenchymal stem cells.23 The effects of LLLT should be more thoroughly investigated before low level laser therapy can be considered as a potential to for bone regeneration.24

Presently, optimal laser irradiation conditions for obtaining the maximum stimulating cell proliferation are still under debate. In several studies concerning the use of laser treatments to improve tissue repair, the authors highlight the difficulty in obtaining comparable results due to different laser sources, treatment protocols, and experimental models used. Some reports have clearly indicated that laser irradiation speeds up tissue repair and a consensus has been reached. (1) For a biological effect, laser wavelengths should be red or near-red (600 to 1200 nm). (2) For biostimulatory effects, the dose or energy density to induce cell proliferation should be between 0.05 and 10J/cm2, whereas energies greater than this value (>10J/cm2) may promote antiproliferative effects.25

Considering evidence that LLLT has a positive effect on bone healing, the aim of the present study was to study the in vitro effects of LLL irradiation (LLLI) by an aluminum gallium indium phosphide (AlGaInP) semiconductor diode laser at a wavelength of 659 nm on the proliferation and differentiation of Saos-2 cells.26 With this aim, it was decided to first investigate the optimal stimulating parameters for Saos-2 proliferation and then evaluate the LLLI capacity under these parameters to induce osteogenic differentiation.

2.

Materials and Methods

2.1.

Irradiation Protocol Settings

The LLLI experiments were performed by using a semiconductor laser-diode, which emits a maximum output power of 130 mW at 659 nm (Table 1). The particular laser diode used for these experiments is based on an AlGaInP semiconductor (model ML101J27-01, produced by Mitsubishi Electric Corp., Tokyo, Japan, Fig. 1), which emits a single transverse-mode laser beam with a typical divergence of 10 and 17 deg in the two directions transverse to the beam propagation axis. After free propagation in air, the central portion of the beam was selected through an iris diaphragm in order to guarantee a constant beam intensity over the entire irradiated field (a circular well with radius r=8mm). The relative positions of the laser source, the mask, and the samples were appropriately chosen to obtain complete illumination of the well containing the test sample, while simultaneously avoiding illumination of adjacent wells. Both the operating temperature and current of the laser source were monitored by proper drivers (by Thorlabs GmbH, Germany). The value of the optical power incident on the samples, taking into account losses due to Fresnel reflection occurring on the cover, was set at 10 mW, corresponding to an intensity of 5mW/cm2. The optical power was periodically monitored to check for any laser performance degradation.

Table 1

Laser irradiation parameters (Mitsubishi laser, model ML101J27-01, Tokyo, Japan).

ParametersValueParametersValue
Beam divergence (parallel)10 degBeam divergence (perpendicular)17 deg
Operating current182 mARadiation wavelength659 nm
Emitted power97 mWPower after the screen11 mW
Power impinging on the sample10 mWPower density on the sample5mWcm2

Fig. 1

Setup used for cell irradiation. Aluminum-gallium-indium-phosphide semiconductor diode laser emitting at 659 nm was perpendicularly positioned above the tissue culture plate containing the SAOS-2 cell monolayer.

JBO_18_12_128006_f001.png

2.2.

Temperature Measurement

In order to evaluate the possible thermal effects induced by the laser irradiation on cell cultures, a commercial temperature probe was used to measure the temperature in the culture dish before turning on the laser source, and also during laser irradiation. A T-type thermocouple (RS Components, code 621-2209, Mi, Italia) was used as the sensor; this has a good sensitivity (0.1°C) and a very broad operating range (from 200 to +350°C). The temperature in the culture dish before laser treatment was 22±0.1°C. The samples were then irradiated for 600 s in a single exposition at dose of 3J/cm2. The final temperature was of 21.8±0.1°C, indicating that the temperature remained unchanged during irradiation.

2.3.

Reagents

The human osteosarcoma cell line Saos-2 was obtained from the American Type Culture Collection (HTB85, ATCC, Manassas, Virginia).26,27 Unless otherwise specified, all reagents were from Sigma Aldrich (St. Louis, Missouri). Dr. Larry W. Fisher (National Institutes of Health, Bethesda, Maryland) provided us with the rabbit polyclonal anti type-I and III collagen, anti-decorin, anti-osteopontin, anti-osteocalcin, anti-osteonectin, and anti-alkaline phosphatase. Polyclonal antibody against human fibronectin (FN) was produced as previously described.28 For the Western blot analysis, we used the anti-actin antibody and the phospho site-specific antibody against Akt-P on Ser 473 and Akt from Cell Signaling Technologies®, Danvers, Massachusetts. Horseradish peroxidase (HRP)-conjugated secondary antibodies (HRP-conjugated from Dako) were used, and detection was performed by enhanced chemiluminescent substrate (ECL) solutions (Pierce Thermo Fisher Scientific, Rockford, Illinois). The TRAPeze™ kit used to detect telomerase activity was purchased from CHEMICON International (Millipore, Billerica, Massachusetts).

2.4.

Cell Culture

Saos-2 cells were cultured in McCoy’s 5A modified medium with L-glutamine and HEPES (Cambrex Bio Science, Baltimore, Maryland), supplemented with 15% fetal bovine serum, 1% L-glutamine, 0.4% antibiotics, 2% sodium pyruvate, and 0.2% fungizone. For osteogenic differentiation analysis, dexamethasone and β-glycerophosphate (both osteogenic factors) were added to the previously indicated medium at a concentration of 108M and 10 mM, respectively. Ascorbic acid, another osteogenic supplement, is a component of McCoy’s 5A modified medium. The cells were cultured at 37°C, 5% CO2, routinely trypsinized after confluence, counted, and seeded at a density of 3×104cells/well in 24-well plates.

2.5.

Cell Viability

2.5.1.

MTT assay

In order to establish an appropriate laser irradiation dose to induce proliferation, cell viability was evaluated after treatment with different doses and irradiation protocols: Saos-2 were divided into five groups: group I, dark control (not exposed); groups II and III, exposed to a single laser dose of 1J/cm2 (200 s irradiation) or 3J/cm2 (600 s irradiation) on day 0, and groups IV and V, exposed for three consecutive days to 1 or 3J/cm2 (multiple doses) on days 0, 1, and 2 (Table 2). The laser doses used have been previously reported to stimulate osteoblast growth in vitro.25 The quantitative 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) test was used to assess cell viability following a single or multiple laser doses at days 1, 2, 3, and 7 of cell culture. The MTT solution (0.5mg/mL, Sigma-Aldrich) was added to cells for 3 h. Absorbance was measured at 570 nm with a microplate reader (BioRad Laboratories, Hercules, California). The optical density value is directly proportional to the number of viable cells in the culture medium.

Table 2

Experimental design.

Scheme of irradiationEnergy density (J/cm2)Time (days)
Proliferation assays
MTT testSingle and multiple doses1 to 31, 2, 3, 7
Fluorescein diacetate assaySingle and multiple doses1 to 37
Morphological observationSingle and multiple doses1 to 37
Western blotSingle dose310, 20, 30 min after irradiation
Telomerase activityMultiple doses37
Differentiation assays
Alkaline phosphatase activityMultiple doses3J/cm214
Calcium quantificationMultiple doses3J/cm214
Real-time polymerase chain reaction (PCR)Multiple doses3J/cm214
ELISAMultiple doses3J/cm214
Confocal laser scanning microscope analysisMultiple doses3J/cm214
SEM analysisMultiple doses3J/cm214
Note: Single irradiation was carried out at day 0; multiple irradiations were performed for three consecutive days at days 0, 1, and 2.

2.5.2.

Fluorescein diacetate assay

At day 7 of culture, a qualitative viability assay [fluorescein diacetate (FDA) assay] was performed on each group. Briefly, cells were washed with phosphate-buffered saline (PBS, pH 7.4) and incubated with 100 μL of FDA working solution for 10 min. After washing with PBS, to stain the nuclei of dead cells, cells were incubated at room temperature (RT) for 3 min with 30 μL of propidium iodide (2μg/ml) and then observed with a confocal fluorescence microscope (Leica TCS SPII Microsystems, Bensheim, Germany).

