Open Access
11 December 2018 Review of light parameters and photobiomodulation efficacy: dive into complexity
Author Affiliations +
Abstract
Photobiomodulation (PBM) therapy, previously known as low-level laser therapy, was discovered more than 50 years ago, yet there is still no agreement on the parameters and protocols for its clinical application. Some groups have recommended the use of a power density less than 100  mW/cm2 and an energy density of 4 to 10  J/cm2 at the level of the target tissue. Others recommend as much as 50  J/cm2 at the tissue surface. The wide range of parameters that can be applied (wavelength, energy, fluence, power, irradiance, pulse mode, treatment duration, and repetition) in some cases has led to contradictory results. In our review, we attempt to evaluate the range of effective and ineffective parameters in PBM. Studies in vitro with cultured cells or in vivo with different tissues were divided into those with higher numbers of mitochondria (muscle, brain, heart, nerve) or lower numbers of mitochondria (skin, tendon, cartilage). Graphs were plotted of energy density against power density. Although the results showed a high degree of variability, cells/tissues with high numbers of mitochondria tended to respond to lower doses of light than those with lower number of mitochondria. Ineffective studies in cells with high mitochondrial activity appeared to be more often due to over-dosing than to under-dosing.

1.

Introduction

Since Mester,1,2 in 1968, accidently discovered the positive effect of a ruby laser beam on hair growth and wound healing in mice, researchers have attempted to uncover the scientific basis for this phenomenon as well to establish the range of optical exposure parameters that lead to successful clinical outcomes. The possibility of stimulating a wide range of cells to improve wound healing and cellular growth has created a science referred to as low-level laser therapy (LLLT) or photobiomodulation therapy (PBMT). As an understanding of basic concepts has emerged, the very wide range of factors contributing to positive outcomes in some cases and negative outcomes in others has stymied the development of definitive protocols.

The multitude of variables to be considered is formidable. More than 1000 research articles have reported that a range of factors can apparently affect the chances of success including wavelength, energy density, power density, total energy, total power, pulse structure, spot size, tissue absorption characteristics, and treatment repetition regimen. Further parameters of lesser importance requiring both control and study are use of combination wavelengths, delivery method (contact, punctual, broad beam), duration of treatment, inadvertent heating of tissue and even whether the source of photons is a laser, light-emitting diode (LED), or broad-spectrum light from a lamp.3,4

It has become apparent that, in order to achieve positive results with PBM, each of these dosimetric parameters must be controlled within a limited range of values. Of the many studies that have been conducted over the past 50 years, a number have attempted to determine the relative contribution of individual parameters to successful outcomes.

Consensus has (almost) been reached on one of the most important concepts in PBM. The so-called Arndt–Schultz law was originally proposed near the end of the 19th century. It states in original form that “For every substance, small doses stimulate, moderate doses inhibit, and large doses kill.”5 This concept6 also forms the basis of the science of “hormesis,” as reviewed by Calabrese and Mattson7

Pharmacological agents used at a therapeutic dose can be very beneficial while the same drug administered at a higher dose may be catastrophic. For many years, this Arndt–Schultz law has been used as a convenient concept to explain the cellular and tissue interactions with light.

Briefly, this law, when applied to PBM, states that, at very low levels of irradiation, photons are absorbed by subcellular chromophores present inside intracellular organelles, most notably, mitochondria. Absorption of energy by cytochrome C oxidase (CCO) in the mitochondrial respiratory chain is the primary initiating interaction triggering PBM effects.8 Both adenosine triphosphate (ATP) production and oxygen consumption by the cells increase. This may lead to changes in nitric oxide (NO) levels, activation of secondary messenger pathways, activation of transcription factors, and growth factor production.9 At this very low level, energy is absorbed by the cell but at such low amounts of energy that there are no observable gross changes (temperature or photochemical damage).

As the number of absorbed photons increases, stimulation of cellular metabolism, as noted above, begins to affect cellular activity, producing positive PBM effects. Both the number of photons and rate at which they are delivered has a significant influence on the response.10,11

As the number of photons increases beyond a particular level, the cellular stimulation disappears, and if the number of photons is even further increased, inhibition and cellular damage occurs. Current theories suggest that the mitochondrial membrane potential having reached a maximum at the optimum dose declines back to baseline and can be lowered below baseline by excessive doses of light.12 ATP reserves within the cell begin to be depleted by excessive doses of light compromising the positive cellular function. Production of excessive reactive oxygen species (ROS), which can be toxic, release of excessive free NO, which can damage cells, and activation of a cytotoxic mitochondrial-signaling pathway leading to apoptosis are also possible theories. At still higher levels of irradiation, depletion of cellular energy reserves or excess levels of the factors noted above become so significant that cellular metabolism falls below normal intrinsic levels and function is actually inhibited eventually leading to cell death.

This concept, represented by the Arndt–Schultz law of biphasic dose response, has become the foundational concept of PBM. However, the appropriate range of values of fluence and irradiance at which these significant transitions occur are not widely agreed upon. Numerous studies suggest that fluences ranging from 3 to 10  J/cm2, at the cellular level, will produce the desired stimulation of metabolic activity.13,14

While this protocol has become widely accepted, some studies suggest that biostimulation will occur in the range of 0.5 to 1  J/cm2 on an open wound and in the range of 2 to 4  J/cm2 to a target through overlaying skin.15 Another respected source suggests that doses used for superficial targets tend to be in the region of 4  J/cm2 with a range of 1 to 10  J/cm2.1618 Doses for deeper-seated targets should be in the 10 to 50/cm2 range.1921

While many studies have shown a positive effect of PBM,17,18,21 a number have failed to show a benefit22,23 and, in fact, some reports have shown negative outcomes at what are reported to be the same parameters of irradiation as other positive studies. Unfortunately, in many of the historical studies, important laser parameters were omitted or incorrectly presented.

Often, laser output total power is reported without consideration of the spot diameter at the surface of the target tissue. Therefore, power density, the most relevant parameter, is not reported and results are, predictably, inconsistent.

Sometimes the distribution of energy across the tissue surface is not noted in published studies introducing profound errors. As an example, most lasers are designed to emit in the TEM00 mode, which produces a Gaussian distribution of beam profile. By mathematical definition, cells in the exact center of the beam will be irradiated at precisely twice the indicated average output power while cells at the periphery of the irradiation spot will only receive about 13% of that power. If irradiation were to be delivered for 30 s, cells at the beam center would receive an energy dose of 6  J/cm2 while those at the periphery would receive 0.39  J/cm2. Obviously, the cellular response, taking into account the Arndt–Schultz law, will be different in each of these tissues. This could result in a conclusion of no-effect, positive effect, or negative effect, depending on which cells were observed in the analysis phase of the study.

Another basic concept that has been suggested to be relevant to the successful application of PBM is the Roscoe–Bunsen law of reciprocity.24 This concept states that the most important parameter in PBM is the total quantity of photons absorbed by the target cells, and it is not important how quickly or how slowly these photons are delivered. This means that 100  mW/cm2 applied for 60 s for a dose of 6  J/cm2 will have the same effect as applying 1  W/cm2 for 6 s (6  J/cm2) or 6  W/cm2 for 1 s (6  J/cm2) using the same spot size.

Numerous studies have shown that, while this law is valuable for many parts of the parameter range, it does not hold true for the entire range.18,25,26 The previously discussed theories of the biphasic dose response, supported by other studies, are the likely reason for this inaccuracy. Within a certain range of parameters, perhaps between 1 and 100  J/cm2, and at power densities from 1 to 100  mW/cm2, this linear reciprocity applies. However, beyond this range, reciprocity does not appear to apply. For instance, there exists a lower threshold (perhaps 0.5  mW/cm2) below which the illumination time could be infinite and would be no different from daylight. Similarly, the upper threshold is fixed by the possible photothermal effect if the power density is too large. The irradiance values, that produce unacceptable heating of the tissue, are governed by the wavelength and are 750  mW/cm2 at 800 to 900 nm, about 300  mW/cm2 at 600 to 700 nm, and as low as 100  mW/cm2 at 400 to 500 nm. Furthermore, the illumination time is also important.17 There exists a certain minimum length of time (few minutes) that the light needs to be on the tissue for the best effects to occur.17

The parameters of most importance in PBM are the power density (irradiance) measured in mW/cm2 and the energy density (fluence) measured in J/cm2. Many of the studies discussed here and, indeed, in most of the research literature, are based on the inaccurate statement of the laser output in Watts. Depending on the area irradiated by this beam of photons, the power density and the cellular effects produced will be very different.

As an example, 1 W delivered through a 400  μm diameter optical fiber will produce a power density of 796  W/cm2 while the same 1 W delivered through an 8-mm diameter therapy hand-piece will produce a power density of only 2  W/cm2.

Energy density is frequently reported in research literature but the spot area at the tissue is often omitted. This error makes it impossible to verify their findings or to see how they calculated the vital energy density information. Inconsistency in reporting these parameters is a major source of contradictory research findings and has done much to hinder the acceptance of PBM effects.

Another important factor that must be taken into account is the optical properties of the tissue itself.27 Since the light is generally delivered as a surface spot shone onto the skin, the number of photons that actually penetrate into the tissue to arrive at the pathological lesion is highly variable.28 The first issue to be addressed is light reflection from the surface of the skin,29 which can be minimized if the optical probe is held in firm contact with the skin.30 The second issue is scattering of light within tissue. Scattering is wavelength dependent with shorter wavelengths undergoing more intense Mie scattering than longer wavelengths.31 The third issue is absorption of the light by chromophores that are not biologically active. These nonactive chromophores are chiefly hemoglobin (both oxyhemoglobin and deoxyhemoglobin), myoglobin, and melanin.27 However, it should be noted that some authors have suggested that photodissociation of oxygen from hemoglobin32 or NO from myoglobin33 could be a relevant mechanism in PBM. There is a growing trend for researchers in PBM to undertake modeling of tissue optical properties either by Monte-Carlo methods34 or by use of tissue phantoms.35

1.1.

Mitochondria and Cells

Mitochondria are highly important intracellular organelles whose main function is to act as “power plant” of the cell, generating ATP which is the main source energy for cellular activity and metabolism. Moreover, mitochondria play important roles in regulation of oxidative stress, calcium metabolism, apoptosis, and a host of signaling pathways.36 It is believed that mitochondria originated when a primitive eukaryotic cell “captured” a primitive prokaryotic bacterium around the time the “great oxygenation event” occurred on the Earth.37

Mitochondria contain the electron transport chain responsible for transferring electrons from NADH through complexes I, II, III, and IV.38

When applying light to cells, mitochondria are the initial sites of light absorption and CCO (particularly, the CuA and CuB metal centers) are believed to be the photoacceptors.39 Photon absorption results in setting in motion a cascade of reactions known as cellular signaling pathways leading to NO dissociation, ROS production, and increased ATP synthesis.9

The number of mitochondria in cells varies widely and it is strongly correlated with the metabolic requirements of the cell (how many chemical reactions the cell has to carry out) and may range from a few to thousands of individual organelles. Cells such as osteoblasts, keratinocytes, and fibroblasts have a lower number of mitochondria, whereas muscle cells, neural cells, cells composing internal organs (liver, kidneys, spleen, etc.), and myocardial cells contain a higher number of mitochondria. Broadly speaking, the proportion of mitochondria in a tissue type can be gauged by observing the color of the tissue (without containing any blood). For instance, dark colored tissues (liver, heart, kidney, gray brain matter) have a high concentration of mitochondria since CCO and other cytochromes are the most important cellular pigments, while light colored tissues (skin, bones, tendons) have few mitochondria. The following reports discuss how mitochondrial numbers and mitochondrial activity have been determined in different cells and tissues.4043

Furthermore, mitochondria in stem cells and induced pluripotent stem cells are poorly developed and low in number; mitochondrial function and structure have even been suggested as indicators of stem cell competence.44

The hypothesis of the present review is that the effects of PBM on different tissues can be explained by taking into account two main factors. First, what is the content of mitochondria in the cells comprising the bulk of the tissue? Second, what is the depth? Cells in vitro are very superficial, skin and some connective tissues are moderately superficial, while other tissues are deeper, bones, joints, brain, organs, etc. Moreover, tissues with high mitochondrial numbers tend to be deeper than those with low mitochondrial numbers.

