ArticlePDF AvailableLiterature Review

Shedding Light on a New Treatment for Diabetic Wound Healing: A Review on Phototherapy

Wiley
The Scientific World Journal
Authors:

Abstract and Figures

Impaired wound healing is a common complication associated with diabetes with complex pathophysiological underlying mechanisms and often necessitates amputation. With the advancement in laser technology, irradiation of these wounds with low-intensity laser irradiation (LILI) or phototherapy, has shown a vast improvement in wound healing. At the correct laser parameters, LILI has shown to increase migration, viability, and proliferation of diabetic cells in vitro; there is a stimulatory effect on the mitochondria with a resulting increase in adenosine triphosphate (ATP). In addition, LILI also has an anti-inflammatory and protective effect on these cells. In light of the ever present threat of diabetic foot ulcers, infection, and amputation, new improved therapies and the fortification of wound healing research deserves better prioritization. In this review we look at the complications associated with diabetic wound healing and the effect of laser irradiation both in vitro and in vivo in diabetic wound healing.
This content is subject to copyright. Terms and conditions apply.
Review Article
Shedding Light on a New Treatment for
Diabetic Wound Healing: A Review on Phototherapy
Nicolette N. Houreld
Laser Research Center, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
Correspondence should be addressed to Nicolette N. Houreld; nhoureld@uj.ac.za
Received  August ; Accepted October ; Published January 
Academic Editors: A. Schreiber and S. Ulisse
Copyright ©  Nicolette N. Houreld. is is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Impaired wound healing is a common complication associated with diabetes with complex pathophysiological underlying
mechanisms and oen necessitates amputation. With the advancement in laser technology, irradiation of these wounds with low-
intensity laser irradiation (LILI) or phototherapy, has shown a vast improvement in wound healing. At the correct laser parameters,
LILI has shown to increase migration, viability, and proliferation of diabetic cells in vitro; there is a stimulatory eect on the
mitochondria with a resulting increase in adenosine triphosphate (ATP). In addition, LILI also has an anti-inammatory and
protective eect on these cells. In light of the ever present threat of diabetic foot ulcers, infection, and amputation, new improved
therapies and the fortication of wound healing research deserves better prioritization. In this review we look at the complications
associated with diabetic wound healing and the eect of laser irradiation both in vitro and in vivo in diabetic wound healing.
1. Introduction
1.1. Diabetes and Wound Healing. Diabetes Mellitus (DM) is a
chronic metabolic disorder due to an absence of, insuciency
in, or resistance to insulin. Complications arise as a result
of elevated glucose levels and protein glycation and include
cardiovascular disease, retinopathy, nephropathy, angiopa-
thy,andneuropathy.Patientsaremorelikelytohavefoot
problemsduetobloodvesselandnervedamageandoen
suer from sensory loss. Small sores can develop on the feet
and oen go unnoticed. ese later develop into deeper ulcers
which become slow to heal, and further complications such
as infection arise which oen necessitate amputation due to
thespreadofinfectiontotheunderlyingtissueandbone.It
is estimated that –% of patients will develop foot ulcers
[,], of which % of these will require hospitalization to
treat these ulcers []. Around –% of these patients will
require lower limb amputation [] and around % of all non
traumatic amputations are as a result of DM []. To further
highlight the seriousness of diabetes associated lower-limb
amputations, the -year mortality rate following amputation
stands at –% [].
To control the development of lower-limb ulcers, patients
are required to check their feet daily, wear the correct
footwear, and regularly visit their health care provider. It
isestimatedthatmorethanmillionpeopleworldwide
suer from DM, and in , . million people died as a
result of DM []. DM and its associated complications impact
heavily on the patient, their family, health care departments,
and countries. e treatment of chronic wounds is becoming
more of a burden due to the increase in health care costs, an
aging population, and an increase in the incidence of diabetes
[].
Despite the huge amount of research into the underlying
pathogenesis of impaired diabetic wound healing, there is still
no clear answer and it appears to be a net result of micro-
and macrovascular disease [] and inadequate angiogenesis
(Figure ). Neuropathy and sensory loss have also been
recognized as a major cause of prolonged healing in diabetic
patients. In addition, advanced glycation end products also
contribute to the pathogenesis [], and hyperglycemia adds
to the oxidative stress.
ere is a decrease in wound strength, reduced angio-
genesis, and poor wound contraction [,]. In DM, there is
a disruption in clot formation and the inammatory phase
is dysregulated [,], oen with a prolonged and excessive
inammatory response. Hypoxia is associated with diabetic
wounds and further amplies the inammatory response [].