2.6.

Western Blot Analysis

Saos-2 cells treated with a laser dose of 3J/cm2 were analyzed for Akt and Akt phosphorylation on Ser 473 by Western blot after 10, 20, and 30 min irradiation. Briefly, cells were scraped from the dish and lysed with ice-cold lysis buffer (50 mM Tris pH 7.5, 50 mM NaCl, 5 mM ethylenediaminetetraacetic acid, 0.1% Triton, and 1 mM sodium sodium orthovanadate) for 30 min on ice. The lysates were centrifuged at 12,000 rpm for 5 min at 4°C, and supernatant protein concentrations were determined. Equivalent samples were subjected to SDS-PAGE on 8% gel. The proteins were then transferred onto nitrocellulose membranes and probed with primary antibodies anti-phospho-Akt on Ser 473 and anti-Akt diluted 12000, followed by secondary antibodies conjugated to HRP (11000). Detection was performed with ECL solution and revealed by autoradiography. Densitometry analysis of the band was performed using Image TM Software. Bands were then quantified by densitometric analysis.

2.7.

Telomerase Assay

The Trapeze® gel-based telomerase detection kit was used to detect and evaluate telomerase activity in cells irradiated with multiple doses of 3J/cm2 and in control cells. This assay is a highly sensitive in vitro assay system for detecting telomerase activity and is based on an improved version of the original method described by Kim et al.28 The assay is a one-buffer, two-enzyme system that uses a polymerase chain reaction (PCR) to enhance the sensitivity of telomerase detection in small samples. For visualization of the reaction products, we used a nonradioactive method: running 25 μl of the products on a 12.5% nondenaturing-page gel in 0.5x Tris/Borate/EDTA buffer. After electrophoresis, the gel was stained with SYBR® green according to the manufacturer’s instructions (Life Technologies, ex Invitrogen, Carlsbad, California). The relative telomerase activity level is expressed as the total product generated (TPG), calculated by the following formula: TPG(units)=(xxo)/c/(rro)/cR×100; x is the signal intensity of 6-bp ladders in the sample; xo is the signal intensity in heat-treated sample; r is the signal intensity in TSR8 control; ro is the signal intensity in lysis buffer; c is the signal intensity in 36-bp internal control in the sample; and cR is the signal intensity in TSR8 control. The intensity of the TRAP product band and standard internal control bands were determined using Image™ Software.

2.8.

Confocal Laser Scanning Microscope Analysis

Saos-2 cells (3×104) were seeded on glass coverslips in growth medium and then processed for confocal laser scanning microscope (CLSM) analysis. For morphological observation, cells were stimulated with a single or multiple laser doses (1 or 3J/cm2). On day 7 of culture, cells were washed with PBS, fixed with 4% (w/v) paraformaldehyde solution for 30 min at 4°C, permeabilized with 0.1% Triton X-100, and finally incubated with the primary antibody anti-α tubulin overnight at 4°C.

For osteogenic protein labeling, cells were irradiated with multiple doses at 3J/cm2 and stained on day 14 of culture both in proliferative medium (PM, without osteogenic factors) and in osteogenic medium (OM, with osteogenic factors). Paraformaldehyde-fixed samples were blocked with PAT [PBS containing 1% (w/v) bovine serum albumin and 0.02% (v/v) Tween 20] for 1 h at RT. Cells were then incubated with specific primary antibodies (anti-type-I collagen, anti-alkaline phosphatase, and anti-osteocalcin rabbit polyclonal antisera) diluted 11000 in PAT overnight at 4°C. Following incubation with the primary antibody, cells were washed once with PBS and incubated with Alexa-Fluor-488 conjugated secondary antibody (diluted 1500 in PAT, Invitrogen, Carlsbad, California) for 1 h at RT. After extensive washes in PBS, nuclei were counterstained with propidium iodide (2μg/mL) for morphological observation and Hoechst 33342 (2μg/mL) for osteogenic protein labeling. Finally, samples were observed with a confocal fluorescence microscope (Leica TCS SPII Microsystems, Wetzlar, Germany).

2.9.

Osteogenic Differentiation

To investigate the effect of LLLI on Saos-2 osteogenic differentiation, cells treated with three consecutive doses at 3J/cm2 and cultured in PM or OM were analyzed on day 14 of culture (Table 2). Control cell groups were unexposed and cultured in PM or OM, respectively.

2.10.

Alkaline Phosphatase Activity

On day 14 of culture, the alkaline phosphatase (ALP) activity from LLLI stimulated and unstimulated samples cultured either in PM or OM was evaluated by a colorimetric end point assay as previously described.2 The assay measures the conversion of the colorless substrate p-nitrophenol phosphate (PNPP) by the enzyme ALP to the yellow product p-nitrophenol, where the rate of the color change corresponds to the amount of enzyme present in the solution. Briefly, an aliquot (1 mL) of 0.3 M PNPP (dissolved in glycine buffer, pH 10.5) was added to each sample at 37°C. After incubation, the reaction was stopped by the addition of 100 mL 5 M NaOH. Standards of PNPP in concentrations ranging from 0 to 50 mM were freshly prepared from dilutions of a 500 mM stock solution and incubated for 10 min with 7U of ALP (Sigma-Aldrich) previously dissolved in 500 mL of ddH2O. The absorbance reading was performed at 405 nm with a microplate reader (BioRad Laboratories, Hercules, California) using 100 mL of standard or sample placed into individual wells of a 96-well plate. Samples were run in triplicate and compared against a calibration curve of p-nitrophenol standards. The enzyme activity was expressed as micromoles of p-nitrophenol produced per minute per milligram of enzyme.

2.11.

Calcium Quantification

On day 14 of culture, the calcium deposition from LLLI stimulated and unstimulated samples cultured either in PM or OM was determined by calcein detection and calcium cresolphthalein complexone methods as previously described.29

2.11.1.

Calcein detection

At the end of cell incubation, each sample was rinsed with sterile PBS and stained with a calcein solution (5 mM in PBS; Invitrogen) for 30 min at 22°C. The samples were counterstained with a solution of Hoechst 33342 (2μg/mL) to target the cellular nuclei and then washed with PBS. The images were taken by blue excitation (bandpass, 450 to 480 nm; dichromatic mirror, DM500; barrier filter, BA515) with a fluorescence microscope at 20× magnification.

2.11.2.

Calcium-cresolphthalein complexone method

The calcium content of each sample was assayed to quantify the amount of mineralized matrix present and was measured using a Calcium Fast kit (Mercury SPA, Naples, Italy) according to the manufacturer’s instructions. The colorimetric end point assay measures the amount of purple-colored calcium-cresolphthalein complexone complex formed when cresolphthalein complexone binds to free calcium in an alkaline solution. Briefly, an aliquot (1 mL) of 1 N HCl was added to each sample and incubated for 24 h at RT to release calcium into solution. The sample supernatant was diluted 1/10 with the Assay Working Solution previously prepared by mixing equal parts of calcium-binding reagent and calcium buffer reagent provided by the Kit. Ca2+ standards in concentrations ranging from 0 to 10mg/mL were prepared from dilutions of a 100mg/mL stock solution of Ca2+. The absorbance reading was performed at 595 nm with a microplate reader (BioRad Laboratories) using 100 mL of standard or sample placed into individual wells of a 96-well plate. Samples were run in triplicate and compared against the standard solution calibration curve.

2.12.

Assay for Gene Expression

Total RNA from LLLI stimulated and unstimulated samples cultured in PM or OM was extracted on day 14 of culture using the Trizol reagent, according to the manufacturer’s protocol (Invitrogen). Reverse transcriptase-polymerase chain reaction (RT-PCR) was performed in order to evaluate the gene expression for bone sialoprotein (BOSP), decorin (DEC), fibronectin (FN), osteocalcin (OC), osteopontin (OP), type-I collagen (COL-I), type-III collagen (COL-III), alkaline phosphatase (ALP), osteonectin (OSN), and the housekeeping gene expression for glyceraldehyde-3-phosphate dehydrogenase (GAPDH). The reverse transcriptase reaction was performed with 300 ng total RNA using the iScript™ cDNA Synthesis kit from Bio-Rad. The primers (Primm s.r.l., Milan, Italy) were designed according to the published gene sequences, and the PCRs were performed with the GeneAmp PCR System 9700 (Applied Biosystems, Foster City, California) as previously reported.29 The primers used are indicated in Table 3.