Therefore, studies were divided into two groups based on the number of mitochondria at the cellular level and the depth of the tissue level.

Cells of tissues with higher numbers of mitochondria were assembled in one group (brain cells, muscle cells, neural cells, macrophages, monocytes) and cells with fewer mitochondria were assembled in another group (keratinocytes, osteoblasts, chondrocytes, fibroblasts, stem cells). Tissues with abundant mitochondria exist in organs, such as muscle, heart, liver, kidney, cells.

The purpose of this review paper was to compare effective and ineffective studies on cells and tissues in each group. Every effort was made to find or calculate relevant parameters even if they were not explicitly stated in the paper.

2.

Materials and Methods

This study was conducted following Preferred Reporting Items of Systematic reviews and Meta-analysis.

Research questions: Is it possible to propose a practical protocol of for PBM or LLLT? What are the best parameters that produce a positive result in different circumstances?

2.1.

Research Strategy for Article Identification

Research was conducted using the following electronic databases: Springer, PubMed, Google Scholar, and Cochrane Database.

Keywords used: LLLT, PBM, LLLT and osseointegration, LLLT and bone graft, LLLT and cells, LLLT and bone regeneration.

After collecting the data, the titles, abstract, and conclusions were checked and unrelated, and obviously biased articles were excluded. Also, all case reports and literature reviews were excluded. Only studies dated from 2007 to 2016 were included.

Evaluations of articles were independently performed by two reviewers. The initial search yielded 250 articles. After exclusion of unrelated articles, only 190 remained. Using the exclusion criteria listed in Table 1 reduced this number to 34 articles.

Table 1

Eligibility criteria adapted from Cericato et al.45 for selection of the studies.

Reason for exclusionPubMedSpringerGoogle ScholarCochraneTotal
Literature and/or systematic review8811633
Article in language other than English1515
Letter from the editor, opinion articles88
Fluence not mentioned3230843
Use of very high fluence: density greater than 500  J/cm2428822
Article did not mention power or fluence rate5216629
Other (book chapter, appendix, bibliograghy, index426
Total exclusion20149230156

2.2.

Assessment of the Studies

After obtaining full texts of all 34 relevant articles, they were evaluated and scored following the checklist using eligibility criteria adapted from Cericato et al.45 described in Table 1. Articles with scores from 0 to 8 points were considered low quality and were excluded. Article with scores from 13 to 15 points were considered high quality while scores from 9 to 12 were considered moderate quality. Table 2 presents the details of the 34 studies finally included in this review.

Table 2

Final list of studies that were included together with Cericato score.

AuthorsScore (Cericato et al.)17
Fernandes et al.4612
Mendez et al.2112
Barbosa et al.2011
Huang et al.4711
Huang et al.4812
Sharma et al.4911
Oron et al.5010
Chen et al.2612
Souza et al.5111
Ferraresi et al.5212
Zhang et al.5312
Wang et al.5412
Amaroli1910
Tschon et al.5511
Pyo et al.5612
Migliario et al.5712
Khadra et al.3211
Skopin et al.5812
Salehpour et al.5911
Wu et al.5812
Lopes-Martins et al.1811
Bozkurt et al.6012
Wang et al.6111
Alves et al.2511
Oron et al.62,6311
Castano et al.1712
Salehpour et al.6411
Leal junior et al.65,6612
Ando et al.1311
Zhang et al.6712
Baroni et al.6811
Leal Junior et al.6911
Blanco et al.7012
Disner et al.7112

3.

Effect of Varying a single parameter on PBM Efficacy

3.1.

I-Effect of varying wavelength on PBM Efficacy

3.1.1.

In vitro studies

It has been shown through many studies that CCO is the most important chromophore that absorbs light. Delpy and Cope72 showed that over 50% of the light absorption between 800 and 850 nm was due to cytochrome c oxidase, with hemoglobin (oxy and deoxy) playing a minor role. CCO has two absorption bands, one in the red spectral region (660  nm) and another in the NIR spectrum (800  nm), which consequently are the wavelengths most often used in PBM3.

In their study, Wang et al.54 found that the mechanisms of action of 810 and 980 nm laser appeared to have significant differences. While the PBM effect occurred at both wavelengths, the chromophore was different between wavelengths. NIR wavelengths, such as 810 nm, stimulate mitochondrial activity and ATP production. At longer wavelengths, the mechanism of action of 980 nm relies on absorption by water leading to the activation of heat (or light)-gated ion channels and promotes cell proliferation via the TRPV1 calcium ion channel pathway.

The same study compared the effect on stem cell differentiation of these two different wavelengths, 810 and 980 nm. For each wavelength, different doses were used from 0.03 to 10  J/cm2, spot size 4  cm2, irradiance 16  mW/cm2, power 64 mW, and time of irradiance (3  J/cm2, 188 s) and (0.3  J/cm2, 18.8 s). The irradiance was adjusted by varying the distance between the laser and the target cells.

Both wavelengths showed a biphasic dose response. At 980 nm, a peak dose response was seen at 0.03 and 0.3  J/cm2 while 810 nm showed a peak response at 3  J/cm2. Moreover, the dose of 0.3  J/cm2 with the 980-nm laser had a better effect than any of the other groups.

A second study by Wang compared the effects of delivering four different wavelengths (420, 540, 660, and 810 nm) using the same parameters of 3  J/cm2 at 16  mW/cm2, on human adipose-derived stem cell differentiation into osteoblasts. They found that 420- and 540-nm wavelengths were more effective in stimulating osteoblast differentiation compared to 660 and 810 nm. Intracellular calcium was higher after 420 and 540 nm and could be inhibited by the TRP channel inhibitors, capsazepine and SKF96365. They concluded that using blue and green wavelengths activated the light-gated calcium channels rather than CCO.61,73

3.1.2.

In vivo studies

Mendez et al.21 compared, histologically, the effect of using two different wavelengths (GaAlAs 830 nm and InGaAl 685 nm) on repair of cutaneous wounds in rats. The control group received no treatment; group II was irradiated with 685 nm, using a fluence of 20  J/cm2 with a spot diameter of 0.6 mm; group III was irradiated using 830 nm, 20  J/cm2; group IV was irradiated with both 830 and 685 nm using a total of 20  J/cm2; group V with 830 nm using 50  J/cm2; group VI with 685 nm, 50  J/cm2 and group VII using 830 and 685 nm, 50  J/cm2. Laser therapy was repeated four times over 7 days at 48 h intervals. They concluded that better results were observed when combining both wavelengths of 830 and 685 nm and attributed this advantage to different absorption and penetration. When comparing the two wavelengths used separately, 830 nm showed better results. While combining the wavelengths provides valuable information, it was not appropriate to include it in the tables of effectiveness.

Barbosa et al.20 compared the effect of light application on bone healing in rats using red and infrared wavelengths. Forty-five rats were divided into three groups after femoral osteotomy: Gr I was used as control; Gr II was submitted to laser treatment using a red wavelength (660 to 690 nm); and Gr III were treated using an infrared laser (790 to 830 nm). Laser therapy was applied immediately after osteotomy and repeated every 48 h, three times a week, for a total of nine sessions over 21 days. The output power was set at 100 mW, energy 4 J, spot size 0.028  cm2, power density 3.5  W/cm2 for 40 s producing a fluence of 140  J/cm2. Animals were sacrificed, the femurs removed and subjected to optical densitometry analysis after 7, 14, and 21 days (five per group).20 After 7 days, both laser-treated groups had significantly higher mean bone optical density compared with the control group but no significant difference between the two laser groups was seen. After 14 days, only Gr III treated with infrared energy showed significantly higher bone density than the control group. After 21 days, no significant difference of the mean bone density between the three groups was seen. They concluded that PBM accelerated bone repair in the initial phase and suggested that PBM in bone repair is both timing and wavelength dependent.

Al-Watban and Zhang16 compared the efficacy of accelerating wound healing in diabetic rats using visible and NIR diode lasers at wavelengths of: 532, 633, 670, 810, and 980 nm. Each wavelength was delivered at doses of 5, 10, 20 and 30  J/cm2, using the same power density for all the wavelength of 22  mW/cm2 except for 633 nm (irradiance used: 15.5  mW/cm2) and 532 nm (10  mW/cm2). Results showed that there was a significant difference between the NIR and visible wavelengths with visible wavelengths being more effective than NIR. They also concluded that the optimum wavelength was 633 nm and the optimum dose was 10  J/cm2.

These studies suggest that the relationship between wavelength and fluence is crucial. If the target is CCO, it is well accepted that red light (630 to 670 nm) or near-infrared light (780 to 940 nm) will have positive effects, using fluences in the stimulatory range of 3 to 10  J/cm2.16

However, if the desired chromophore is ion channels within cells, the wavelengths that best affect the calcium channels are in the range of 420 to 540 nm.54,61 Delivering just 3  J/cm2 when using 16  mW/cm2 will have the best effect. Using the higher wavelength of 980 nm may also have a beneficial effect for targeting water as a chromophore.54

Disner et al.71 studied the effect of PBMT delivered to the head (over right prefrontal cortex) combined with attention bias modification (ABM) therapy on 51 human patients with elevated symptom of depression. PBMT was administered before and after blocks of ABM using 1064 nm, 3.4 W, irradiance of 250  mW/cm2 (3,400  mW/13.6  cm2=250  mW/cm2) for 4 min and a cumulative fluence of 60  J/cm2 (0.25  W/cm2×240  s=60  J/cm2). They found that PBMT led to greater symptom improvement especially among participants, whose attention span was responsive to ABM, and they concluded that the beneficial effect of ABM could be improved by adjunctive interventions, such as right prefrontal PBMT.

3.2.

II-Effect of Varying Energy Density and Power Density on PBM Efficiency

3.2.1.

In vitro studies with cells with high number of mitochondria

Fernandez et al.46 stimulated the M1 profile (macrophages can have two different phenotypes called M1 and M2 depending on the type of cytokines they produce) of macrophages by using two different sets of laser parameters: 660 nm, 15 mW, 0.375  W/cm2, 20 s for 7.5  J/cm2 and 780 nm, 70 mW, 1.75  W/cm2, 1.5 s for 2.6  J/cm2 (the spot area calculated by current authors from available information was 0.04  cm2). Results showed that both lasers were able to decrease TNFα and iNOS expression but parameters used for 780 nm gave an additional decrease. Also, parameters used for 660 nm gave an up-regulation of IL-6 expression and production. They concluded that using 780 nm with high power and low energy density or 660 nm with low power and high energy density achieved similar results and the additional decrease by the parameters used with 780 nm suggest that this wavelength returned the cells to a nonstimulated state.

Lopes-Martins et al.74 found a true biphasic response occurred in the neutrophils isolated from mice treated with different energy densities (1, 2.5, and 5  J/cm2) with a maximum effect at 2.5  J/cm2.

Huang et al.47 irradiated cortical neuronal cells with a diode laser using 810 nm, 20  mW/cm2, 3  J/cm2, spot size of 5 cm, 150 s. They found that laser treatment reduced oxidative stress in primary cortical neurons in vitro.