Hindawi Publishing Corporation
e Scientific World Journal
Volume 2014, Article ID 398412, 13 pages
http://dx.doi.org/10.1155/2014/398412
e Scientic World Journal
Impaired
diabetic
wound
healing
Neuropathy Ischemia Hypoxia
macrovascular disease Sensory loss
Hyperglycemia
Oxidative
stress
Inflammation Circulation Immunity
Angiogenesis
Infection
Fibroblast
migration
Advanced glycation
end products
Disruption in
extracellular
matrix
Micro- &
F : Some of the underlying pathogenesis of impaired diabetic wound healing.
Formation of the extracellular matrix (ECM) is a crucial
step in wound healing and provides structural integrity to
tissue. In diabetic wound healing, there is a malformation
of the ECM due to the disruption in ECM-growth factor
interactions and impaired migration and proliferation of
broblasts []. Collagen is an important component of
the ECM and is synthesized and maintained by a balance
between matrix synthesis and degradation. In DM, there is an
imbalance between matrix degrading enzymes, matrix met-
alloproteases (MMPs), their inhibitors, and tissue inhibitor
metalloproteinases (TIMPs). e loss of collagen which is
associated with diabetes can be due to decreased levels of its
synthesis, enhanced metabolism, or a combination of both
[]. Nonhealing diabetic foot wounds display elevated MMP
activity, with a - to -fold increase in MMP- and MMP-
[,]. Dysregulated cellular functions also play a part,
such as defective T cell immunity, leukocyte chemotaxis,
phagocytosis, and bactericidal capacity [].
Infection of diabetic ulcers remains a real problem. It
can become life threatening and is one of the most common
causes of lower-limb amputation and appropriate treatment
is essential [].WoundsarecommonlyinfectedwithPseu-
domonas aeruginosa and Staphylococcus []. Infection can
spread from one ulcer to another as the foot has several
intercommunicating compartments, and combined with sen-
sory loss patients can continue walking on these infected
ulcers further facilitating their spread []. Ischemia com-
plicates matters further by reducing defense mechanisms.
Administration of antibiotics has its own complications
especially with the emergence of antibiotic resistant bacteria,
poor arterial supply which aects antibiotic delivery, correct
duration of treatment, and toxicity and allergy to patients.
1.2. Treatment of Diabetic Wounds. Management of the dia-
betic foot is multidisciplinary and can become problematic.
Treatment is both local (treating of the diabetic foot) and
systemic (glycemic control). Treatment of the diabetic foot
is extensive and can encompass mechanical and surgical
debridement, management of the wound base, antibiotic
therapy to treat infection, revascularization, prophylactic foot
surgery, mechanical o-loading, accommodative orthotics,
andanalterationoffootwear[,]. Hyperbaric oxygen
therapy (HBOT), which entails delivering % oxygen at
pressures above one atmosphere, increases the amount of
oxygen dissolved in the blood and has been used to treat a
variety of wounds [,]. HBOT has been used as an adjunc-
tive treatment for diabetic foot ulcers; however its evidence
of eciency is limited []. Kaya and colleagues []treated
 patients with diabetic foot ulcers with HBOT. Sixty-three
percent of patients responded to treatment, while % showed
no improvement and % underwent amputations. Com-
plications associated with HBOT are not common but can
include claustrophobia, ear, sinus, or lung damage, temporary
worsening of short sightedness, and oxygen poisoning [].
A number of clinical applications have been found for
lasers in a variety of medical specialities and have been used
indentistry,dermatology,osteology,physiotherapy,acupunc-
ture, surgery, photodynamic cancer therapy, and chiropractic
and veterinary science. Lasers have also been used in the
treatment of chronic wounds, including diabetic ulcers.
2. Phototherapy
Phototherapy, also known as photobiomodulation, low-level
laser therapy (LLLT), involves the application of light (oen
e Scientic World Journal
laser light of a specic wavelength or a light emitting diode,
LED) to stimulate cellular processes.
e eects of phototherapy are chemical and not thermal.