Table 3

Primers used for qRT-PCR.

GenesUpstream primerDownstream primerAmplicon size (bp)
ALP5’- ACCTCGTTGACACCTGGAAG-3’5’-CCACCATCTCGGAGAGTGAC-3’189
BOSP5’-TGAGGCTGAGAATACCACAC-3’5’-GCCTAGTGGTGTGTTCTTAG-3’380
Col-I5’-TGTAAGCGGTGGTGGTTATG-3’5’-GGTAGCCATTTCCTTGGAAG-3’450
Col-III5’-TGGATCAGATGGTCTTCCA-3’5’-TCTCCATAATACGGGGCAA-3’620
Dec5’-CGAGTGGTCCAGTGTTCTGA-3’5’-AAAGCCCCATTTTCAATTCC-3’400
Fn5’-TGGAACTTCTACCAGTGCGAC-3’5’-TGTCTTCCCATCATCGTAACAC-3’500
GAPDH5’-TTCACCACCATGGAGAAGGC-3’5’-GGCATGGACTGTGGTCATGA-3’236
OC5’-GGCAGCGAGGTAGTGAAGAG-3’5’-CTGGAGAGGAGCAGAACTGG-3’230
OSN5’-CTTCAGACTGCCCGGAGA-3’5’-GAAAGAAGATCCAGGCCCTC-3’110
OP5’-TCACTGATTTTCCCACGGAC-3’5’-TCATAACTGTCCTTCCCACG-3’280
Note: ALP, alkaline phosphatase; BOSP, bone sialoprotein; Col-I, type-I collagen; Col-III, type-III collagen; Dec, decorin; Fn, fibronectin; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; OC, osteocalcin; OSN, osteonectin; and OP, osteopontin.

2.13.

Real-Time PCR

Total RNA from LLLI stimulated and unstimulated samples cultured in PM or OM was extracted on day 14 of culture with the NucleoSpin® RNA XS kit (Macherey-Nagel, Duren, Germany) and retro-transcribed to c-DNA with the iScript cDNA Synthesis Kit (BioRad Laboratories, Marnes-La-Coquette, France). A quantitative RT-PCR analysis was performed in a 48-well optical reaction plate using a MiniOpticon® Real Time PCR System (BioRad Laboratories) as previously described.2 Gene expression was analyzed in triplicate and normalized to GAPDH gene expression, using the Livak method.30 Analysis was performed in a total volume of 20 μL amplification mixture containing 2x (10 μL) Brilliant® SYBR® Green QPCR Master Mix (Stratagene, La Jolla, California), 2 μL cDNA, 0.4 μL of each primer, and 7.2 μL H2O. Thermal cycling was initiated by denaturation at 95 deg for 3 min, followed by 40 cycles at 95 deg for 5 s and 60 deg for 20 s. To perform the real-time analysis, we used Invitrogen™ (Carlsbad, California) primers for the genes listed in Table 3.

2.14.

Extraction of the Extracellular Matrix Proteins and ELISA Assay

On day 14 of culture, in order to evaluate the amount of extracellular matrix proteins produced by LLLI stimulated and unstimulated samples cultured either in PM or OM, an ELISA assay was performed as previously described.29 Briefly, the samples were washed extensively with sterile PBS to remove culture medium and then incubated for 24 h at 37°C with 1 mL of sterile sample buffer [20 mM Tris-HCl, 4 M GuHCl, 10 mM EDTA, 0.066% (w/v) sodium dodecyl sulphate (SDS), pH 8.0]. At the end of the incubation period, the total protein concentration in both culture systems was evaluated with the BCA Protein Assay Kit (Pierce Biotechnology Inc., Rockford, Illinois). Calibration curves to measure COL-I, COL-III, DEC, OP, OC, OSN, FN, and ALP were performed as previously described.2 We have taken into consideration that an underestimation of the absolute protein deposition is possible because the sample buffer used for matrix extraction contains SDS, which may interfere with protein adsorption during the ELISA assay. The amount of extracellular matrix constituents from samples was expressed as pg/(cells per well).

2.15.

Scanning Electron Microscopy Analysis

Saos-2 were seeded on plastic cell culture coverslip disks (Thermanox Plastic, Nalge Nunc International, Rochester, New York) and irradiated with multiple laser doses at 3J/cm2 in PM or OM. Control groups were treated under the same conditions. On day 14 of culture, cells were treated as previously described.2 The specimens were sputter coated with gold and observed at 250× and 1000× magnification, respectively, with a Leica Cambridge Stereoscan 440 microscope (Leica Microsystems) at 8 kV.

2.16.

Statistical Analysis

Each experimental condition was performed in triplicate in three separate experiments. Differences between groups were tested by one-way analysis of variance. Tukey’s test was used to correct for multiple comparisons. Statistical significance was established at two-tailed p0.05. All calculations were generated using GraphPad Prism 5.0 (GraphPad Inc., San Diego, California).

3.

Results

3.1.

Effect of Low-Level Laser Irradiation on Cell Proliferation and Morphology

To test whether LLLI can act as a proliferative factor, the experimental setup was performed as indicated in Table 2. At all levels of applied irradiation and at each time interval, cell vitality and morphology were evaluated by MTT assay, FDA, and CLSM, respectively (Fig. 2). The results of cell viability after a single dose at 1 or 3J/cm2 is shown in Fig. 2(Aa). Significant differences were detected in cell proliferation after a single dose at 1 or 3J/cm2 with respect to the dark control (p<0.05) on day 2. However, on days 3 and 7, these differences were not statistically significant. The results of cells treated with daily doses are also presented in Fig. 2(Ab). The viability analysis showed that repeated irradiation on three consecutive days with doses of 1 or 3J/cm2 resulted in a significantly higher proliferation when compared to the dark control group on days 2, 3, and 7 (p<0.05). It is possible to hypothesize that single dose may exert an effect on cell proliferation in the first few days after exposure, but this effect may not last, as previously reported.23 The effect of irradiation was then qualitatively evaluated by FDA assay at day 7 of culture in untreated/laser-treated cells after single or multiple doses [Fig. 2(B)]. Results showed comparable cell viability between the samples. The Saos-2 cell morphology after laser exposure was evaluated by CLSM and no differences in the cytoskeletal organization of alpha tubulin between the experimental groups was observed [Fig. 2(C)]. To further complete the analysis on proliferative activity following laser treatment, the activation of mitogenic kinase Akt was analyzed (Fig. 3). This protein was previously reported to be involved in LLLI-induced cell proliferation.31

Fig. 2

Time course of cell Saos-2 proliferation following irradiation with a single and daily laser doses. (Aa) The viability of groups irradiated with a single dose at 1 or 3J/cm2 on day 0. (Ab) The viability of groups irradiated with a daily dose of 1 or 3J/cm2 on days 0, 1, and 2. The MTT test was performed to evaluate cell viability after a single dose or daily dose exposures at days 1, 2, 3, and 7 of cell culture. After seven days of culture, all experimental groups were treated with fluorescein diacetate (green cells alive) and propidium iodide (red cells dead) (B). Dark control and groups irradiated with one or three consecutive doses at 3J/cm2 are shown (20× magnification, the scale bar represents 50 μm). SAOS-2 cell morphological analysis after laser treatment: alfa-tubulin is shown in green and nuclei in red (40× magnification, the scale bar represents 50 μm) (C). The dark control and irradiated sample with one or three consecutive doses at 3J/cm2 are shown.