Studies using PBM in vitro on cells with high numbers of mitochondria that reported positive results are summarized in Table 3. Ineffective parameters in vitro in cells with high numbers of mitochondria are reported in Table 7. In some cases, the same studies are included in both Tables 3 and 7 (effective and ineffective parameters) when the authors varied the parameters.

Table 3

Effective treatment of PBM: in vitro studies in cells with higher number of mitochondria.

AuthorsWavelength (nm)FluenceIrradianceCell type
Fernandes et al.467802.6  J/cm21.75  W/cm2; 70 mW, 0.04  cm2, 1.5 sMacrophage
Huang et al.478103  J/cm220  mW/cm2; 150 s, spot size 5 cmNeural cells
Huang et al.488103  J/cm225  mW/cm2, 2 min, spot size 5 cmNeural cells
Sharma et al.498100.03, 0.3, 3, 10, peak at 3  J/cm225  mW/cm2Mouse cortical neuron
Oron et al.508080.05  J/cm250  mW/cm2Human neural cells
Chen et al.268081  J/cm244.7  mW/cm2
170 mW, 3.8  cm2, 22.4 s
Monocyte
Souza et al.517803  J/cm2275  mW/cm2 [Power=70  mW,
1.5 s (2×) effective power 53.9 mW]
Area=0.196  cm2
Beam spot area=0.04  cm2
Macrophage
Ferraresi et al.52Cluster 40 LEDs (20 infrared 850 nm and 20 red 630 nm)2.5  J/cm228  mW/cm2
50 mW (IR) and 25 mW (red)
Cluster: 1000 mW (IR) and 500 mW (red)
45  cm2, 90 s, distance: 156 mm
Myotube C2C12
Amaroli et al.198083.0  J/cm2100  mW/cm2
100 mW
spot area:1  cm2
Paramecium
Amaroli1980864  J/cm21000  mW/cm2
100 mW, spot area=1  cm2
Paramecium
Chen et al.266601  J/cm20.8  mW/cm2
6 mW, 7.5  cm2
1250 s
Monocyte
Chen et al.266602  J/cm20.8  mW/cm2
6 mW, 7.5  cm2
2500 s
Monocyte
Souza et al.516607.5  J/cm2 effective fluence 1.15  J/cm257.4  mW/cm2
Power=15  mW, 20 s
Effective power  11.25  mW
Irradiated area=0.196  cm2
Beam spot area=0.04  cm2
Macrophage
Fernandez et al.466607.5  J/cm20.375  W/cm2
15 mW, 0.04  cm2, 20 s
Macrophage

3.2.2.

In vitro studies with cells with lower numbers of mitochondria

Tschon et al.55 irradiated osteoblast–like cells using a 915-nm diode laser at the following parameters: 100 Hz pulsed mode, 50% duty cycle, and output power of 0.575 W. Laser energy was delivered in defocused mode using a concave lens to cover the growth area (1.91  cm2) at distance of 19 mm (power density calculated by current authors from available information was 150  mW/cm2). The laser was applied for 48, 96, and 144 s producing doses of 5, 10, and 15  J/cm2 (energy density calculated by current authors from available information was 7.2, 14.4, and 21.56  mJ/cm2), and specimens were examined after 4, 24, 48, and 72 h. In vitro scratch wounds treated with 5 and 10J/cm2 were the first to reach complete coverage after 72 h, followed by 15  J/cm2, which reached complete healing after 96 h.

Pyo et al.56 studied the effect of hypoxia and PBM on the expression of bone morphogenetic protein-2 (BMP-2); transforming growth factor-beta-1 (TGF-β1); type I collagen, osteocalcin; hypoxia inducible factor-1 (HIF-1) and AKT. Osteoblast cells were cultured under 1% oxygen tension and then exposed to hypoxia. These cells were then irradiated with an 808 nm diode laser; 1000 mW, continuous wave (CW) for 15 s for a stated energy density of 1.2  J/cm2 at each session (power density calculated by current authors from available information was 80  mW/cm2). Other cells were cultured 24 h more under hypoxia and irradiated a second and third time for a total energy density of 1.2, 2.4, and 3.6J/cm2. Finally, further hypoxia was applied to the cells after irradiation. Cells were not exposed to laser energy in the control groups and were incubated under hypoxia at 1, 24, and 48 h. Results showed that hypoxia did not affect osteoblast viability (in the control group) and BMP-2, but it resulted in a decrease in osteocalcin, TGF-β, and expression of type I collagen. However, PBM applied to hypoxic osteoblasts stimulated osteoblast differentiation and proliferation through an increased expression of BMP-2, osteocalcin, and TGF-β. In addition, PBM inhibited HIF-1 expression and inhibited production of Akt.

Migliario et al.57 irradiated murine preosteoblasts (MC-3 T3 –E1) in order to evaluate the effect of PBM on ROS in cells labeled with an ROS marker. They used a diode laser at 930 nm, 1 W, irradiation time of 1, 5, 10, 25, and 50 s, for a delivered fluence of 1.57, 7.87, 15.74, 39.37, and 78.75  J/cm2 (spot area calculated by current authors from available information was 0.63  cm2 and irradiance of 1.57  W/cm2). The laser application was delivered three times at 0, 24, and 48 h. They found that ROS generation was dose dependent and doubled at higher fluences (25 to 50  J/cm2). Also, laser irradiation was able to increase preosteoblast proliferation starting from a fluence of 5  J/cm2. Increasing the fluence produced an increase in cell proliferation up to 25  J/cm2 and then a decrease at 50  J/cm2. The peak of cell proliferation occurred at 10  J/cm2. These results are partially in disagreement with other studies that suggest that 1 to 5  J/cm2 was optimal for cell proliferation. Contradictory results may be due to differences in irradiation parameters (wavelength, output power, energy density).

Zhang et al.53 irradiated fibroblast cells with 628 nm. Power output was constant at 15 mW, irradiance 11.46  mW/cm2, and distance of 0.75 cm. Samples were irradiated for various time periods to yield final energy doses of 0.44, 0.88, 2.00, 4.40, and 9  J/cm2. They found a maximum increase in human fibroblast cell proliferation with a fluence of 0.88  J/cm2 and a reduction in the proliferation at 9  J/cm2.

Khadra et al.75 investigated the effect of single and multiple doses on attachment and proliferation of human fibroblasts. Cells were cultured on titanium implants and divided into three groups: group I was used as a control, group II received GaAlAs 830 nm, output power 84 mW, 9 cm distance to the cells, a single dose of 3  J/cm2, 360 s (spot area calculated by current authors from available information was 10  cm2 and irradiance of 0.0084  W/cm2), group III was divided into three subgroups and exposed to multiple doses (one dose on each of three consecutive days) of 0.75, 1.5, and 3  J/cm2 corresponding to exposure times of 90, 180, and 360 s (spot area calculated by current authors from available information was 10  cm2). Results indicated that samples exposed to multiple doses of 1.5 and 3  J/cm2 showed a significantly proliferation. They concluded that the attachment of human fibroblasts to the titanium implant was enhanced by PBM. Both multiple and single doses significantly increased cellular attachment. Finally, 0.75  J/cm2 did not promote proliferation and cell attachment.

Skopin and Molitor58 studied the effect of using different doses and different irradiances on wound healing in fibroblast cultures using 980-nm diode laser. They applied an irradiance of: 26, 49, 73, 97, and 120  mW/cm2 for a constant 2 min each, delivering 3.1, 5.9, 8.8, 11.6, and 14.4  J/cm2. They found a significant increase in cell division when using 26, 73, and 97  mW/cm2. This effect was negated at 120  mW/cm2.

Al-Watban and Andres76 studied the effect of He–Ne laser on the proliferation of hamster ovary and human fibroblasts. Irradiance was held constant at 1.25  mW/cm2 using an accumulated dose over three consecutive days of 60 to 600  mJ/cm2. They found a peak response at 180  mJ/cm2. This study suggested that there is activation at a lower dose from 2  mJ/cm2 with a peak at 180  mJ/cm2. At higher doses, greater than 300  mJ/cm2, there was bioinhibition.

Studies using PBM in vitro on cells with low numbers of mitochondria that reported positive results are summarized in Table 4. Ineffective parameters in vitro in cells with low numbers of mitochondria are reported in Table 8. In some cases, the same studies are included in both Tables 3 and 7 (effective and ineffective parameters) when the authors varied the parameters.

Table 4

Effective treatment of PBM: in vitro studies in cells with lower number of mitochondria.

AuthorsWavelength (nm)Fluence (J/cm2)IrradianceCell type
Wang et al.54420316  mW/cm2
4  cm2, 188 s
Adipose stem cells
Wang et al.54540316  mW/cm2
4  cm2, 188 s
Adipose stem cells
Zhang et al.536280.8811.46  mW/cm2
Output power 15 mW, 0.76 cm distance to the surface, area=9.6  cm2
Fibroblast
Zhang et al.536282.011.46  mW/cm2
Output power 15 mW, 0.76 cm distance to the surface, area=9.6  cm2
Fibroblast
Zhang et al.536284.411.46  mW/cm2
Output power 15 mW, 0.76 cm distance to the surface, area=9.6  cm2
Fibroblast
Khadra et al.328301.58.4  mW/cm2
84 mW, 10  cm2, 9 cm distance to cells
Fibroblast
Khadra et al.328303.08.4  mW/cm2
84 mW, 10  cm2, 360 s, 9 cm distance to cells
Fibroblast
Tschon et al.559157.2150  mW/cm2, 100 Hz, 50% duty cycle,
power 0.575 W, 48 s
Osteoblast
Tschon et al.5591514.4150  mW/cm2
50% duty cycle, power 0.575 W, 96 s
Osteoblast
Migliario et al.579307.81580  mW/cm2
1 W, 5 s, 0.63  cm2
Preosteoblast
Migliario et al.57930151580  mW/cm2
1 W, 10 s, 0.63  cm2
Preosteoblast
Migliario et al.57930391580  mW/cm2
1 W, 25 s, 0.63  cm2
Preosteoblast
Pyo et al.568081.280  mW/cm2
15 s, 1 W
Osteoblast
Skopin et al.589803.126.7  mW/cm2Fibroblast
Skopin et al.589808.873  mW/cm2Fibroblast
Skopin et al.5898011.697  mW/cm2Fibroblast
Bozkurt et al.60940180.3  W/cm2
0.3 W, 60 s, distance: 0.5 to 1 mm
Cementoblast
Wang et al.73810316  mW/cm2
4  cm2, 188 s
Adipose stem cells
Wang et al.619800.316  mW/cm2
4  cm2, 18.8 s
Adipose stem cells

3.2.3.

In vivo studies in tissues with high number of mitochondria: heart, brain, muscle, inflammation

Oron et al.62 treated myocardial infarction with LLLT using an 810-nm laser. Fluence was held constant at 0.9  J/cm2 while irradiance was varied to deliver 2.5, 5, and 25  mW/cm2. A peak response was found at 5  mW/cm2, while treatment was less effective when using 2.5 and 25  mW/cm2.