Energy which is delivered to cells produces insignicant and
minimal temperature changes, typically in the range of .
.C[]. Cellular responses are the result of changes in
photoacceptor molecules, or chromophores. Photoacceptors
take part in cellular metabolism and are not connected to a
light response, such as chlorophyll which is a photoreceptor
[]. Once the photon energy is absorbed, the photoacceptor
assumes an electronically excited state [], which in turn
stimulates cellular metabolism [,]byactivatingor
deactivating enzymes which alter other macromolecules such
asDNAandRNA[,]. e energy which is absorbed
bythephotoacceptorcanbetransferredtoothermolecules
causing chemical reactions in the surrounding tissue; this
then gives rise to observable eects at a biological level [,
]. Photon energy is absorbed by the chromophores and
there is an increase in adenosine triphosphate (ATP) [,]
and cell membrane permeability, which leads to activation
of secondary messengers which in turn activate a cascade
of intracellular signals []. ere is also an increase in
mitochondrial membrane potential and proton gradient [].
e exact mechanisms of action following laser irradi-
ation are not well understood, and a number of theories
exist, the most studied and best understood being that of
cytochrome-c oxidase (cyt 𝑎/𝑎3),theterminalenzymeinthe
eukaryotic mitochondrial respiratory chain (complex IV).
Cytochrome c oxidase facilitates the transfer of electrons to
molecular oxygen. e end product of this complex is the pro-
duction of ATP. Cytochrome c oxidase has two heme moieties
(heme 𝑎and heme 𝑎3) and two redox-active copper sites (CuA
and CuB), and these are the possible absorbing chromophores
for visible red and near infrared (NIR) light [,,]. When
photon energy is absorbed by cytochrome c oxidase, there is
achangeinthemitochondrialredoxstateand/orpumping
of protons across the inner mitochondrial membrane []
and an increase in ATP synthesis. ere is also an increase
in intracellular calcium ([Ca2+]i) which stimulates DNA and
RNA synthesis [].IthasbeenspeculatedbyKaru[]
that photoirradiation may intensify the transfer of electrons
within cytochrome c oxidase by making more electrons
available. An increase in the transfer of electrons and protons
accelerates oxidative metabolism which ultimately leads to
increased ATP []. Photoirradiation causes the reduction
or oxidation of cytochrome c oxidase and is dependent
on the initial redox status of the enzyme at the time of
irradiation []. Silveira and colleagues [,]showed
that LILI produced an increase in mitochondrial complexes
I, II, III, and IV, as well as succinate dehydrogenase. Hu
and colleagues [] also found an increase in cytochrome
c oxidase activity and concluded there was a cascade of
reactions which altered cellular homeostasis. Houreld et
al., [] showed that irradiation of isolated mitochondria
resulted in an increase in cytochrome c oxidase (complex
IV) activity. ere is also an increase in the concentration
of active mitochondria in irradiated cells. Both eects lead
to an increase in ATP. e eect of laser irradiation on
the mitochondria at a transcriptional level was also investi-
gated, and there is evidence that that there is an upregulation
of genes involved in complexes I, IV, and V [](Figure ).
Asecondpossibilityisthelocalizedtransientheating
of the photoacceptor which may cause structural changes
and trigger mechanisms such as activation or inhibition of
enzymes []. Another theory is the release of nitric oxide
(NO) from reduced cytochrome c oxidase which reverses the
signalling consequences of excessive NO binding [,,],
as NO in very low concentrations inhibits cytochrome c
oxidase by competing with oxygen [,].
2.1. In Vitro Eects of Photoirradiation. A number of studies,
on various cell types, have shown positive eects of photoir-
radiation. Studies have been conducted on stem cells [],
keratinocytes [,], mast cells [,], broblasts [],
smooth muscle cells [], osteoblasts [,], and schwann
cells []tonamebutafew.