JBO_18_12_128006_f002.png

Fig. 3

Representative Western blot analysis for Akt phosphorylation up to 30 min after a single laser dose at 3J/cm2. SAOS-2 were cultured for 24 h in serum free medium, irradiated, and collected at the indicated time after laser treatment and then analyzed for Akt phosphorylation (Ser-473) by Western blot. Akt and b-actin were used as controls. Results represent one of three replicates (a). Bar graph represents the phosphorylation level of the signal protein calculated by the ratio between the phosphorylated and total protein obtained in three different experiments. *p<0.05 versus time 0 (b).

JBO_18_12_128006_f003.png

Western blot analysis showed that laser treatment with a dose of 3J/cm2 is able to induce a transient increase in Akt phosphorylation/activation, which gradually reached a maximum level of activation 20 min after stimulation [Figs. 3(a) and 3(b)]. The correlation between telomerase activity and cell proliferation after laser irradiation was also investigated (Fig. 4). Telomerase activity was measured quantitatively with the TRAPEZE Gel Based Telomerase Detection Kit. As shown in Fig. 4, both stimulated and unstimulated samples were telomerase positive, but telomerase activity was not significantly increased in stimulated cells compared to the dark control (p>0.05).

Fig. 4

Electrophoresis image of telomerase activity in Saos-2 after irradiation for three consecutive days at a dose of 3J/cm2 in proliferative medium using the TRAPeze™ kit. One representative telomerase assay out of three similar ones is presented (a). Lane 1: TRAPeze quality control cell lysate (HeLa cells) heat-treated; lane 2: non-heat-treated extract of control HeLa cells; lane 3: heat-treated dark control cells; lane 4: non-heat-treated dark control cells; lane 5: heat-treated irradiated cells; lane 6: non-heat-treated irradiated cells; lanes 7 and 8: TSR8 control and 1X CHAP lysis buffer, respectively. Bar graph represents the telomerase products quantification (b). Telomerase activity (in total product generated units) was calculated by comparing the ratio of telomerase products to an internal standard for each lysate, as described by CHEMICON International.

JBO_18_12_128006_f004.png

3.2.

Effect of Low-Level Laser Irradiation on Cell Differentiation

To investigate whether low-level laser treatment was able to influence Saos-2 osteogenic differentiation (Table 2, for experimental setup), all analyses were performed on day 14 of culture on multiple-stimulated or unstimulated cells, with/without the addition of osteogenic factors in the culture medium. ALP activity, calcium deposition, gene expression, and bone extracellular matrix protein production were evaluated. The level of ALP activity was higher in cells in osteogenic medium (both unstimulated/laser stimulated) than cells in proliferative medium (both unstimulated/laser stimulated) [Fig. 5(B)]. The ALP activity was different between cells that were laser treated or untreated cultured in PM as well as in OM. Interestingly, the ALP activity of LLLI-exposed samples in PM was considerably higher when compared to unexposed samples in PM (*p<0.05); on the contrary, ALP activity of laser-exposed samples in OM was slightly lower than that of unexposed samples in OM (#p>0.05). These data are in accordance with the immunolocalization of ALP on the cell surface as observed by CLSM. A more intense green fluorescence signal was observed on laser-stimulated samples in PM and on stimulated/unstimulated samples in OM [Figs. 5(Ab), 5(Ac), and 5(Ad)] than on unstimulated cells in PM [Fig. 5(Aa)]. Figure 6 shows the calcium deposition detected by calcein assays: a more intense green fluorescence signal was observed on laser-stimulated cells in PM and laser-stimulated/unstimulated cells in OM [Figs. 6(Ab), 6(Ac), and 6(Ad)] than on unstimulated cells maintained in PM [Fig. 6(Aa)]. A significant difference was also observed between laser-exposed/unexposed cultures in PM. The results were quantitatively confirmed with the calcium creosolphatein complexone method [Fig. 6(B)].

Fig. 5

Alkaline phosphatase (ALP) deposition and activity of Saos-2 cells exposed to three consecutive doses at 3J/cm2 and cultured in proliferative medium (PM) or in osteogenic medium (OM). ALP presence was determined by confocal laser microscopy (40× magnification, the scale bar represents 50 μm) (A), and ALP activity was measured colourimetrically, corrected for the protein content measured with the BCA Protein Assay Kit and expressed as millimoles of p-nitrophenol produced per minute per milligram of protein (B). Bars express the mean values±SD of results from three experiments (*p<0.05 versus PM).

JBO_18_12_128006_f005.png

Fig. 6

Calcium matrix produced by Saos-2 cells irradiated with three consecutive doses at 3J/cm2 and cultured in PM or in OM. Calcium deposition was determined with a confocal laser scanning microscope [(A), 20× magnification, the scale bar represents 50 μm] and by quantification of calcium content as reported in the Materials and Methods section (B). Results are presented as an average±SD (*p<0.05 versus PM).

JBO_18_12_128006_f006.png

In order to characterize bone-specific gene expression, an RT-PCR analysis was performed at 14 days culture with/without osteogenic factors on unstimulated/stimulated samples. The qualitative RT-PCR (qRT-PCR) showed differences for the transcripts specific for COL-I, BOSP, and OP (Fig. 7). To further expand these data, a qRT-PCR for the ALP, BOSP, COL-I, and OP in laser-stimulated/unstimulated cells cultured in PM or OM was performed at 14 days using the ΔΔCt method. These genes are involved in the osteogenic process, and they are the most characterized to evaluate bone differentiation of cells.3236 As shown in Fig. 8, the real-time PCR data revealed no significant differences between laser-treated/untreated cells cultured in PM in the expression of ALP and COL-I, while a statistically significant down regulation of BOSP and OP expressions (p<0.05), which are genes related to the final phases of osteogenic differentiation, was observed [Fig. 8(a)]. No evident fold-increase was detected for these genes between laser-stimulated/unstimulated cells cultured in OM [Fig. 8(b)]. These findings were confirmed by the confocal analysis of COL-1 and osteocalcin (OCN) expression in laser-treated and untreated cultures in PM [Figs. 9(A) and 9(B)]. Instead, a more intense green fluorescence signal for COL-I and OCN in laser-stimulated/unstimulated cells was observed in osteogenic culture conditions compared with PM [Figs. 9(A) and 9(B)]. In Table 4, data are reported for the extracellular matrix protein deposition on day 14 of culture. An enhancement in COL-III, ALP, and DEC deposition was observed in laser-treated cells in PM. These were, respectively, 1.8-, 2-, and 1.8-fold greater when compared with unstimulated cells in PM (p<0.05). On the contrary, the level of bone proteins was not statistically different when measured in cells stimulated with laser and cultured in the presence of osteogenic factors compared with osteogenic control (Table 3; p>0.05). Moreover, two weeks after seeding, SEM revealed that cells formed a confluent multilayer with a more evident cellular density in laser-stimulated/unstimulated samples in PM [Figs. 10(a) and 10(b)] as compared with OM cultures [Figs. 10(c) and 10(d)]. At higher magnification, the characteristic cell morphology of PM and OM cultures following laser irradiation were observed: in PM, cells exhibited a round shape, while in OM, they were flat and elongated, typical of differentiated cells. These results suggested that the laser treatment did not interfere with proliferation and differentiation processes.

Fig. 7

Assay for gene transcription of cells cultured in PM, or irradiated with three consecutive doses at 3J/cm2 and cultured in PM, or cultured in OM, or irradiated with three consecutive doses at 3J/cm2 and cultured in OM. The indicated reverse transcriptase-polymerase chain reaction (RT-PCR) products were subjected to electrophoresis on 2% agarose gel and visualized by UV exposure. The level of specific bands was normalized for glyceraldehyde-3-phosphate dehydrogenase cDNA.

JBO_18_12_128006_f007.png

Fig. 8

Expression of the indicated bone-specific genes as determined by qRT-PCR. Saos-2 were stimulated for three consecutive days with a dose of 3J/cm2 and then cultured in PM (a) or in OM (b). The graph shows the fold induction of gene expression (arbitrary units), setting the expression of the indicated genes in cells grown in the absence of laser treatment at day 14 of culture. A p<0.05 was considered statistically significant. Data are representative of one of the three experiments performed.