Castano et al.17 studied inflammatory arthritis in rats, comparing the effect of using high and low fluences (3 to 30  J/cm2) delivered at high and low irradiance (5 to 50  mW/cm2). Effective treatment was observed when using: 30  J/cm2 at 50  mW/cm2 for 10 min and 30  J/cm2 at 5  mW/cm2 for 100 min. Low fluence of 3  J/cm2 at 5  mW/cm2 for 10 min was also effective. Only the dose of 3  J/cm2 at 50  mW/cm2 for 1 min was ineffective. They concluded that at higher fluence (30  J/cm2), the PBM effect on arthritis did not depend on irradiance as both high and low irradiance were effective, while at a lower fluence of 3  J/cm2, only the lower irradiance was effective. Therefore, they concluded that the duration of the light exposure was of great importance. While some studies found (3  J/cm2, 50  mW/cm2) beneficial, this study did not. They suggest that because the duration was only 1 min, the light did not have sufficient time to produce a sufficient activation of cellular metabolism.17

Salehpour et al.77 compared the therapeutic effect of a 10-Hz pulsed wave of NIR (810 nm) and red (630 nm) lasers with citalopram in rats that had been subjected to a model of chronic mild stress that causes depression. After inducing stress in rats, they were divided into: group I receiving PBM using NIR 810 nm and group II receiving 630-nm coherent light using identical parameters of: 10-Hz gated wave (50% duty cycle), fluence of 1.2  J/cm2 per session, output power 35 and 240 mW, respectively, 2 ms duration for both type of lasers, beam diameter of 3 mm, contact mode, and spot size of 0.07  cm2. Laser power was set at 6.2 W in the red wavelength and 39.3 W in the infrared wavelength for an irradiance of 89 and 562  mW/cm2, respectively. The average fluence for each session was 1.2  J/cm2 and totaling 14.4  J/cm2 for the entire 12 session treatment. Finally, group III was treated with the antidepressant drug citalopram that works by decreasing cortisol levels. Results showed that PBM using 10-Hz pulsed NIR laser had a better effect than red laser and the same effect as citalopram.

Salehpour et al.59 studied brain mitochondrial function in mice after inducing mitochondrial dysfunction by administration of D-galactose. This model is considered to be a model of age-related cognitive dysfunction. Animals were treated with wavelengths of 660 and 810 nm at two different fluences: 4 and 8  J/cm2, 10 Hz, 4.75  W/cm2, 88% duty cycle, 200 mW, in contact, three times a week, 48 h between sessions, and 7-mm diameter power meter sensor. They found poor results with both wavelengths at 4  J/cm2 and an amelioration of the aging-induced mitochondrial dysfunction with 8  J/cm2

Wu et al.78 induced traumatic brain injury (TBI) in mice and treated the animals using 660, 730, 810, or 980 nm, single dose treatment of 36  J/cm2 using an irradiance of 15  mW/cm2, 4-min duration, 4 h after injury. They found a significant improvement for mice having moderate to severe injury only when using 660 nm and 810 nm. The most desirable effect was seen at 810 nm, and both 730 and 980 nm did not produce any benefit.

Lopes-Martins et al.18 investigated the effect of PBM on muscular fatigue in rats during tetanic contractions. Four groups of 32 rats received different doses of PBMT (0.5, 1.0, and 2.5  J/cm2), using parameters of 655 nm, spot area 0.08  cm2, 25 mW, 2.5 mW; 31.25  mW/cm2. Groups: 0.5  J/cm2 (32 s), 1  J/cm2 (80 s), 2.5  J/cm2 (160 s). Only the groups of 0.5 and 1  J/cm2 prevented the development of muscular fatigue in rats during repeated tetanic contractions.

Lopes-Martins et al.74 in another study used 650-nm wavelength on acute inflammatory pleurisy in mice. Using the same power of 2.5 mW but different fluences of 3, 7.5, and 15  J/cm2. They found that under these conditions, 7.5  J/cm2 were more effective than either 3 or 15  J/cm2.

De Almeida et al.79 studied muscle performance after inducing muscle contraction in 30 rats. Using 904 nm, 15-mW average power and different energies (0.1, 0.3, 1.0, and 3.0 J) they found that the 1.0 and 3.0 J groups showed significant enhancement (P<0.01) in total work. They conclude that 1.0 J decreased postexercise muscle damage and enhanced muscle performance.

Studies using PBM in vivo in tissues with high numbers of mitochondria that reported positive results are summarized in Table 5. Ineffective parameters PBM in vivo in tissues with high numbers of mitochondria are reported in Table 9. In some cases, the same studies are included in both Tables 5 and 9 (effective and ineffective parameters) when the authors varied the parameters.

Table 5

Effective PBM treatment: in vivo on tissues with higher number of mitochondria.

AuthorsWavelength (nm)FluenceIrradianceTissue type
Alves et al.25808142.4  J/cm21.78  W/cm2
4 J, 50 mW, 0.028  cm2, 80 s per point
Arthritis
Oron et al.62,638100.3  J/cm25  mW/cm2
5 mW, area 1.1  cm2, 60 s
Heart
Oron et al.62,638100.9  J/cm25  mW/cm2Myocardium tissue
Castano et al.1781030  J/cm250  mW/cm2Arthritis
Castano et al.1781030  J/cm25  mW/cm2Arthritis
Castano et al.178103  J/cm25  mW/cm2Arthritis
Salehpour et al.598101.2  J/cm2560  mW/cm2
39.3 W, spot size 0.07  cm2
Brain
Salehpour et al.648108  J/cm289  mW/cm2
6.2 W, spot size 0.07  cm2
Brain
Wu et al.7881036  J/cm215  mW/cm2Brain
Blanco et al.701064250  mW/cm260  J/cm2Brain (human)
Disner et al.711064250  mW/cm260  J/cm2Brain (human)
Ando et al.1381036  J/cm250  mW/cm2TBI
Zhang et al.67810Fluence reaching the cortex 1.8 to 2.5  J/cm2
Average irradiance 36  J/cm2
150  mW/cm2
Pulse freq 10 Hz, pulse duration 50 ms, 4 min
TBI
Salehpour et al.688101.2  J/cm289 and 562  mW/cm2
35 and 240 mW
10 Hz, 50% duty cycle; 0.07  cm2
Brain
Baroni et al.68Cluster with 69 LEDs 660/850 nm206.89  J/cm26.89  W/cm2
200 mW; 6 J per diode (30 s); 0.02  cm2
30 J per application point (5×6  J)6 application points: total energy 180 J
Femoral quadriceps
Zhang et al.806350.96  J/cm26.37  mW/cm2
5 mW, laser beam width 10 mm, 150 s
Preconditioning myocardium
Salehpour et al.596608  J/cm24.75  W/cm2
88% duty cycle, 200 mW, in contact three times a week, 7 mm diameter
Brain
Wu et al.7866036  J/cm215  mW/cm2Brain
Lopes-Martins et al.186550.5  J/cm231.25  mW/cm2
2.5 mW, spot area 0.08  cm2, 25 mW, 32 s
Muscle
Lopes-Martins et al.186551  J/cm231.25  mW/cm2
2.5 mW, spot area 0.08  cm2, 25 mW, 80 s, 2.5 mW
Muscle

3.2.4.

In vivo studies in tissues with a lower number of mitochondria: skin, bone, cartilage

Lanzafame et al.15 treated pressure ulcers in mice with a 670-nm diode laser. Maintaining a constant fluence of 5  J/cm2 and using different irradiances (0.7, 2, 8, 40  mW/cm2), they found a significant improvement at 8  mW/cm2.

Prabhu et al.81 found a biphasic dose response on excisional wound healing in mice when using a He–Ne laser (632 nm, 7 mW, 4  mW/cm2 at different fluences (1, 2, 3, 4, 6, 8, and 10  J/cm2). A clear biphasic dose response occurred with a peak benefit at a fluence of 2  J/cm2 and an inhibitory effect at the higher dose of 10  J/cm2.

Gal et al.82 compared the wound tensile strength in rats at different power densities using 670 nm. A positive effect was seen when using 4  mW/cm2 delivered for 20 min, 50 s, (5  J/cm2), but this effect was not seen when using 15  mW/cm2 delivered for 5 min, 33 s, (5  J/cm2) at the same wavelength. This suggests that delivering the same fluence at a lower irradiance over more time was more effective.

Al-Watban and Delgado83 studied, in vivo, the effect of laser irradiation on burn wound healing in rats. They created a superficial burn with an area of 1.534  cm2 and irradiated the wound with a diode laser at 670 nm, 200 mW, three times per week for 12 weeks at different doses of 1, 5, 9, and 19  J/cm2. Only the groups receiving the lower doses of 1 and 5  J/cm2 showed significantly better wound healing compared to the control, with the greatest effect obtained at 1  J/cm2.

Studies using PBM in vivo in tissues with low numbers of mitochondria that reported positive results are summarized in Table 6. Ineffective parameters PBM in vivo in tissues with low numbers of mitochondria are reported in Table 10. In some cases, the same studies are included in both Tables 6 and 10 (effective and ineffective parameters) when the authors varied the parameters.

Table 6

Effective PBM treatment: in vivo on tissues with lower number of mitochondria.

AuthorsWavelength (nm)Fluence (J/cm2)IrradianceTissue type
Mendez et al.2183050125  mW/cm2
35 mW 0.6 cm diameter
Wound healing
Lanzarfane et al.1567058  mw/cm2Ulcer
Prabhu et al.2163224.0  mW/cm2
7 mw, 1.75  cm2
Wound healing
Gal et al.21670515  mw/cm2Wound tensile strength
Al-Watban et al.216701 and 5130  mW/cm2
200 mW, 1.534  cm2
Wound healing
Mendez et al.2183020125  mW/cm2
35 mW, 0.6 cm diameter
Wound healing
Barbosa et al.207901403500  mW/cm2
100 mW, 4 J, spot size 0.028  cm2
Bone
Barbosa et al.208301403500  mW/cm2
100 mW, 4 J, spot size 0.028  cm2
Bone

3.3.

III-Effect of Varying the Mode of Delivery on PBM Efficiency: CW or Pulsed

In a comprehensive literature review,84 Hamblin included 33 studies, nine of them directly comparing pulsed wave and CW. Six of these studies found that pulsed wave offered better results than CW; one study found that both modes were equally effective and only two studies reported better result using CW. Hamblin et al. concluded from this review that pulsed light may be superior to CW light, particularly for wound healing and poststroke management, whereas CW may be more beneficial in patients requiring nerve regeneration. In addition, they concluded that it is impossible to draw any correlation between pulse frequency and pathological condition. They found that no particular frequency appears to be more or less effective than others. Finally, this review reported that the following frequencies were beneficial: 2, 10, 25, 50 Hz when using (670 nm, 20 mW, energy density, 2  J/cm2), 100 Hz when using (808 nm, 37.5  mW/cm2, 0.9  J/cm2) 292 Hz when using (800  mW/cm2; 21.6  J/cm2), 600 Hz when using (670 nm, 10 mW, 5  J/cm2), 1000 Hz when using (808 nm, 7.5  mW/cm2, 0.9–1.2 J, duty cycle, 30%), 1500 Hz when using (5  mW/cm2); 3000 Hz when using (10  mW/cm2) and 8000 Hz (N/A).

Gigo-Benato et al.14 compared the effect of combined CW and pulsed laser (CW+PW) using 808 nm (CW) and 905 nm (PW) to either the CW (808 nm) or PW (905 nm) laser used separately. CW was applied at 29  J/cm2 while the pulsed wave laser was applied at 40  J/cm2. Results suggested that the combined laser was more effective in nerve regeneration than the CW alone or the PW alone.

Al-Watban and Zhang16 evaluated the effects of using both pulsed and CW PBM in rats wound healing. After creation of elliptical wounds, animals were treated with a 635-nm diode laser, average power of 3.4 mW, spot size of 3.8  cm2, wound size of 1.04  cm2, irradiance of 0.89  mW/cm2, treatment duration 18.7 min and fluence of 1  J/cm2, three times per week. The dose was delivered using either CW or pulsed mode at: 100, 200, 300, 400, or 500 Hz. They found that the effect of using CW was more efficient than using pulsed laser and, when comparing different frequencies, 100 Hz had better effect on wound healing than the other frequencies.