Impaired diabetic wound healing has been associated
with impaired cellular function, and there is a decrease
in cellular migration, proliferation, NO synthesis, growth
factors, and collagen synthesis. ere is also an increase in
proteinases that degrade the extracellular matrix and collagen
(MMPs) and cells appear to be stuck in the inammatory
phase of wound healing. e increase in oxidative stress also
leads to increased cell death. Laser irradiation in vitro has
shown that these cells respond in a favourable fashion, even
irradiation of diabetic cells (Tab l e ). ere is an increase
in cellular migration [,], proliferation [,,],
viability [,], collagen production [,,], ATP
[], mitochondria concentration [], cytochrome c oxidase
activity [], NO [], growth factors [,,,], and
gene regulation []. ere is also a decrease in MMPs [],
apoptosis [,,] and proinammatory cytokines [].
Irradiation of hypoxic cells has also shown favourable
eects, with an increase in ATP and cyclic adenosine
monophosphate (cAMP) [], proliferation [,], viability
[], transforming growth factor-𝛽(TGF-𝛽) [], intra-
cellular Ca2+ [] and mitochondrial membrane potential
[], and a decrease in apoptosis and the pro-inammatory
cytokine tumour necrosis factor alpha (TNF-𝛼)[]. Irra-
diation of hypoxic/ischemic cells resulted in reduced ROS,
which results in increased angiogenesis []. Laser irradiation
restores homeostasis of injured and stressed cells, resulting in
improved repair and wound healing.
Not all studies have shown positive eects. Pereira et al.
[]andMarquesetal.[] showed that laser irradiation of
broblast cells had no eect on the synthesis of procollagen.
In fact there were ultrastructural changes to the endoplasmic
reticulum which may have resulted in a disruption in protein
synthesis []. Damante et al. [] demonstrated that irra-
diation at  nm had no eect on basic broblast growth
factor (bFGF). Interestingly, irradiation of the same cells
at  nm signicantly increased bFGF. Hakki and Bozkurt
[] irradiated human gingival broblasts by dierent laser
parameters and found no increase in proliferation at each of
the parameters used. However, they did nd an increase in
e Scientic World Journal
R
O
S
R
O
S
R
O
SR
O
S
cAMP
cAMP
cAMP
cAMP
Gene
transcription
Transcription
factors
ATP
ATPATP
ATP
ATP
NO
NO
NO
NO
NO
Mitochondria
Nucleus
Visible red
Laser light
Proliferation
Migration
Growth factors
ECM deposition
Inflammation
Apoptosis
iCa2+
iCa2+
iCa2+
iCa2+
F : Laser light is absorbed by chromophores in the cell, mitochondria in the case of visible red light. is leads to an increase in
adenosine triphosphate (ATP), reactive oxygen species (ROS), nitric oxide (NO), and intracellular calcium (iCa2+). ere is an activation of
transcription factors which get translocated to the nucleus and activate gene transcription. is leads to increased cell survival and wound
healing.
the transcription of various growth factors, namely, insulin-
like growth factor (IGF), vascular endothelial growth factor
(VEGF), and transforming growth factor-beta (TGF-𝛽). Irra-
diation of hypertrophic scar-derived broblasts and normal
dermal broblasts at a wavelength of  nm and a uence
of . and J/cm2had an inhibitory eect []. Pereira and
colleagues [] found no benet when they irradiated human
dentalpulpstemcellsatnmusingvariousuencies(.,
., , and  J/cm2). Schwartz-Filho and colleagues []
showed that irradiation at a wavelength of  nm with a
density of , , or  J/cm2had no eect on osteogenic cell
growth or viability.
ese adverse eects and dierence can be explained
by dierences in laser parameters. e eects of laser irra-
diation are highly dependent on the laser parameters such
as wavelength, power density, and uence. Cells respond to
LILI in a dose- and wavelength-dependent manner, and the
number of exposures as well as the time between exposures
plays an important role [,]. Higher uencies have
a negative eect on cells, while too low uences have no
eect. e inuence of wavelength was demonstrated by
Gupta et al., [] who demonstrated that irradiation at 
andnmhadapositiveeectonwoundhealing,while
awavelengthofandnmhadnoeect.iscan
be explained by the absorption spectrum of chromophores
which absorb light at dierent wavelengths.
2.2. In Vivo Eects of Photoirradiation. Alimitednumberof
clinical studies have been done on diabetic wound healing
(Table ). A reason for the small number of randomized trials
may be due to ethical issues associated with doing human
clinical trials []. A number of studies using phototherapy
in animal models have been done (Table ).