JBO_18_12_128006_f008.png

Fig. 9

Representative images of osteoblast protein markers after laser treatment by confocal laser scanning microscope. Saos-2 were treated for three consecutive days at 3J/cm2 and then cultured in PM [(Aa), (Ab), (Ba), and (Bb)] or in OM [(Ac), (Ad), (Bc), and (Bd)]. Immunofluorescence analysis was performed at day 14 of culture. Coll-1 (A) and osteocalcin (B) (in green), nuclei (in blue) (40× magnification, scale bar represents 50 μm).

JBO_18_12_128006_f009.png

Table 4

Normalized amount of extracellular matrix constituents secreted and deposited by Saos-2 with or without three doses laser of 3  J/cm2 in proliferative medium (PM) or osteogenic medium (OM) after two weeks of cell culture (pg/cell×well). In comparison to unstimulated samples, a p value <0.05 was considered statistically significant (*).

ProteinsPMMultiple doses of 3J/cm2+PMMultiple doses of 3J/cm2+PM/PMOMMultiple doses of 3J/cm2+OMMultiple doses of 3J/cm2+OM/OM
Alkaline phosphatase3.8±0.2337.6±1.062.0*17.8±1.210.7±2.00.6
Decorin11.7±1.5121.4±2.141.8*81.3±3.350.3±8.70.6
Fibronectin3.8±1.1310.7±1.572.8*25.4±2.215.2±1.50.6
Osteocalcin1.5±0.271.8±0.781.26.1±0.73.6±0.050.6
Osteonectin0.9±0.0421.07±0.0361.21.8±0.031.1±0.060.6
Osteopontin9.2±1.7612.2±2.221.330.5±2.019.8±3.10.6
Type-1 collagen11.7±1.2115.3±1.031.340.6±2.824.4±1.30.6
Type-3 collagen16.9±2.0630.16±3.861.8*91.5±3.360.9±11.20.7

Fig. 10

SEM images of Saos-2 cells after laser treatment. Cells were irradiated with three consecutive doses at 3J/cm2 and cultured in PM [(a) and (b)] or in OM [(c) and (d)] at 250× magnification, scale bar represents 10 μm (insert at 1000×, scale bar represents 10 μm).

JBO_18_12_128006_f010.png

4.

Discussion and Conclusions

In this study, we report the results obtained from the application of AlGaInP laser stimuli on adherent human osteoblastic Saos-2 cells. Through a systematic, analytical study, we provide evidence that, besides being able to modulate inflammation and the wound healing process in bone, LLLI is a positive proliferative factor in osteoblast-like cells. Within the parameters assessed in this study, we demonstrated that LLLI (1) enhances the proliferation potential of Saos-2 cells without increasing their tumorigenic characteristics and (2) does not induce morphological damage nor affect their osteoblastic phenotype. In the field of regenerative medicine, tissue engineering offers therapeutic alternatives for autologous bone grafts. The key to successful engineering of bone with optimal restoration of function lies in finding the optimal combination of biomaterial, biofactors, and cells.37 A growing body of evidence shows that different laser systems can lead to enhanced proliferative potential in various cell lines, including stem cells, without compromising their innate characteristics and properties.38 Many investigators have reported that low-level laser treatment positively affects bone regeneration both in vitro and in vivo.3941 Several in vitro experiments were performed to determine the optimum procedure and to elucidate the molecular mechanisms of LLLI. Recently, Yamamoto et al.7 applied diode laser to mouse osteoblast culture and observed that laser treatment accelerated cell proliferation via mouse minichromosome maintenance genes, which are regulators of DNA replication. Moreover, Hirata and his colleagues42 demonstrated that the pro-osteogenic effect of LLLI is related to the stimulation of bone morphogenetic proteins/Smad signaling pathway.

Our study focused on an evaluation of the effects of diode laser treatments on osteoblast-like cell proliferation and bone differentiation. The design of the LLLI protocol used in this study was based on an extensive review of previous data obtained in vitro in bone and rat calvarian cells.21,43 The choice of the model cell line was based on Saos-2 osteoblastic features. To evaluate the effects of LLLI on cell proliferation and differentiation, the Saos-2 cell line was selected as it exhibits several fundamental osteoblast characteristics27 and represents a widely used model for in vitro osteoblast study. An ability to induce the formation of new bone at a specific site would represent a significant advance in bone repair and tissue engineering: this property seems to belong to Saos-2 cells. These osteoblasts uniquely contain an osteoinductive activity, whereas other human osteosarcoma cells, such as U-2 OS, cannot replicate that bone-inducing ability.44 Devitalized Saos-2 cells, extracts, and secretions induced the formation of new bone when implanted subcutaneously in nu/nu mice.26,45 These osteoblasts can be grown, virtually indefinitely, to produce large quantities of osteoinductive factors, such as BOSP and OSN.44 The use of this cell line showed the potential of the LLLI stimulations; nevertheless, by appropriately tuning the LLLI parameters, a better result could be obtained with autologous bone marrow stromal cells instead of Saos-2 osteoblasts for total immunocompatibility with the patient. Initially, we focused on establishing the best irradiation protocol to obtain the maximum proliferation rate. An analysis was performed using red laser wavelength (659 nm) and considering different doses (1 or 3J/cm2) and different irradiation schemes (single irradiation and irradiation repeated for three consecutive days). Proliferative assays showed that an initial effect on cell proliferation was already evident 24 h after the first laser application of 1 or 3J/cm2. However, we found that the effect of a single laser dose on cell proliferation was transitory; in fact, following one laser treatment, after 96 h, no significant differences in proliferation were observed when compared with the dark control. Instead, when samples were irradiated for three consecutive days, the differences in growth rate between controls and treated samples were always statistically significant (p<0.05), suggesting that this treatment was necessary to boost Saos-2 growth. LLLI is reported to stimulate cell proliferation through a wide network of signals. In order to explain our proliferation results, we analyzed the activation of the mitogenic protein Akt, which has been reported to play a key role in proliferation induction by LLLI.31 These data confirmed the involvement of Akt in proliferation of treated cells. In agreement with Zhang et al.,31 we observed, by Western blot analysis, that LLLI is able to induce a transient but strong increase of Akt phosphorylation, which reaches a maximum 20 min after treatment. It was reported that LLLI induces the expression of growth factors, such as insulin-like growth factor-1,46 vascular endothelial growth factor,47 and transforming growth factor-beta.48 We speculate that the relatively prolonged effect on cell proliferation that we observed (up to seven days after multiple laser doses) may be attributed to the growth factors synthesized and secreted by the cells in response to irradiation. After observing that laser treatment promotes in vitro proliferation of these cells, we tested whether this effect was supported by an increase in telomerase activity, whose up regulation is considered to be responsible for the unlimited proliferation of cancer cells.49 In our experiment, laser treatment did not increase the average level of telomerase activity, and these results led us to conclude that this treatment schedule induces proliferation of Saos-2 cells without changing their telomerase expression pattern. In addition, CLSM observations performed on laser-treated/untreated cells showed that irradiated cells did not change their morphology after laser application. After identifying the optimal laser protocol, we studied whether laser treatment influenced osteoblastic differentiation of Saos-2 cells. In most bone tissue engineering studies, bone-promoting factors are used to induce differentiation of Saos-2 along the osteogenic pathway. To screen laser effects on Saos-2 differentiation, we stimulated and cultured cells in the presence/absence of osteoinductive conditioned medium in comparison with unstimulated cultures in the same media conditions used as controls. The cultures were harvested on day 14, representing the almost completely differentiated and mineralized stage of Saos-2 cells. When this cell line is cultured with medium supplemented with osteogenic factors, osteoblasts will differentiate and form a calcified matrix.50 It has been reported that an enhancement in bone formation depends on an increase in extracellular matrix synthesis.51 In this osteogenic condition, Saos-2 cells showed a significant level of bone matrix constituents such as COL-I, COL-III, ALP, OP, OC, OSN, and DEC. In particular, COL-I is the most important and abundant structural protein of the bone matrix;32 DEC is a proteoglycan that is considered to be a key regulator for the assembly and the function of many extracellular matrix proteins with a major role in the lateral growth of the collagen fibrils, delaying the lateral assembly on the surface of the fibrils;33 OP is an extracellular glycosylated bone phosphoprotein secreted at the early stages of the osteogenesis before the onset of the mineralization; it binds calcium, is likely to be involved in the regulation of the hydroxyapatite crystal growth, and through specific interaction with the vitronectin receptor, promotes the attachment of the cells to the matrix;34 OC is secreted after the onset of mineralization, and it binds to bone minerals.35

In this study, we consistently observed that during 14 days of culture in OM, Saos-2 cells exhibited the typical fully differentiated and mineralized stage of osteoblasts with respect to those maintained in PM as documented by ALP activity and calcium deposition. The same features were observed for laser-stimulated cells cultured in OM.