This article contradicts Hamblin, who concluded that pulsed mode was more effective than CW in wound healing. Perhaps, Al-Watban found that CW was more efficient because he did not use the same fluence in CW that he used in pulsed mode. Moreover, he used gated CW rather than true pulsed wave.16

Ando et al.13 treated TBI in mice comparing pulsed and CW 810-nm laser irradiation. The parameters used were: 810-nm diode laser, irradiance of 50  mW/cm2, spot diameter of 1 cm onto the injured head with a 12-min exposure giving a fluence of 36  J/cm2. They found that 10 Hz produced better results than 100 Hz or continuous mode.

el Sayed and Dyson85 compared the effect of four different frequencies (2.5, 20, 292 and 20,000 Hz) and found that only 20 and 292 Hz were beneficial.

Sushko et al.86 investigated pain induced in mice by hypodermic injection of 20 ml of 5% formalin solution into the footpad. They irradiated the mice using 640 and 880 nm LED in continuous or pulsed mode for 10 min. They found that pulsed mode was more effective than CW and frequencies of 10 and 8000 Hz were most effective, whereas pulse repetition rates of 200 and 600 Hz were less effective.

Ueda and Shimizu87 studied the effect of three different pulse repetition rates on osteoblast-like cells from rats using these parameters (830 nm, 500 mW, 0.48 to 3.84  J/cm2) in CW mode and (1, 2, and 8 Hz) in pulsed mode. They found that 1 and 2 Hz markedly stimulated cellular proliferation, ALP activity, ALP gene expression, and bone nodule formation, and that 2 Hz was the best pulse repetition rate to stimulate bone nodule formation.

4.

Review of Which Parameters Lead to Effective and Ineffective PBMT

It is difficult to compare studies done with different parameters, protocols, treatment objectives, and biological target tissues. Often, parameters are not completely presented or are of questionable accuracy. In this part of the review analysis, an attempt is made to draw at least some general inferences from the data presented in Tables 3Table 4Table 5Table 6Table 7Table 8Table 910.

Table 7

Ineffective treatment of PBM: in vitro studies in cells with higher number of mitochondria.

AuthorsWavelength (nm)Fluence (J/cm2)IrradianceCell type
Sharma et al.498103025  mW/cm2Mouse cortical neurons
Chen et al.2666030.8  mW/cm2
6 mW, 7.5  cm2
3750 s
Monocyte
Chen et al.2666020.8  mW/cm2
6 mW, 7.5  cm2
2500 s
Monocyte
Amaroli et al.198083.01000  mW/cm2, 1 W, 1  cm2 spot areaParamecium
Amaroli et al.1980864100  mW/cm2, 1 W, 1  cm2 spot areaParamecium

Table 8

Ineffective treatment of PBM in vitro studies in cells with lower number of mitochondria.

AuthorsWavelength (nm)Fluence (J/cm2)IrradianceCell type
Tschon et al.5591520.56150  mW/cm2
100 Hz, 50% duty cycle, power 0.575 W 144 s
Osteoblast
Migliario et al.579301.571580  mW/cm2
1 W,1 s, 0.63  cm2
Preosteoblast
Migliario et al.5793078.71580  mW/cm2
1 W, 50 s, 0.63  cm2
Preosteoblast
Skopin et al.589805.949  mW/cm2Fibroblast
Skopin et al.5898014.4120  mW/cm2Fibroblast
Zhang et al.536289.011.4  mW/cm2
15 mW, distance of 0.75 cm
Fibroblast
Khadra et al.758300.758.4  mW/cm2
84 mW, 10  cm2, 360 s, 9 cm distance to cells
Fibroblast
Wang et al.739802016  mW/cm2
4  cm2, 1 W
Adipose stem cells

Table 9

Ineffective PBM treatment in vivo on tissues with higher number of mitochondria.

AuthorsWavelength (nm)Fluence (J/cm2)IrradianceTissue type
Oron et al.62,638100.32.5  mW/cm2
5 mW, area of irradiation of 1.1  cm2
Heart
Oron et al.62,638100.325  mW/cm2
5 mW, area of irradiation of 1.1  cm2
Heart
Salehpour et al.5966044.75  W/cm2
10 Hz, 4.75  W/cm2, 88% duty cycle, 200 mW
Brain
Salehpour et al.5981044.75  W/cm2
10 Hz, 4.75  W/cm2, 88% duty cycle, 200 mW
Brain
Wu et al.789803615  mW/cm2Brain
Alves et al.25808142.43.57  W/cm2
4 J, 50 mW, 0.028  cm2, 80 s per point
Arthritis
Lopes-Martins et al.186552.531.25  mW/cm2
2.5 mW, spot area 0.08  cm2, 25 mW, 160 s, 2.5 mW
Muscle

Table 10

Ineffective PBM treatment in vivo on tissues with lower number of mitochondria.

AuthorsWavelength (nm)Fluence (J/cm2)IrradianceTissue type
Lanzafame et al.156705.00.7  mW/cm2Ulcers (wound healing)
Lanzafame et al.156705.02.0  mW/cm2Ulcers (wound healing)
Gal et al.826705.015  mW/cm2Wound healing
Lanzafame et al.156705.040  mW/cm2Wound healing
Prabhu et al.81632104.0  mW/cm2
7 mw, 1.75  cm2
Wound healing
Al-Watban et al.836709.0130  mW/cm2
200 mW, 1.534  cm2
Wound healing
Al-Watban et al.8367019130  mW/cm2
200 mW, 1.534  cm2
Wound healing
Kilik et al.8863651  mW/cm2
Probe to wound 10 cm
Wound healing

4.1.

Wavelength

Wavelength affects tissue penetration. Shorter wavelengths (600 to 700 nm) are considered best to treat superficial tissue, whereas longer wavelengths (780 to 950 nm) are preferred to treat deeper tissues. Red wavelengths penetrate 0.5 to 1 mm and near-infrared energy penetrates 2 mm before losing 37% of its intensity.8991

The infrared wavelengths show better effects on bone repair compared to red wavelengths because red light has less capacity to penetrate compared to the infrared laser.

According to Karu,8 wavelengths between 700 and 770 nm do not have any significant activity. Wu et al.78 used a 730-nm laser on TBI in mice and found it to be ineffective while 660 and 810 nm lasers were effective. Gupta et al.92 carried out a similar comparison on wound healing in mice and again found that 660- and 810-nm lasers were effective, while a 730-nm laser was not effective.

Barbosa et al.20 concluded that the PBM effects of NIR were effective for more than 14 days, whereas the effects of red wavelength are lost after 14 days.

The combination of two wavelengths gives an additional effect of PBM. When comparing 830 and 685 nm, Mendez et al.21 found that 830 nm offered better results. Much work still remains to define the optimal wavelengths. Nevertheless, NIR wavelengths are preferable for deep tissues and targets within the body, which require substantial doses of light.

4.2.

Laser Versus Noncoherent Light

Both coherent lasers and noncoherent LEDs are used in PBMT. Laser beams are collimated and the light is more likely to be forward scattered within the tissue than noncollimated LED light.5 This means that the penetration depth is likely to be deeper with lasers provided all the other characteristics are identical. Moreover, lasers emit coherent light, while LED light is noncoherent. The coherence length is higher for smaller bandwidths. For instance, gas lasers such as He–Ne laser have very long coherence lengths. Diode lasers have somewhat greater bandwidths and consequently shorter coherence lengths. When coherent laser light interacts with tissue, small imperfections in the tissue structure lead to different phases occurring in the individual wavefronts leading to mutual interference patterns. These interference patterns are called “laser speckles” and the size of the speckles is related to the light wavelength. In the visible range, the sizes are less than 1  μm. Subcellular organelles (such as mitochondria) have dimensions of this order and a theory proposes that the laser speckles are better to stimulate mitochondria than noncoherent LED light.9395 A recent review concluded that there were no substantial differences between lasers and LEDs for PBM applications provided all the other light parameters were equal.96

4.3.

Fluence and Irradiance

The photon intensity i.e., irradiance (W/cm2 or spectral irradiance), must be adequate. Using higher intensity, the photon energy will be transformed to excessive heat in the target tissue and, using lower intensity, photons absorption will be insufficient to achieve the goal.

The dose also must be adequate (J/cm2). Using low irradiance and prolonging the irradiation time to achieve the ideal fluence or dose will not give an adequate final result. The Bunsen–Roscoe law of reciprocity, termed the second law of photobiology,97 does not hold true for low incident power densities.

There is no fixed value of dose or fluence that always produces a positive PBM effect. Even within different studies on the same animal models, there can be contradictory findings. For instance, three papers looked at peri-implant bone regeneration after PBM. Menezes et al.98 found that 20  J/cm2 was the best dose to deliver, whereas Massotti et al.22 and Mayer et al.23 found that 20  J/cm2 was the worst dose to deliver.

The optimal doses are directly related to different factors:

  • Wavelength

  • Type of treatment being delivered: pain relief, wound healing, or tissue regeneration

  • Power density or irradiance

  • Energy density or fluence

  • Depth of the target tissue being treated

  • Spot size of the beam reaching the tissue surface and the actual target tissue.

In an attempt to determine whether the delivered fluence (J/cm2) was more or less important than the irradiance (mW/cm2), we constructed scatter plots (Figs. 1Fig. 2Fig. 34) of both the effective and ineffective studies arranged according to our categorization of the studies in Tables 2Table 3Table 4Table 5Table 6Table 7Table 89.

Fig. 1

Studies on PBM of cells in vitro with higher numbers of mitochondria. (a) Effective (positive studies), (b) ineffective (negative studies), and (c) combination of effective (positive studies), and ineffective (negative studies).

JBO_23_12_120901_f001.png

Fig. 2

Studies on PBM of cells in vitro with lower numbers of mitochondria. (a) Effective (positive studies), (b) ineffective (negative studies), and (c) combination of effective (positive studies) and ineffective (negative studies).

JBO_23_12_120901_f002.png

Fig. 3

Studies on PBM of tissues in vivo with higher numbers of mitochondria. (a) Effective (positive studies), (b) ineffective (negative studies), and (c) combination of effective (positive studies) and ineffective (negative studies).

JBO_23_12_120901_f003.png

Fig. 4

Studies on PBM of tissues in vivo with lower numbers of mitochondria. (a) Effective (positive studies), (b) ineffective (negative studies), and (c) combination of effective (positive studies) and ineffective (negative studies).

JBO_23_12_120901_f004.png

4.3.1.

In vitro studies

Figure 1(a) shows the plot of in vitro studies in cells with higher numbers of mitochondria, whereas Fig. 1(b) shows the corresponding plot for cells with lower numbers of mitochondria. The following observations can be made. In all the effective studies, the fluence was relatively low (<7.5  J/cm2) and in several cases, less than 1  J/cm2. However, in the ineffective studies, the fluence values were larger (all >3  J/cm2), and in two cases, very large values (30 and 65  J/cm2). There were more studies in the effective group (11) than in the ineffective group (5). This suggests that high-mitochondrial cells respond well to PBM and that ineffective studies are more likely to be due to over-dosing than to under-dosing.