Al-Watban [] irradiated Sprague-Dawley rats (𝑛=
893) to dierent wavelengths (, , , , , nm,
and – nm LED cluster) and uencies (, , ,
and  J/cm2). He showed that phototherapy at a wave-
length of  nm accelerated healing and was the best
for alleviating diabetic wounds and burn healing. It was
suggested that phototherapy with  nm should be given
three times a week at a uence of . J/cm2per dose
for the treatment of diabetic burn wounds or . J/cm2
e Scientic World Journal
T : Summary of in vitro and in vivo studies done on various cell types and animal models, respectively, using low level laser irradiation (LILI).
Species/cell type Study design Outcomes Author reference
In vitro studies
Diabetic wounded human
skin broblasts
Cells were irradiated at  nm with J/cmand incubated for 
or  h. Control cells received no laser irradiation.
Irradiation resulted in increased cellular migration, viability,
proliferation, and collagen production. Ayu k et al. []
Diabetic wounded and
hypoxic human skin
broblast cells (WS)
Cells were irradiated at nm with J/cmand incubated for or
 h. Control cells received no laser irradiation.
Irradiated diabetic wounded cells showed increased cellular
migration, viability, and proliferation and a decrease in apoptosis
(caspase /) and proinammatory cytokine interleukin (IL)-𝛽.
NuclearfactorkappaB(NF-𝜅B) also translocated into the nucleus.
Irradiated hypoxic cells regained their normal morphology and
showed an increase in cellular viability, proliferation, and IL- and
decreased apoptosis (caspase /) and proinammatory cytokine
tumor necrosis factor (TNF)-𝛼.NF-𝜅Balsotranslocatedintothe
nucleus.
Sekhejane et al. []
Human skin broblasts
(HSFs)
Cells were cultured in physiologic glucose (. mM/L) or high
glucose concentration (. and mM/L) and irradiated at
. nm with ., , and J/cmon consecutive days.
Densities of . and J/cmhad stimulatory eects on the viability
and proliferation rate of HSFs cultured in physiologic glucose.
Densities of ., , and J/cmhad stimulatory eects on the
proliferation rate of HSFs cultured in high glucose concentrations.
Esmaeelinejad et al.
[]
Diabetic wounded skin
broblast cells (WS)
Cells were irradiated at .nm with or  J/cm.Controlcells
received no laser irradiation.
Cells irradiated at J/cmshowed increased cellular migration and
proliferation, while cells irradiated at  J/cmshowed decreased
cellular migration and proliferation.
Houreld and
Abrahamse []
NIH T broblast cells
For proliferation studies, cells were grown in .% foetal bovine
serum (FBS) and irradiated at  nm. Cells received two
applications ( h interval) of J/cme ach ( J/cmtotal); J/cm
and then J/cm( J/cmtotal); J/cmand then J/cm( J/cm
total). Control cells received no laser irradiation. Cells were
incubated for , , , and days. For procollagen studies, cells were
grown in .% FBS and irradiated at  nm, J/cmand
incubated for days.
CellsirradiatedwithandJ/cm
showed increased cellular
proliferation. No signicant increase in procollagen was seen in
any of the irradiated cells.
Pereira et al. []
Murine broblast T
cells and primary human
keloid broblast cell
cultures
Cells were irradiated at  nm for consecutive days (, , and
 h) with or  J. For the MTT assay (proliferation), a power
density of . W/cmwas use d, while . W/cmwas used for
viability assays (Trypan blue).
A dose of J stimulated proliferation, while  J inhibited
proliferation of human keloid broblast cells. Laser irradiation is
aected by the physiological state of the cells; high-metabolic rate
and short-cell-cycle T cells were not responsive to LILI. A dose
of J reduced cell death but did not stimulate cell cycle. A dose of
 J had negative eects on the cells, as it increased cell death and
inhibited cell proliferation.
Frigo et al. []
HIG- rabbit synovial
broblasts
Cells were synchronized at G by serum starvation (.% FBS for
 h) and irradiated at  nm with ., ., or . J/cmand
cultured for another h. Control cells received no laser
irradiation.