Furthermore, qRT-PCR analysis on osteoblastic differentiation markers showed comparable expression of BOSP, COL-I, and OP, and a slight increase in ALP and OP was visible in laser-treated cells cultured in OM. All together these data suggest that laser stimulation does not block the progression of osteoblast differentiation, as also directly confirmed by the Calcein green staining. More interesting were the results obtained in the unconditioned medium (PM). We demonstrated that when cells were treated with laser in PM, the expression of osteoblast markers, ALP activity, and the mineralization of Saos-2 were significantly enhanced, compared to unstimulated cells in PM (Figs. 5 and 6). Apparently, laser exposition by itself, without adding osteogenic factors, seems to direct cells toward the bone differentiation pathway. An increase in the secretion of proteins specific for differentiation toward bone was detected. An almost twofold increase in ALP production was observed in the laser-stimulated cells in PM (Table 4). It is generally believed that the matrix mineralization is initiated by the expression of membrane-bound glycoprotein ALP on osteoblasts. It has been previously reported that ALP is expressed in large amounts in osteoblasts in vivo36 as well as in vitro differentiation studies with osteoblast-like cell lines.52 The elevated expression of ALP and bone proteins in the samples exposed to multiple laser doses may be attributable to the ability of laser irradiation to accelerate cellular activity, for example, ATP synthesis,53 early osteoblastic differentiation,54 and release of growth factors.55 Our data are in agreement with Dörtbudak et al.56 who reported a marked increase in bone matrix production between days 12 and 16 after three laser irradiation treatments (diode laser, wavelength 690 nm). In contrast to data reported by Coombe et al.,24 with our experimental conditions, a significant increase in ALP activity in the irradiated group as compared with the unexposed control was detected. Unexpectedly, the qRT-PCR results showed no significant difference in the gene expression of ALP between laser-stimulated and unstimulated cells when cultured in medium without osteo-inductive factors for 14 days. Moreover, the late osteoblastic markers OP and BOSP were significantly down regulated. The early bone COL-I marker, which is known to be up regulated at the proliferation stage and down regulated at subsequent stages, was quite similar between stimulated/unstimulated samples. Hypothetically, bone protein transcription was immediately activated after laser exposition, allowing protein translation, and the effect was lost thereafter. On the other hand, protein transcription stopped earlier with respect to protein translation. Further experiments need to be performed to explain these results. In this study, we properly answered the question of whether laser stimulation could positively affect cell proliferation and activate the differentiation process: based on our findings performed in our experimental conditions, we conclude that laser treatment supports, in particular, the in vitro proliferation of Saos-2 cells. However, we also demonstrated that in the absence of osteostimulatory factors, the laser treatment can influence the activation of the osteogenic pathway, as demonstrated by the increased calcified bone extracellular matrix deposition.

On the other side, we showed that the cell differentiation due to laser exposition and in the presence of chemical osteostimulatory factors was not improved: the differentiation process was not significantly incremented when compared to unexposed cells cultivated in the presence of osteogenic factors. Our observations are in accordance with those of Ozawa and colleagues who suggested that a beneficial laser effect on bone could be achieved with a number of applications, but not after only a single application using the same parameters.39

In conclusion, our results demonstrate the effectiveness of laser treatment in modulating cellular functions in vitro. Saos-2 cells cannot be utilized in clinical applications, but they can be used as an in vitro model to further investigate the cellular mechanisms underlying laser treatment, which are as yet unknown. We further propose that this experiment design be utilized to stimulate the conversion of bone marrow mesenchymal cells to the osteogenic phenotype; the resulting data could lead to a potential application in regenerative medicine.

Acknowledgments

We would like to acknowledge financial support from the Compagnia S. Paolo (Genova) Grant titled “Uso di cellule staminali mesenchimali sottoposte a campo elettromagnetico: un approccio innovativo per l’osteointegrazione di impianti di titanio” (2011). M.G.C.De A. also thanks the INAIL project “Malattie professionali e infortuni” titled “Effetti dei campi elettromagnetici sulla salute umana: modelli sperimentali in vitro” (2010). We are grateful to P. Vaghi (Centro Grandi Strumenti, University of Pavia, Pavia, Italy) and D. Picenoni (Politecnico di Milano, Milano, Italy) for technical assistance in the confocal laser scanning microscope and SEM studies, respectively. A special thanks to Natascia Brondino for helping us in statistical analysis and Laurene Kelly for correcting the English of the manuscript.

References

1. 

R. H. FunkT. MonseesN. Ozkucur, “Electromagnetic effects—from cell biology to medicine,” Prog. Histochem. Cytochem., 43 (4), 177 –264 (2009). http://dx.doi.org/10.1016/j.proghi.2008.07.001 PHCCAS 0079-6336 Google Scholar

2. 

E. Sainoet al., “In vitro calcified matrix deposition by human osteoblasts onto a zinc-containing bioactive glass,” Eur. Cell. Mater., 21 (14), 59 –72 (2011). ECMUBB 1473-2262 Google Scholar

3. 

N. D. SearbyC. SteeleR. K. Globus, “Influence of increased mechanical loading by hypergravity on the microtubule cytoskeleton and prostaglandin E2 release in primary osteoblasts,” Am. J. Physiol. Cell. Physiol., 289 (1), 148 –158 (2005). http://dx.doi.org/10.1152/ajpcell.00524.2003 0363-6143 Google Scholar

4. 

E. MesterA. F. MesterA. Mester, “The biomedical effects of laser application,” Lasers Surg. Med., 5 (1), 31 –39 (1985). http://dx.doi.org/10.1002/(ISSN)1096-9101 LSMEDI 0196-8092 Google Scholar

5. 

L. J. Walsh, “The current status of low level laser therapy in dentistry. Part 1. Soft tissue applications,” Aust. Dent. J., 42 (4), 247 –254 (1997). http://dx.doi.org/10.1111/j.1834-7819.1997.tb00129.x ADEJA2 0045-0421 Google Scholar

6. 

A. N. Pereiraet al., “Effect of low-power laser irradiation on cell growth and procollagen synthesis of cultured fibroblasts,” Lasers Surg. Med., 31 (4), 263 –267 (2002). http://dx.doi.org/10.1002/(ISSN)1096-9101 LSMEDI 0196-8092 Google Scholar

7. 

M. Yamamotoet al., “Stimulation of MCM3 gene expression in osteoblast by low level laser irradiation,” Lasers Med. Sci., 16 (3), 213 –217 (2001). http://dx.doi.org/10.1007/PL00011357 LMSCEZ 1435-604X Google Scholar

8. 

Y. Morimotoet al., “Effect of low-intensity argon laser irradiation on mitochondrial respiration,” Lasers Surg. Med., 15 (2), 191 –199 (1994). http://dx.doi.org/10.1002/(ISSN)1096-9101 LSMEDI 0196-8092 Google Scholar

9. 