Figure 2(a) shows the effective in vitro studies in cells with lower mitochondrial numbers. Again, the positive studies outweigh the negative studies [Fig. 2(b)] (15 to 8). The fluence values in the positive studies in the lower mitochondrial number subgroup appeared to be overall higher than the fluences used in the positive studies in the higher mitochondrial number subgroup. The fluences used in the negative studies in the lower mitochondrial number subgroup were only a little higher than those in the positive studies, suggesting that over-dosing was not such a big problem as it was in the higher mitochondrial number subgroup [Fig. 1(b)]. There were three positive studies that used relatively high irradiances (>1.5  W/cm2), as opposed to only one study in the positive high-mitochondrial subgroup.

4.3.2.

In vivo studies

Figure 3(a) shows the plot of effective or positive studies in vivo on tissues composed of cells with higher numbers of mitochondria, whereas Fig. 3(b) shows the corresponding plot for ineffective or negative studies on tissues composed of higher mitochondrial number cells. Here, a difference is seen when comparing the two plots and with the analogous two plots from the in vitro studies. In the in vivo case, the fluence values in the effective studies subgroup [Fig. 3(a)] are higher than those in the ineffective studies subgroup [Fig. 3(b)]. This is the opposite of what was found in the in vitro case with cultured cells [compare Figs. 1(a) with 1(b)]. Hence, these observations tend to suggest that failure, in vivo, could be due to under-dosing while failure, in vitro, could equally well be due to over-dosing. In vivo, the depth of the tissue is important, while cells, in vitro culture, are generally a single monolayer. It is a fact that tissues with higher numbers of mitochondria (brain, heart, muscles, inflammatory cells) tend to be deeper within the body than tissues with lower numbers of mitochondria (skin, tendons, cartilage). There are, of course, some exceptions (bones and bone marrow), which have lower numbers of mitochondria but are still deep within the body.

Figure 4(a) shows the plot of effective treatment in tissue with a lower number of mitochondria, whereas Fig. 4(b) shows the plot of ineffective treatment on tissue with a lower number of mitochondria.

The following observation can be made:

The fluence values used in the positive studies are much higher than those in the negative studies, particularly when the tissue is deeper (such as bone). In addition, some studies used very low fluences of less than 1  J/cm2 to treat superficial tissue (wound healing) and had positive results.

Fluences used in the negative studies are generally less than 10  J/cm2, most of them used low irradiance. There are three studies that use lower fluence in combination with higher irradiance and produced positive results.

This would suggest that ineffective studies for tissue with lower mitochondria are more likely to be due to under-dosing rather than over-dosing. Fluence and irradiance are both important in determining the success of in vivo studies.

5.

Conclusions

The limitation of this analysis was the relatively small number of studies that passed our inclusion and exclusion criteria. Nevertheless, some tentative conclusions can be drawn from the analysis that we can at least propose for other researchers to confirm or refute, as more well-documented studies continue to be published in the coming years.

  • 1. Cells with higher numbers of mitochondria respond better to PBM than cells with lower numbers of mitochondria.

  • 2. Ineffective studies on cells with higher numbers of mitochondria are as likely to be due to over-dosing as they are to under-dosing.

  • 3. It is less likely that ineffective studies in cells with lower numbers of mitochondria will be due to over-dosing.

  • 4. The fluence delivered is more important in determining the success or failure of an in vitro study than the irradiance employed.

  • 5. Tissues with higher numbers of mitochondria tend to be deeper within the body than tissues with lower numbers of mitochondria, therefore, over-dosing is less likely.

  • 6. Ineffective studies in vivo are more likely to be due to under-dosing regardless of the number of mitochondria.

Disclosures

M.R.H. is on the Scientific Advisory Boards of the following companies: Transdermal Cap Inc., Cleveland, Ohio; Photothera Inc., Carlsbad, California; BeWell Global Inc., Wan Chai, Hong Kong; Hologenix Inc., Santa Monica, California; LumiThera Inc., Poulsbo, Washington; Vielight, Toronto, Canada; Bright Photomedicine, Sao Paulo, Brazil; Quantum Dynamics LLC, Cambridge, Massachusetts; Global Photon Inc., Bee Cave, Texas, Medical Coherence, Boston, Massachusetts; NeuroThera, Newark DE JOOVV Inc., Minneapolis-St. Paul, Minnesota; Illumiheal & Petthera, Shoreline, Washington; MB Lasertherapy, Houston, Texas and has consulted for: USHIO Corp,. Japan; Merck KGaA, Darmstadt, Germany; Philips Electronics Nederland B.V.; Johnson & Johnson Inc., Philadelphia, Pennsylvania; UVLRx Therapeutics, Oldsmar, Florida; Ultralux UV Inc., Lansing MI; AIRx Medical, Pleasanton, California; FIR Industries, Inc., Ramsey, New Jersey.

Acknowledgments

M.R.H. was supported by US NIH Grant Nos. R01AI050875 and R21AI121700.

References

1. 

E. Mester et al., “Stimulation of wound healing by means of laser rays. (Clinical and electron microscopical study),” Acta Chir. Acad. Sci. Hung., 19 (2), 163 –170 (1978). ACAHA3 0001-5431 Google Scholar

2. 

E. Mester, B. Szende and P. Gartner, “The effect of laser beams on the growth of hair in mice,” Radiobiol. Radiother. (Berl), 9 (5), 621 –626 (1968). Google Scholar

3. 

H. Chung et al., “The nuts and bolts of low-level laser (light) therapy,” Ann. Biomed. Eng., 40 (2), 516 –533 (2012). https://doi.org/10.1007/s10439-011-0454-7 ABMECF 0090-6964 Google Scholar

4. 

P. A. Jenkins and J. D. Carroll, “How to report low-level laser therapy (LLLT)/photomedicine dose and beam parameters in clinical and laboratory studies,” Photomed. Laser Surg., 29 (12), 785 –787 (2011). https://doi.org/10.1089/pho.2011.9895 Google Scholar

5. 

F. Martius, “Das Amdt-Schulz Grandgesetz,” Munch Med. Wschr, 70 1005 –1006 (1923). Google Scholar

6. 

E. J. Calabrese, “Originator of the hormesis concept: Rudolf Virchow or Hugo Schulz,” Hum. Exp. Toxicol., 37 (9), 889 –890 (2017). https://doi.org/10.1177/0960327117751237 HETOEA 0960-3271 Google Scholar

7. 

E. J. Calabrese and M. P. Mattson, “How does hormesis impact biology, toxicology, and medicine?,” NPJ Aging Mech. Dis., 3 13 (2017). https://doi.org/10.1038/s41514-017-0013-z Google Scholar

8. 

T. I. Karu, “Multiple roles of cytochrome c oxidase in mammalian cells under action of red and IR-A radiation,” IUBMB Life, 62 (8), 607 –610 (2010). https://doi.org/10.1002/iub.359 1521-6543 Google Scholar

9. 

L. F. de Freitas and M. R. Hamblin, “Proposed mechanisms of photobiomodulation or low-level light therapy,” IEEE J. Sel. Top. Quantum Electron., 22 (3), 348 –364 (2016). https://doi.org/10.1109/JSTQE.2016.2561201 IJSQEN 1077-260X Google Scholar

10. 

Y. Y. Huang et al., “Biphasic dose response in low level light therapy,” Dose Response, 7 (4), 358 –383 (2009). https://doi.org/10.2203/dose-response.09-027.Hamblin Google Scholar

11. 

Y. Y. Huang et al., “Biphasic dose response in low level light therapy—an update,” Dose Response, 9 (4), 602 –618 (2011). https://doi.org/10.2203/dose-response.11-009.Hamblin Google Scholar

12. 

M. R. Hamblin, “Mechanisms and mitochondrial redox signaling in photobiomodulation,” Photochem. Photobiol., 94 (2), 199 –212 (2018). https://doi.org/10.1111/php.12864 PHCBAP 0031-8655 Google Scholar

13. 

T. Ando et al., “Comparison of therapeutic effects between pulsed and continuous wave 810-nm wavelength laser irradiation for traumatic brain injury in mice,” PLoS One, 6 (10), e26212 (2011). https://doi.org/10.1371/journal.pone.0026212 POLNCL 1932-6203 Google Scholar

14. 

D. Gigo-Benato et al., “Low-power laser biostimulation enhances nerve repair after end-to-side neurorrhaphy: a double-blind randomized study in the rat median nerve model,” Lasers Med. Sci., 19 (1), 57 –65 (2004). https://doi.org/10.1007/s10103-004-0300-3 Google Scholar

15. 

R. J. Lanzafame et al., “Reciprocity of exposure time and irradiance on energy density during photoradiation on wound healing in a murine pressure ulcer model,” Lasers Surg. Med., 39 (6), 534 –542 (2007). https://doi.org/10.1002/lsm.20519 LSMEDI 0196-8092 Google Scholar

16. 

F. A. Al-Watban and X. Y. Zhang, “The comparison of effects between pulsed and CW lasers on wound healing,” J. Clin. Laser Med. Surg., 22 (1), 15 –18 (2004). https://doi.org/10.1089/104454704773660921 JCLSEO Google Scholar

17. 

A. P. Castano et al., “Low-level laser therapy for zymosan-induced arthritis in rats: Importance of illumination time,” Lasers Surg. Med., 39 (6), 543 –550 (2007). https://doi.org/10.1002/lsm.20516 LSMEDI 0196-8092 Google Scholar

18. 

R. A. Lopes-Martins et al., “Effect of low-level laser (Ga-Al-As 655 nm) on skeletal muscle fatigue induced by electrical stimulation in rats,” J. Appl. Physiol. (1985), 101 (1), 283 –288 (2006). https://doi.org/10.1152/japplphysiol.01318.2005 Google Scholar

19. 

A. Amaroli et al., “Photobiomodulation by infrared diode laser: effects on intracellular calcium concentration and nitric oxide production of paramecium,” Photochem. Photobiol., 92 (6), 854 –862 (2016). https://doi.org/10.1111/php.2016.92.issue-6 PHCBAP 0031-8655 Google Scholar

20. 

D. Barbosa et al., “Effects of low-level laser therapy (LLLT) on bone repair in rats: optical densitometry analysis,” Lasers Med. Sci., 28 (2), 651 –656 (2013). https://doi.org/10.1007/s10103-012-1125-0 Google Scholar

21. 

V. T. M. Mendez et al., “Assessment of the influence of the dose and wavelength of LLLT on the repair of cutaneous wounds,” Proc. SPIE, 4950 (2003). https://doi.org/10.1117/12.476431 PSISDG 0277-786X Google Scholar

22. 

F. P. Massotti et al., “Histomorphometric assessment of the influence of low-level laser therapy on peri-implant tissue healing in the rabbit mandible,” Photomed. Laser Surg., 33 (3), 123 –128 (2015). https://doi.org/10.1089/pho.2014.3792 Google Scholar

23. 

L. Mayer et al., “Histologic and resonance frequency analysis of peri-implant bone healing after low-level laser therapy: an in vivo study,” Int. J. Oral Maxillofac Implants, 30 (5), 1028 –1035 (2015). https://doi.org/10.11607/jomi.3882 Google Scholar

24. 

R. W. Bunsen and H. E. Roscoe, “Bunsen-Roscoe law,” Photochem. Stud. Ann. Phys., 108 193 (1859). Google Scholar

25. 

A. C. Alves et al., “Effect of low-level laser therapy on the expression of inflammatory mediators and on neutrophils and macrophages in acute joint inflammation,” Arthritis Res. Ther., 15 (5), R116 (2013). https://doi.org/10.1186/ar4296 Google Scholar

26. 

C. H. Chen et al., “Effects of low-level laser therapy on M1-related cytokine expression in monocytes via histone modification,” Mediators Inflammation, 2014 1 –13 (2014). https://doi.org/10.1155/2014/625048 Google Scholar

27. 