Cellular proliferation was signicantly stimulated at . and
. J/cm, while no eect was observed at . J/cm. e proportion
of cells at S phase in the laser irradiation group (. J/cm)was
signicantly higher; thus LILI enhances cell cycle progression and
as it promotes synovial broblast proliferation.
Taniguchi et al. []
e Scientic World Journal
T : Cont i n u e d .
Species/cell type Study design Outcomes Author reference
Porcine primar y
aortic smooth muscle cells
(SMCs)
Cells were irradiated at nm with or J/cm.Cellswere
incubated for dierent time periods depending on the assay.
LILI stimulated porcine aortic SMC proliferation, increased
collagen synthesis, modulated activity and expression of matrix
metalloproteinase (MMP)-, gene expression of MMP-, and tissue
inhibitor of metalloproteinases (TIMP)-, and inhibited gene
expression of proinammatory cytokine IL-𝛽.
Gavish et al. []
Primary human gingival
broblasts (GF)
Cells were irradiated at nm with dierent settings used in
dentistry:power:W,pulseinterval:ms,pulselength:ms,
 s/cm,  J/cm(infected pocket setting); power: . W, pulse
interval:  ms, pulse length:  ms,  s/cm,  J/cm(Perio
pocket setting); power: . W in continuous wave,  s/cm,
J/cm(biostimulation setting).
No signicant dierence in proliferation was observed in the
dierent laser applications when compared to the control group.
Signicantly increased insulin-like growth factor (IGF) and
vascular endothelial growth factor (VEGF) mRNA was observed in
all irradiated groups. A signicant increase in collagen type I
mRNA expression was noted in only the biostimulation setting.
Hakki and Bozkurt
[]
Human foreskin broblast
HS cells
Cells were grown in % FBS for h and then irradiated with a
light emitting diode (LED) array (nm) with or J. Cells were
incubated for or days. Control cells received no laser irradiation.
A dose of J induced a signicant increase in viability. Irradiation
increased the mRNA expression level of type I collagen and also
aected basic broblast growth factor (bFGF) secretion levels.
Huang et al. []
Human dermal broblasts
LED array populated with  and  nm LEDs. e ratios of
visible to infrared (IR) light were decreased (in the case of visible)
and increased (in the case of IR) in series of % increments from
no IR to fully IR. Cells were incubated for  h.
Photomodulation with a /nm LED array in dierent ratios
has an eect on gene expression proles and is eective for altering
gene expression, collagen synthesis, and reduction of MMP-
expression.
McDaniel et al. []
Human gingival
broblasts, FMM cells
Cells were irradiated at  nm with J/cmand incubated for
days. Control cells received no laser irradiation.
Irradiation produced no dierence in the amount of procollagen
between groups, and the amount of type I collagen as well as the
total protein content was signicantly smaller in control cultures.
ere were also ultrastructural changes in cytoplasmic organelles,
especially the mitochondria and rough endoplasmic reticulum.
Marques et al. []
Diabetic and ischemic
skin broblast cells (WS)
Whole cells or isolated mitochondria were irradiated at nm
with or  J/cm. Control cells received no laser irradiation.
Irradiation of mitochondria with J/cmresulted in increased
adenosine triphosphate (ATP) production, a higher accumulation
of activated mitochondria in diabetic cells, an increase in complex
IV activity, and a decrease in complex III activity. ere was an
increase in complex IV activity in mitochondria and a higher
accumulation of activated mitochondria in diabetic cells irradiated
with J/cm. Irradiated ischemic cells showed no signicant
dierences compared to their nonirradiated control.
Houreld et al. []
Diabetic wounded skin
broblast cells (WS)
Cells were irradiated at nm with J/cm. Control cells received
no laser irradiation.Cells were incubated for min, , , or  h.
Irradiation resulted in increased cellular proliferation ( and
 h), nitric oxide ( min), and reactive oxygen species ( min)
and decreased apoptosis ( h), TNF-𝛼( and  h), and IL-𝛽
( h).
Houreld et al. []
Primary human gingival
broblasts (hGF)
Cells irradiated at  nm with J/cmand incubated for  h. Two
study groups, namely, cells which were irradiated once (single-dose
group)andcellswhichwereirradiatedtwicewithhinterval
(double dose). Control cells received no laser irradiation.