J. A. de VilliersN. N. HoureldH. Abrahamse, “Influence of low intensity laser irradiation on isolated human adipose derived stem cells over 72 hours and their differentiation potential into smooth muscle cells using retinoic acid,” Stem Cell, 7 (4), 869 –882 (2011). http://dx.doi.org/10.1007/s12015-011-9244-8 STCEDI 1066-5099 Google Scholar

10. 

T. Fushimiet al., “Green light emitting diodes accelerate wound healing: characterization of the effect and its molecular basis in vitro and in vivo,” Wound Repair Regen., 20 (2), 226 –235 (2012). http://dx.doi.org/10.1111/wrr.2012.20.issue-2 1067-1927 Google Scholar

11. 

B. MvulaT. J. MooreH. Abrahamse, “Effect of low-level laser irradiation and epidermal growth factor on adult human adipose-derived stem cells,” Lasers Med. Sci., 25 (1), 33 –39 (2010). http://dx.doi.org/10.1007/s10103-008-0636-1 LMSCEZ 1435-604X Google Scholar

12. 

D. H. HawkinsH. Abrahamse, “The role of laser fluence in cell viability, proliferation, and membrane integrity of wounded human skin fibroblasts following helium-neon laser irradiation,” Lasers Surg. Med., 38 (1), 74 –83 (2006). http://dx.doi.org/10.1002/(ISSN)1096-9101 LSMEDI 0196-8092 Google Scholar

13. 

I. Saygunet al., “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., 30 (3), 149 –154 (2012). http://dx.doi.org/10.1089/pho.2011.3079 PLDHA8 1549-5418 Google Scholar

14. 

C. H. ChenH. S. HungS. H. Hsu, “Low-energy laser irradiation increases endothelial cell proliferation, migration, and eNOS gene expression possibly via PI3K signal pathway,” Lasers Surg. Med., 40 (1), 46 –54 (2008). http://dx.doi.org/10.1002/(ISSN)1096-9101 LSMEDI 0196-8092 Google Scholar

15. 

T. N. Demidova-Riceet al., “Low-level light stimulates excisional wound healing in mice,” Lasers Surg. Med., 39 (9), 706 –715 (2007). http://dx.doi.org/10.1002/(ISSN)1096-9101 LSMEDI 0196-8092 Google Scholar

16. 

T. I. KaruS. F. Kolyakov, “Exact action spectra for cellular responses relevant to phototherapy,” Photomed. Laser Surg., 23 (4), 355 –361 (2005). http://dx.doi.org/10.1089/pho.2005.23.355 PLDHA8 1549-5418 Google Scholar

17. 

D. Pastoreet al., “Increase in b-H+/e- ratio of the cytochrome c oxidase reaction in mitochondria irradiated with helium-neon laser,” Biochem. Mol. Biol. Int., 34 (4), 817 –826 (1994). BMBIES 1039-9712 Google Scholar

18. 

J. Pouysseguret al., “Cytoplasmic pH, a key determinant of growth factor-induced DNA synthesis in quiescent fibroblasts,” FEBS Lett., 190 (1), 115 –119 (1985). http://dx.doi.org/10.1016/0014-5793(85)80439-7 FEBLAL 0014-5793 Google Scholar

19. 

H. TubyL. MaltzU. Oron, “Low-level laser irradiation (LLLI) promotes proliferation of mesenchymal and cardiac stem cells in culture,” Lasers Surg. Med., 39 (4), 373 –378 (2007). http://dx.doi.org/10.1002/(ISSN)1096-9101 LSMEDI 0196-8092 Google Scholar

20. 

M. A. TrellesE. Mayayo, “Bone fracture consolidates faster with low power laser,” Lasers Surg. Med., 7 (1), 36 –45 (1987). http://dx.doi.org/10.1002/(ISSN)1096-9101 LSMEDI 0196-8092 Google Scholar

21. 

E. Steinet al., “Initial effects of low-level laser therapy on growth and differentiation of human osteoblast-like cells,” Wien Klin Wochenschr, 120 (3–4), 112 –117 (2008). http://dx.doi.org/10.1007/s00508-008-0932-6 WKWOAO 0043-5325 Google Scholar

22. 

M. Khadraet al., “Low-level laser therapy stimulates bone-implant interaction: an experimental study in rabbits,” Clin. Oral Implants Res., 15 (3), 325 –332 (2004). http://dx.doi.org/10.1111/clr.2004.15.issue-3 0905-7161 Google Scholar

23. 

W. T. LiY. C. LeuJ. L. Wu, “Red-light light-emitting diode irradiation increases the proliferation and osteogenic differentiation of rat bone marrow mesenchymal stem cells,” Photomed. Laser Surg., 28 (1), 157 –165 (2010). http://dx.doi.org/10.1089/pho.2009.9945 PLDHA8 1549-5418 Google Scholar

24. 

A. R. Coombeet al., “The effects of low level laser irradiation on osteoblastic cells,” Clin. Orthod. Res., 4 (1), 3 –14 (2001). http://dx.doi.org/10.1034/j.1600-0544.2001.040102.x 1397-5927 Google Scholar

25. 

K. M. AlghamdiA. KumarN. A. Moussa, “Low-level laser therapy: a useful technique for enhancing the proliferation of various cultured cells,” Lasers Med. Sci., 27 (1), 237 –249 (2012). http://dx.doi.org/10.1007/s10103-011-0885-2 LMSCEZ 1435-604X Google Scholar

26. 

H. C. Andersonet al., “The mechanism of bone induction and bone healing by human osteosarcoma cell extracts,” Clin. Orthop. Relat. Res., 313 129 –134 (1995). CORTBR 0009-921X Google Scholar

27. 

H. C. Andersonet al., “Bone-inducing agent in Saos-2 cell extracts and secretions,” Cells Mater., 8 89 –99 (1998). CMBLFF 1425-8153 Google Scholar

28. 

N. W. Kimet al., “Specific association of human telomerase activity with immortal cells and cancer,” Science, 266 (5193), 2011 –2015 (1994). http://dx.doi.org/10.1126/science.7605428 SCIEAS 0036-8075 Google Scholar

29. 

E. Sainoet al., “In vitro enhancement of SAOS-2 cell calcified matrix deposition onto radio frequency magnetron sputtered bioglass-coated titanium scaffolds,” Tissue Eng. Part A, 16 (3), 995 –1008 (2010). http://dx.doi.org/10.1089/ten.tea.2009.0051 1937-3341 Google Scholar

30. 

K. J. LivakT. D. Schmittgen, “Analysis of relative gene expression data using real-time quantitative PCR and the 2(-delta delta C(T)) method,” Methods, 25 (4), 402 –408 (2001). http://dx.doi.org/10.1006/meth.2001.1262 MTHDE9 1046-2023 Google Scholar

31. 

L. Zhanget al., “Low-power laser irradiation promotes cell proliferation by activating PI3K/Aktpathway,” J. Cell Physiol., 219 (3), 553 –562 (2009). http://dx.doi.org/10.1002/jcp.v219:3 JCLLAX 0021-9541 Google Scholar

32. 

T. A. Owenet al., “Progressive development of the rat osteoblast phenotype in vitro: reciprocal relationships in expression of genes associated with osteoblast proliferation and differentiation during formation of the bone extracellular matrix,” J. Cell Physiol., 143 (3), 420 –430 (1990). http://dx.doi.org/10.1002/(ISSN)1097-4652 JCLLAX 0021-9541 Google Scholar

33. 

F. Azariet al., “Intracellular precipitation of hydroxyaptite mineral and implications for pathological calcification,” J. Struct. Biol., 162 (3), 468 –479 (2008). http://dx.doi.org/10.1016/j.jsb.2008.03.003 JSBIEM 1047-8477 Google Scholar

34. 

S. Van Dijket al., “Evidence that a non-RGD domain in rat osteopontin is involved in cell attachment,” J. Bone Miner. Res., 8 (12), 1499 –1506 (1993). http://dx.doi.org/10.1002/jbmr.5650081213 JBMREJ 0884-0431 Google Scholar

35. 