S. L. Jacques, “Optical properties of biological tissues: a review,” Phys. Med. Biol., 58 (11), R37 –R61 (2013). https://doi.org/10.1088/0031-9155/58/11/R37 PHMBA7 0031-9155 Google Scholar

28. 

H. Kolarova, D. Ditrichova and J. Wagner, “Penetration of the laser light into the skin in vitro,” Lasers Surg. Med., 24 (3), 231 –235 (1999). LSMEDI 0196-8092 Google Scholar

29. 

K. Yoshida et al., “Relationship between microstructure of the skin surface and surface reflection based on geometric optics,” J. Dermatol. Sci., 66 (3), 225 –232 (2012). https://doi.org/10.1016/j.jdermsci.2012.03.011 JDSCEI 0923-1811 Google Scholar

30. 

T. Ohshiro, “The proximal priority theory: an updated technique in low level laser therapy with an 830 nm GaAlAs laser,” Laser Ther., 21 (4), 275 –285 (2012). https://doi.org/10.5978/islsm.12-OR-16 LATHE5 0898-5901 Google Scholar

31. 

A. Bhandari et al., “Modeling optical properties of human skin using Mie theory for particles with different size distributions and refractive indices,” Opt. Express, 19 (15), 14549 –14567 (2011). https://doi.org/10.1364/OE.19.014549 OPEXFF 1094-4087 Google Scholar

32. 

Y. Xu et al., “Study on mechanism of release oxygen by photo-excited hemoglobin in low-level laser therapy,” Lasers Med. Sci., 33 (1), 135 –139 (2018). https://doi.org/10.1007/s10103-017-2363-y Google Scholar

33. 

A. Keszler et al., “Wavelength-dependence of vasodilation and NO release from S-nitrosothiols and dinitrosyl iron complexes by far red/near infrared light,” Arch. Biochem. Biophys., 649 47 –52 (2018). https://doi.org/10.1016/j.abb.2018.05.006 Google Scholar

34. 

K. Kwon et al., “Enhancement of light propagation depth in skin: cross-validation of mathematical modeling methods,” Lasers Med. Sci., 24 (4), 605 –615 (2009). https://doi.org/10.1007/s10103-008-0625-4 Google Scholar

35. 

A. Shuaib and A. K. Bourisly, “Photobiomodulation optimization for spinal cord injury rat phantom model,” Transl. Neurosci., 9 67 –71 (2018). https://doi.org/10.1515/tnsci-2018-0012 Google Scholar

36. 

D. G. Nicholls, “Mitochondrial function and dysfunction in the cell: its relevance to aging and aging-related disease,” Int. J. Biochem. Cell Biol., 34 (11), 1372 –1381 (2002). https://doi.org/10.1016/S1357-2725(02)00077-8 IJBBFU 1357-2725 Google Scholar

37. 

B. F. Lang, M. W. Gray and G. Burger, “Mitochondrial genome evolution and the origin of eukaryotes,” Annu. Rev. Genet., 33 351 –397 (1999). https://doi.org/10.1146/annurev.genet.33.1.351 ARVGB7 0066-4197 Google Scholar

38. 

S. W. Perry et al., “Mitochondrial membrane potential probes and the proton gradient: a practical usage guide,” Biotechniques, 50 (2), 98 –115 (2011). https://doi.org/10.2144/000113610 BTNQDO 0736-6205 Google Scholar

39. 

J. S. Winterle and O. Einarsdottir, “Photoreactions of cytochrome C oxidase,” Photochem. Photobiol., 82 (3), 711 –719 (2006). https://doi.org/10.1562/2005-09-14-RA-684 PHCBAP 0031-8655 Google Scholar

40. 

E. D. Robin and R. Wong, “Mitochondrial DNA molecules and virtual number of mitochondria per cell in mammalian cells,” J. Cell Physiol., 136 (3), 507 –513 (1988). https://doi.org/10.1002/jcp.1041360316 Google Scholar

41. 

N. San Martin et al., “Mitochondria determine the differentiation potential of cardiac mesoangioblasts,” Stem Cells, 29 (7), 1064 –1074 (2011). https://doi.org/10.1002/stem.654 Google Scholar

42. 

K. J. Ahlqvist, A. Suomalainen and R. H. Hamalainen, “Stem cells, mitochondria and aging,” Biochim. Biophys. Acta, 1847 (11), 1380 –1386 (2015). https://doi.org/10.1016/j.bbabio.2015.05.014 BBACAQ 0006-3002 Google Scholar

43. 

Y. Shen et al., “The role of mitochondria in methamphetamine-induced inhibitory effects on osteogenesis of mesenchymal stem cells,” Eur. J. Pharmacol., 826 56 –65 (2018). https://doi.org/10.1016/j.ejphar.2018.02.049 Google Scholar

44. 

R. Bukowiecki, J. Adjaye and A. Prigione, “Mitochondrial function in pluripotent stem cells and cellular reprogramming,” Gerontology, 60 (2), 174 –182 (2014). https://doi.org/10.1159/000355050 GERNDJ 0304-324X Google Scholar

45. 

G. O. Cericato, M. A. Bittencourt and L. R. Paranhos, “Validity of the assessment method of skeletal maturation by cervical vertebrae: a systematic review and meta-analysis,” Dentomaxillofac. Radiol., 44 (4), 20140270 (2015). https://doi.org/10.1259/dmfr.20140270 Google Scholar

46. 

K. P. Fernandes et al., “Photobiomodulation with 660-nm and 780-nm laser on activated J774 macrophage-like cells: effect on M1 inflammatory markers,” J. Photochem. Photobiol. B, 153 344 –351 (2015). https://doi.org/10.1016/j.jphotobiol.2015.10.015 JPPBEG 1011-1344 Google Scholar

47. 

Y. Y. Huang et al., “Low-level laser therapy (LLLT) reduces oxidative stress in primary cortical neurons in vitro,” J. Biophotonics, 6 (10), 829 –838 (2013). https://doi.org/10.1002/jbio.201200157 Google Scholar

48. 

Y. Y. Huang et al., “Low-level laser therapy (810 nm) protects primary cortical neurons against excitotoxicity in vitro,” J. Biophotonics, 7 (8), 656 –664 (2014). https://doi.org/10.1002/jbio.v7.8 Google Scholar

49. 

S. K. Sharma et al., “Dose response effects of 810 nm laser light on mouse primary cortical neurons,” Lasers Surg. Med., 43 (8), 851 –859 (2011). https://doi.org/10.1002/lsm.21100 LSMEDI 0196-8092 Google Scholar

50. 

U. Oron et al., “Ga-As (808 nm) laser irradiation enhances ATP production in human neuronal cells in culture,” Photomed. Laser Surg., 25 (3), 180 –182 (2007). https://doi.org/10.1089/pho.2007.2064 Google Scholar

51. 

N. H. Souza et al., “Effect of low-level laser therapy on the modulation of the mitochondrial activity of macrophages,” Braz. J. Phys. Ther., 18 (4), 308 –314 (2014). https://doi.org/10.1590/bjpt-rbf.2014.0046 Google Scholar

52. 

C. Ferraresi et al., “Low-level laser (light) therapy increases mitochondrial membrane potential and ATP synthesis in C2C12 myotubes with a peak response at 3–6 hours,” Photochem. Photobiol., 91 (2), 411 –416 (2014). https://doi.org/10.1111/php.12397 PHCBAP 0031-8655 Google Scholar

53. 

Y. Zhang et al., “cDNA microarray analysis of gene expression profiles in human fibroblast cells irradiated with red light,” J. Invest. Dermatol., 120 (5), 776 –780 (2003). https://doi.org/10.1046/j.1523-1747.2003.12130.x JIDEAE 0022-202X Google Scholar

54. 

Y. Wang et al., “Photobiomodulation (blue and green light) encourages osteoblastic-differentiation of human adipose-derived stem cells: role of intracellular calcium and light-gated ion channels,” Sci. Rep., 6 33719 (2016). https://doi.org/10.1038/srep33719 SRCEC3 2045-2322 Google Scholar

55. 

M. Tschon et al., “Photobiomodulation with low-level diode laser promotes osteoblast migration in an in vitro micro wound model,” J. Biomed. Opt., 20 (7), 078002 (2015). https://doi.org/10.1117/1.JBO.20.7.078002 JBOPFO 1083-3668 Google Scholar

56. 

S. J. Pyo et al., “Low-level laser therapy induces the expressions of BMP-2, osteocalcin, and TGF-beta1 in hypoxic-cultured human osteoblasts,” Lasers Med. Sci., 28 (2), 543 –550 (2013). https://doi.org/10.1007/s10103-012-1109-0 Google Scholar

57. 

M. Migliario et al., “Laser-induced osteoblast proliferation is mediated by ROS production,” Lasers Med. Sci., 29 (4), 1463 –1467 (2014). https://doi.org/10.1007/s10103-014-1556-x Google Scholar

58. 

M. D. Skopin and S. C. Molitor, “Effects of near-infrared laser exposure in a cellular model of wound healing,” Photodermatol. Photoimmunol. Photomed., 25 (2), 75 –80 (2009). https://doi.org/10.1111/ppp.2009.25.issue-2 PPPHEW 0905-4383 Google Scholar

59. 

F. Salehpour et al., “Transcranial low-level laser therapy improves brain mitochondrial function and cognitive impairment in D-galactose-induced aging mice,” Neurobiol. Aging, 58 140 –150 (2017). https://doi.org/10.1016/j.neurobiolaging.2017.06.025 NEAGDO 0197-4580 Google Scholar

60. 

S. B. Bozkurt et al., “Biostimulation with diode laser positively regulates cementoblast functions, in vitro,” Lasers Med. Sci., 32 (4), 911 –919 (2017). https://doi.org/10.1007/s10103-017-2192-z Google Scholar

61. 

Y. Wang et al., “Photobiomodulation of human adipose-derived stem cells using 810nm and 980nm lasers operates via different mechanisms of action,” Biochim. Biophys. Acta, 1861 (2), 441 –449 (2017). https://doi.org/10.1016/j.bbagen.2016.10.008 BBACAQ 0006-3002 Google Scholar

62. 

U. Oron et al., “Attenuation of infarct size in rats and dogs after myocardial infarction by low-energy laser irradiation,” Lasers Surg. Med., 28 (3), 204 –211 (2001). https://doi.org/10.1002/lsm.1039 LSMEDI 0196-8092 Google Scholar

63. 

U. Oron et al., “Low-energy laser irradiation reduces formation of scar tissue after myocardial infarction in rats and dogs,” Circulation, 103 (2), 296 –301 (2001). https://doi.org/10.1161/01.CIR.103.2.296 CIRCAZ 0009-7322 Google Scholar

64. 

F. Salehpour et al., “Transcranial near-infrared photobiomodulation attenuates memory impairment and hippocampal oxidative stress in sleep-deprived mice,” Brain Res., 1682 36 –43 (2018). https://doi.org/10.1016/j.brainres.2017.12.040 BRREAP 0006-8993 Google Scholar

65. 

E. C. Leal Junior et al., “Effect of low-level laser therapy (GaAs 904 nm) in skeletal muscle fatigue and biochemical markers of muscle damage in rats,” Eur. J. Appl. Physiol., 108 (6), 1083 –1088 (2010). https://doi.org/10.1007/s00421-009-1321-1 EJAPFN 1439-6319 Google Scholar

66. 

E. C. Leal Junior et al., “Effects of low-level laser therapy (LLLT) in the development of exercise-induced skeletal muscle fatigue and changes in biochemical markers related to postexercise recovery,” J. Orthop. Sports Phys. Ther., 40 (8), 524 –532 (2010). https://doi.org/10.2519/jospt.2010.3294 Google Scholar

67. 