Cells in the single-dose group showed a signicant increase in
proliferation and growth factors bFGF and IGF-, with no change
in IGF-binding protein (IGFBP). Cells in the double dose group
showed a signicant increase in proliferation and growth factors
bFGF, IGF-, and IGFBP.
Saygun et al. []
e Scientic World Journal
T : Cont i n u e d .
Species/cell type Study design Outcomes Author reference
Human gingival broblast
cell line (FGH)
Cells were grown in % FBS for h and then irradiated in media
containing % FBS. Cells irradiated twice at  or nm with
or J/cmwith h between irradiations.
ere was no signicant dierence in the expression of
keratinocyte growth factor (KGF), while bFGF was signicantly
increased in cells irradiated at  nm (no dierence at  nm).
Damante et al. []
Wounded, diabetic
wounded, and ischemic
skin broblast cells (WS)
Cells were irradiated at  nm with J/cm. Control cells received
no laser irradiation. Cells were incubated for  min.
Irradiation upregulated the expression of mitochondrial genes
COXB (complex IV), COXC (complex IV), and PPA (complex
V) in diabetic wounded cells and ATPB (complex V) and
ATPG (complex V) in ischemic cells. COXC (complex IV),
ATPF (complex V), NDUFA (complex I), and NDUFS
(complex I) were upregulated in wounded cells.
Masha et al. []
Isolated mouse embryonic
broblasts
Cells irradiated at  nm with ., ., ., , or  J/cm.
Control cells received no laser irradiation.
A dose of ., , and  J/cmproduced an increase in reactive
oxygenspecies(ROS).NoincreaseinATPwasseenwith
. J/cm, a small increase was s een at . J/cmand a large
increase was seen with uencies of ., , and  J/cm.Adoseof
. J/cmincreased NF-𝜅B h aer irradiation. Activation of
NF-𝜅B is mediated via ROS generation.
Chen et al. []
In vivo studies
Rat, Sprague-Dawley,
diabetic (streptozotocin
induced), and nondiabetic
Full-thickness wound (. ±mm
)oraburn(±. mm)
was made on each rat. Rats were irradiated with various
wavelengths (, , , , and , nm) and polychromatic
LED clusters (–, –, –, –, and
nm)withadoseof,,,orJ/cm
three times per
week.
e best eects on wound and burn healing were exhibited with a
laser with a wavelength of  nm. Based on the results,
phototherapy at  nm, . J/cm, times/week is recommended
for diabetic burn wounds, and phototherapy at nm, . J/cm,
times/week for diabetic wounds is recommended for human
clinical trials.
Al-Watban []
Mice, diabetic
(BKS.Cg-m+/+Lepr db/J),
male and female
A full-thickness circular wound was made on the le ank in each
mouse using a sterile mm diameter skin punch, and the wound
extended down to the fascial layer over the abdominal
musculature. Wounds were irradiated at  nm, with , ., ., or
. J/day. Mice were euthanized on day .
Irradiation of splintered wounds at  nm with . J/day
(.–. J/cm/day) for days was shown to cause the maximal
stimulation of healing on day . Wounds healed mainly by
reepithelization and granulation tissue formation.
Chung et al. []
Rats, Wistar, diabetic
(streptozotocin induced),
and male
A×cmcutaneousapwasraisedonthedorsumofeach
animal. A plastic sheet was introduced between the ap and the
bed to impair blood supply, and the ap was then sutured. Rats
were treated transcutaneously every other day with  or  nm,
on  contact points at the wound margin (.J/cm/point; total of
 J/cm). Rats were euthanized on day .
e results suggest that the best responses of the aps were
observed on irradiated subjects, in particular those treated with
 nm. ere was increased angiogenesis, reduced tissue necrosis
and inammation, and increased broblastic proliferation.
Santos et al. []
Rats, Wistar, diabetic
(streptozotocin induced),
and male
Rats were divided into groups:
control (untreated, nondiabetic); laser (laser treated, nondiabetic);
diabetic (diabetic rats, nonlaser treated); and diabetic + laser
(diabetic rats laser treated). Scars were irradiated once at  nm
with J/cm, and rats were euthanized  h aer irradiation.