J. E. AubinF. Liu, “The osteoblast lineage,” Principles of Bone Biology, 51 –67 Academic Press, San Diego (1996). Google Scholar

36. 

J. ZernickK. TwarogW. B. Upholt, “Regulation of alkaline phosphatase and alfa 2(1) procollagen synthesis during early intramembranous bone formation in the rat mandible,” Differentiation, 44 (3), 207 –215 (1990). http://dx.doi.org/10.1111/dif.1990.44.issue-3 DFFNAW 0301-4681 Google Scholar

37. 

C. Vinatieret al., “Cartilage engineering: a crucial combination of cells, biomaterials and biofactors,” Trends Biotechnol., 27 (5), 307 –314 (2009). http://dx.doi.org/10.1016/j.tibtech.2009.02.005 TRBIDM 0167-7799 Google Scholar

38. 

G. Sheferet al., “Skeletal muscle cell activation by low energy laser irradiation: a role for the MAPK/ERK pathway,” J. Cell Physiol., 187 (1), 73 –80 (2001). http://dx.doi.org/10.1002/(ISSN)1097-4652 JCLLAX 0021-9541 Google Scholar

39. 

Y. Ozawaet al., “Stimulatory effects of low-power laser irradiation on bone formation in vitro,” Proc. SPIE, 330 281 –288 (1995). http://dx.doi.org/10.1117/12.207040 PSISDG 0277-786X Google Scholar

40. 

L. Abramovitch-Gottlibet al., “Low-level laser irradiation stimulates osteogenic phenotype of mesenchymal stem cells seeded on a three-dimensional biomatrix,” Lasers Med. Sci., 20 (3–4), 138 –146 (2005). http://dx.doi.org/10.1007/s10103-005-0355-9 LMSCEZ 1435-604X Google Scholar

41. 

M. Soleimaniet al., “The effects of low-level laser irradiation on differentiation and proliferation of human bone marrow mesenchymal stem cells into neurons and osteoblasts—an in vitro study,” Lasers Med. Sci., 27 (2), 423 –430 (2012). http://dx.doi.org/10.1007/s10103-011-0930-1 LMSCEZ 1435-604X Google Scholar

42. 

S. Hirataet al., “Low-level laser irradiation enhances BMP-induced osteoblast differentiation by stimulating the BMP/Smad signaling,” J. Cell Biochem., 111 (6), 1445 –1452 (2010). http://dx.doi.org/10.1002/jcb.22872 JCEBD5 0730-2312 Google Scholar

43. 

Y. Ozawaet al., “Low-energy laser irradiation stimulated bone nodule formation at early stages of cell cultured in rat calvarial cell,” Bone, 22 (4), 347 –354 (1998). http://dx.doi.org/10.1016/S8756-3282(97)00294-9 BONEDL 8756-3282 Google Scholar

44. 

H. C. Andersonet al., “Selective synthesis of bone morphogenetic proteins-1, -3, -4 and bone sialoprotein may be important for osteoinduction by SAOS-2 cells,” J. Bone Miner. Metab., 20 (2), 73 –82 (2002). http://dx.doi.org/10.1007/s007740200010 JBMME4 0914-8779 Google Scholar

45. 

H. C. Andersonet al., “Bone-inducing agent (BIA) from cultured human SAOS-2 osteosarcoma cells,” Bone Miner., 16 (1), 49 –62 (1992). http://dx.doi.org/10.1016/0169-6009(92)90821-T BOMIET 0169-6009 Google Scholar

46. 

I. Saygunet al., “Effects of laser irradiation on the release of basic fibroblast growth factor (bFGF), insulin like growth factor-1 (IGF-1), and receptor of IGF-1 (IGFBP3) from gingival fibroblasts,” Lasers Med. Sci., 23 (2), 211 –215 (2008). http://dx.doi.org/10.1007/s10103-007-0477-3 LMSCEZ 1435-604X Google Scholar

47. 

H. TubyL. MaltzU. Oron, “Modulations of VEGF and iNOS in the rat heart by low level laser therapy are associated with cardioprotection and enhanced angiogenesis,” Lasers Surg. Med., 38 (7), 682 –688 (2006). http://dx.doi.org/10.1002/(ISSN)1096-9101 LSMEDI 0196-8092 Google Scholar

48. 

S. M. Safaviet al., “Effects of low-level He-Ne laser irradiation on the gene expression of IL-1beta, TNF-alpha, IFN-gamma, TGF-beta, bFGF, and PDGF in rat’s gingival,” Lasers Med. Sci., 23 (3), 331 –335 (2008). http://dx.doi.org/10.1007/s10103-007-0491-5 LMSCEZ 1435-604X Google Scholar

49. 

H. VaziriS. Benchimol, “From telomere loss to p53 induction and activation of a DNA-damage pathway at senescence: the telomere loss/DNA damage model of cell aging,” Exp. Gerontol., 31 (1–2), 295 –301 (1996). http://dx.doi.org/10.1016/0531-5565(95)02025-X EXGEAB 0531-5565 Google Scholar

50. 

S. B. Rodanet al., “Characterization of a human osteosarcoma cell line (SAOS-2) with osteoblastic properties,” Cancer Res., 47 (18), 4961 –4966 (1987). CNREA8 0008-5472 Google Scholar

51. 

S. C. Manolagas, “Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis,” Endocr. Rev., 21 (2), 115 –137 (2000). http://dx.doi.org/10.1210/er.21.2.115 ERVIDP 0163-769X Google Scholar

52. 

L. C. Gerstenfeldet al., “Expression of differentiated function by mineralizing cultures of chicken osteoblasts,” Dev. Biol., 122 (1), 49 –60 (1987). http://dx.doi.org/10.1016/0012-1606(87)90331-9 DEBIAO 0012-1606 Google Scholar

53. 

T. KaruL. PyatibratG. Kalendo, “Irradiation with He-Ne laser increases ATP level in cells cultivated in vitro,” J. Photochem. Photobiol. B, 27 (3), 219 –223 (1995). http://dx.doi.org/10.1016/1011-1344(94)07078-3 JPPBEG 1011-1344 Google Scholar

54. 

R. Lubartet al., “Effects of visible and near-infrared lasers and cell cultures,” J. Photochem. Photobiol. B., 12 (3), 305 –310 (1992). http://dx.doi.org/10.1016/1011-1344(92)85032-P JPPBEG 1011-1344 Google Scholar

55. 

W. YuJ. O. NaimR. J. Lanzafame, “The effect of laser irradiation on the release of bFGF from 3T3 fibroblasts,” Photochem. Photobiol., 59 (2), 167 –170 (1994). http://dx.doi.org/10.1111/php.1994.59.issue-2 PHCBAP 0031-8655 Google Scholar

56. 

O. DörtbudakR. HaasG. Mailath-Pokorny, “Effect of low-power laser irradiation on bony implant sites,” Clin. Oral Implants Res., 13 (3), 288 –292 (2002). http://dx.doi.org/10.1034/j.1600-0501.2002.130308.x 0905-7161 Google Scholar
© 2013 Society of Photo-Optical Instrumentation Engineers (SPIE) 0091-3286/2013/$25.00 © 2013 SPIE
Nora Bloise, Gabriele Ceccarelli, Paolo Minzioni, Marco Vercellino, Laura Benedetti, Maria Gabriella C. De Angelis, Marcello Imbriani, and Livia Visai "Investigation of low-level laser therapy potentiality on proliferation and differentiation of human osteoblast-like cells in the absence/presence of osteogenic factors," Journal of Biomedical Optics 18(12), 128006 (23 December 2013). https://doi.org/10.1117/1.JBO.18.12.128006
Published: 23 December 2013
Lens.org Logo
CITATIONS
Cited by 48 scholarly publications.
Advertisement
Advertisement
RIGHTS & PERMISSIONS
Get copyright permission  Get copyright permission on Copyright Marketplace
KEYWORDS
Bone

Laser therapeutics

Phase modulation

Proteins

Aluminium phosphide

Calcium

Laser irradiation

Back to Top