Q. Zhang et al., “Low-level laser therapy effectively prevents secondary brain injury induced by immediate early responsive gene X-1 deficiency,” J. Cereb. Blood Flow Metab., 34 (8), 1391 –1401 (2014). https://doi.org/10.1038/jcbfm.2014.95 Google Scholar

68. 

B. M. Baroni et al., “Effect of light-emitting diodes therapy (LEDT) on knee extensor muscle fatigue,” Photomed. Laser Surg., 28 (5), 653 –658 (2010). https://doi.org/10.1089/pho.2009.2688 Google Scholar

69. 

E. C. Leal Junior et al., “Comparison between cold water immersion therapy (CWIT) and light emitting diode therapy (LEDT) in short-term skeletal muscle recovery after high-intensity exercise in athletes--preliminary results,” Lasers Med. Sci., 26 (4), 493 –501 (2011). https://doi.org/10.1007/s10103-010-0866-x Google Scholar

70. 

N. J. Blanco, W. T. Maddox and F. Gonzalez-Lima, “Improving executive function using transcranial infrared laser stimulation,” J. Neuropsychol., 11 (1), 14 –25 (2017). https://doi.org/10.1111/jnp.2017.11.issue-1 Google Scholar

71. 

S. G. Disner, C. G. Beevers and F. Gonzalez-Lima, “Transcranial laser stimulation as neuroenhancement for attention bias modification in adults with elevated depression symptoms,” Brain Stimul., 9 (5), 780 –787 (2016). https://doi.org/10.1016/j.brs.2016.05.009 Google Scholar

72. 

M. T. Delpy and M. Cope, “Quantification in tissue near-infrared spectroscopy,” Phil. Trans. R. Soc. Lond. B, 352 649 –659 (1997). https://doi.org/10.1098/rstb.1997.0046 PTRBAE 0962-8436 Google Scholar

73. 

Y. Wang et al., “Red (660 nm) or near-infrared (810 nm) photobiomodulation stimulates, while blue (415 nm), green (540 nm) light inhibits proliferation in human adipose-derived stem cells,” Sci. Rep., 7 (1), 7781 (2017). https://doi.org/10.1038/s41598-017-07525-w SRCEC3 2045-2322 Google Scholar

74. 

R. A. Lopes-Martins et al., “Spontaneous effects of low-level laser therapy (650 nm) in acute inflammatory mouse pleurisy induced by carrageenan,” Photomed. Laser Surg., 23 (4), 377 –381 (2005). https://doi.org/10.1089/pho.2005.23.377 Google Scholar

75. 

M. Khadra et al., “Effect of laser therapy on attachment, proliferation and differentiation of human osteoblast-like cells cultured on titanium implant material,” Biomaterials, 26 (17), 3503 –3509 (2005). https://doi.org/10.1016/j.biomaterials.2004.09.033 BIMADU 0142-9612 Google Scholar

76. 

F. A. Al-Watban and B. L. Andres, “The effect of He-Ne laser (632.8 nm) and Solcoseryl in vitro,” Lasers Med. Sci., 16 (4), 267 –275 (2001). https://doi.org/10.1007/PL00011363 Google Scholar

77. 

F. Salehpour et al., “Therapeutic effects of 10-Hz pulsed wave lasers in rat depression model: a comparison between near-infrared and red wavelengths,” Lasers Surg. Med., 48 (7), 695 –705 (2016). https://doi.org/10.1002/lsm.v48.7 LSMEDI 0196-8092 Google Scholar

78. 

Q. Wu et al., “Low-level laser therapy for closed-head traumatic brain injury in mice: effect of different wavelengths,” Lasers Surg. Med., 44 218 –226 (2012). https://doi.org/10.1002/lsm.v44.3 LSMEDI 0196-8092 Google Scholar

79. 

P. de Almeida et al., “Low-level laser therapy improves skeletal muscle performance, decreases skeletal muscle damage and modulates mRNA expression of COX-1 and COX-2 in a dose-dependent manner,” Photochem. Photobiol., 87 (5), 1159 –1163 (2011). https://doi.org/10.1111/php.2011.87.issue-5 PHCBAP 0031-8655 Google Scholar

80. 

H. Zhang et al., “Low level laser irradiation precondition to create friendly milieu of infarcted myocardium and enhance early survival of transplanted bone marrow cells,” J. Cell Mol. Med., 14 (7), 1975 –1987 (2010). https://doi.org/10.1111/j.1582-4934.2009.00886.x Google Scholar

81. 

V. Prabhu et al., “Development and evaluation of fiber optic probe-based helium-neon low-level laser therapy system for tissue regeneration--an in vivo experimental study,” Photochem. Photobiol., 86 (6), 1364 –1372 (2010). https://doi.org/10.1111/php.2010.86.issue-6 PHCBAP 0031-8655 Google Scholar

82. 

P. Gal et al., “Effect of equal daily doses achieved by different power densities of low-level laser therapy at 635 nm on open skin wound healing in normal and corticosteroid-treated rats,” Lasers Med. Sci., 24 (4), 539 –547 (2009). https://doi.org/10.1007/s10103-008-0604-9 Google Scholar

83. 

F. A. Al-Watban and G. D. Delgado, “Burn healing with a diode laser: 670 nm at different doses as compared to a placebo group,” Photomed. Laser Surg., 23 (3), 245 –250 (2005). https://doi.org/10.1089/pho.2005.23.245 Google Scholar

84. 

J. T. Hashmi et al., “Effect of pulsing in low-level light therapy,” Lasers Surg. Med., 42 (6), 450 –466 (2010). https://doi.org/10.1002/lsm.v42:6 LSMEDI 0196-8092 Google Scholar

85. 

S. O. el Sayed and M. Dyson, “Effect of laser pulse repetition rate and pulse duration on mast cell number and degranulation,” Lasers Surg. Med., 19 (4), 433 –437 (1996). https://doi.org/10.1002/(SICI)1096-9101(1996)19:4<433::AID-LSM8>3.0.CO;2-T LSMEDI 0196-8092 Google Scholar

86. 

B. S. Sushko, P. Lymans’kyi Iu and S. O. Huliar, “[Action of the red and infrared electromagnetic waves of light-emitting diodes on the behavioral manifestation of somatic pain],” Fiziol Zh., 53 (3), 51 –60 (2007). Google Scholar

87. 

Y. Ueda and N. Shimizu, “Effects of pulse frequency of low-level laser therapy (LLLT) on bone nodule formation in rat calvarial cells,” J. Clin. Laser Med. Surg., 21 (5), 271 –277 (2003). https://doi.org/10.1089/104454703322564479 JCLSEO Google Scholar

88. 

R. Kilik et al., “Effect of equal daily doses achieved by different power densities of low-level laser therapy at 635 nm on open skin wound healing in normal and diabetic rats,” Biomed. Res. Int., 2014 1 –9 (2014). https://doi.org/10.1155/2014/269253 Google Scholar

89. 

R. R. Anderson and J. A. Parrish, “The optics of human skin,” J. Invest. Dermatol., 77 (1), 13 –19 (1981). https://doi.org/10.1111/1523-1747.ep12479191 JIDEAE 0022-202X Google Scholar

90. 

T. A. Henderson and L. D. Morries, “Near-infrared photonic energy penetration: can infrared phototherapy effectively reach the human brain?,” Neuropsychiatr. Dis. Treat., 11 2191 –2208 (2015). https://doi.org/10.2147/NDT Google Scholar

91. 

M. V. Padalkar and N. Pleshko, “Wavelength-dependent penetration depth of near infrared radiation into cartilage,” Analyst, 140 (7), 2093 –2100 (2015). https://doi.org/10.1039/C4AN01987C ANLYAG 0365-4885 Google Scholar

92. 

A. Gupta, T. Dai and M. R. Hamblin, “Effect of red and near-infrared wavelengths on low-level laser (light) therapy-induced healing of partial-thickness dermal abrasion in mice,” Lasers Med. Sci., 29 (1), 257 –265 (2014). https://doi.org/10.1007/s10103-013-1319-0 Google Scholar

93. 

Z. Zalevsky and M. Belkin, “Coherence and speckle in photomedicine and photobiology,” Photomed. Laser Surg., 29 (10), 655 –656 (2011). https://doi.org/10.1089/pho.2010.2939 Google Scholar

94. 

L. Hode, “The importance of the coherency,” Photomed. Laser Surg., 23 (4), 431 –434 (2005). https://doi.org/10.1089/pho.2005.23.431 Google Scholar

95. 

K. C. Smith, “Laser (and LED) therapy is phototherapy,” Photomed. Laser Surg., 23 (1), 78 –80 (2005). https://doi.org/10.1089/pho.2005.23.78 Google Scholar

96. 

V. Heiskanen and M. R. Hamblin, “Photobiomodulation: lasers vs light emitting diodes?,” Photochem. Photobiol. Sci., 17 (8), 1003 –1017 (2018). https://doi.org/10.1039/C8PP00176F PPSHCB 1474-905X Google Scholar

97. 

J. R. Bolton, I. Mayor-Smith and K. G. Linden, “Rethinking the concepts of fluence (UV Dose) and fluence rate: the importance of photon-based units—a systemic review,” Photochem. Photobiol., 91 (6), 1252 –1262 (2015). https://doi.org/10.1111/php.2015.91.issue-6 PHCBAP 0031-8655 Google Scholar

98. 

R. F. Menezes et al., “Assessment of the effect of laser photobiomodulation on periimplant bone repair through energy dispersive X-ray fluorescence a study in dogs,” Proc. SPIE, 9695 96950T (2016). https://doi.org/10.1117/12.2214262 PSISDG 0277-786X Google Scholar

Biography

Randa Zein graduated as a dental surgeon from Lebanese University and became a restorative and aesthetic specialist in Toulouse, France. She holds her master’s degree of laser dentistry in Genoa, Italy, and currently, she is enrolled in the PhD program of tissue engineering at Cardiff University, United Kingdom. Her field of interest is the use of technology to find new technical ways to treat Parkinson disease and tumor metastases.

Wayne Selting holds a BSc degree in electrical engineering and a MSc degree in biomedical engineering as well as a DDS. He is a former codirector of a postdoctoral master’s degree in laser dentistry at Ospedale San Martino, University of Genoa, Italy. His research interests are in tissue penetration of laser and LED treatment modalities as well as optimizing parameters for both laser surgical intervention and photobiomodulation.

Michael R. Hamblin is a principal investigator at the Wellman Center for Photomedicine at Massachusetts General Hospital and an associate professor at Harvard Medical School since 1994. His research interests are in photodynamic therapy for antimicrobial applications and the treatment of localized infections, and for the stimulation of antitumor immunity. In photobiomodulation, he works on cellular and molecular mechanisms, applications to Alzheimer’s and other brain diseases, pain, inflammation, and wound healing.

© 2018 Society of Photo-Optical Instrumentation Engineers (SPIE)
Randa Zein, Wayne Selting, and Michael R. Hamblin "Review of light parameters and photobiomodulation efficacy: dive into complexity," Journal of Biomedical Optics 23(12), 120901 (11 December 2018). https://doi.org/10.1117/1.JBO.23.12.120901
Received: 4 April 2018; Accepted: 14 November 2018; Published: 11 December 2018
Lens.org Logo
CITATIONS
Cited by 182 scholarly publications and 9 patents.
Advertisement
Advertisement
RIGHTS & PERMISSIONS
Get copyright permission  Get copyright permission on Copyright Marketplace
KEYWORDS
Tissues

Wound healing

In vitro testing

In vivo imaging

Bone

Brain

Photons

RELATED CONTENT


Back to Top