In untreated diabetic rats there was increased MMP- and MMP-
expression compared to untreated nondiabetic rats. Irradiation of
diabetic rats signicantly reduced MMP- and MMP- expression
compared to untreated diabetic rats, and there was also increased
production of collagen.
Aparecida et al. []
e Scientic World Journal
T : Cont i n u e d .
Species/cell type Study design Outcomes Author reference
Rats, Wistar, diabetic
(streptozotocin induced),
male
Full-thickness wounds were made in the hard palates using a mm
biopsy punch. Rats were divided into groups: control group
(nonirradiated) and experimental group (irradiated). Wounds
were irradiated at  nm with  J/cmaer surgery and on days
, , and aer surgery. Rats were euthanized on days , , and .
Irradiation resulted in decreased numbers of inammatory cells
and increased mitotic activity of broblasts, collagen synthesis, and
vascularization. Oxidative status was also signicantly decreased
on day .
Decreased inammatory cells, and oxidative stress and increased
collagen and vascularization Firat et al. []
Rat, Sprague-Dawley,
normal or diabetic
(streptozotocin induced),
male
Le and right maxillary rst molars were extracted, and extraction
sockets on the le were not irradiated, while the right ones were
irradiated at  nm with . J/cm. Rats were euthanized , , ,
or  days aer extraction.
Irradiation promoted new bone formation. In normal rats,
osteoblasts and osteoid tissue were observed at day , which was
earlier than in the control group, and new bone reached the top of
the extraction socket at day . In diabetic irradiated rats, less
inltration of inammatory cells and blood clots were observed at
day , and more new bone formed at days and  than in the
nonirradiated diabetic group. Laser irradiation stimulated the
dierentiation of osteoblasts and increased the expression of
collagen type I and osteocalcin mRNA.
Park and Kang []
Rats, Wistar, diabetic
(streptozotocin induced)
male
Rats were divided into groups: control (normoglycemic, no
injury), diabetic (no injury), sham (Normoglycemic, sham
irradiated), diabetic sham, nondiabetic cryoinjured submitted to
LLLT, diabetic cryoinjured submitted to LLLT, and diabetic
cryoinjured nontreated. Cryoinjury was carried out on the le
posterior leg: the muscle fascia was carefully removed, and the
tibialis anterior muscle was surgically exposed and cryoinjured for
 s with a cooled (in liquid nitrogen) round mm metal probe.
Aer the frozen muscle had thawed, the procedure was repeated
on the same area for another  s. Surgical wounds were closed
with sutures and rats were allowed recovering. Two hours aer
injury, the muscle was irradiated at  nm with J/cmto points
within the area (energy per point was . J, totalizing . J per
treatment). Irradiations were performed daily (h interval). Rats
that were euthanized on day received treatments, while rats
euthanized on day  received  treatments.
Diabetic animals that received LLLT exhibited morphological
aspects of skeletal muscle healing similar to those found in the
normoglycemic animals having received LLLT, with the
organization of immature bers in the collagen meshwork. e
diabetic sham irradiated group exhibited brosis. us, LLLT can
help avoid brosis and reduce muscle atrophy
Franc¸a et al. []
Double-blind, randomized,
placebo-controlled study.
Twenty patients with 
chronic lower extremity
venous ulcers
Inclusion criteria included the following: ulcer in the lower
extremity, () ulcers larger than . cm, () ulcer duration >wk,
() presence of classical signs of venous insuciency such as
edema, varicosities, lipodermatosclerosis, eczema, and
elephantiasis nostra, () and () controlled systemic arterial
hypertension (diastolic arterial pressure < mm Hg). Each group
of ulcers was treated x/week. Ulcers were covered with % silver
sulfadiazine (SDZ) cream, dressed, and then bandaged. Group
received placebo phototherapy; group were irradiated at  and
 nm (LEDs)  s per point until the entire ulcer surface was
treated with the probe; and the control group received standard
care without phototherapy. Ulcers were treated for a maximum of
 days.
Laser therapy increased wound healing. At all time points, light
treated ulcers healed faster than the control group treated with
SDZ cream dressing alone, as well as the placebo treatment group.
Caetano et al. []
e Scientic World Journal
T : Cont i n u e d .
Species/cell type Study design Outcomes Author reference
Double-blind, randomized
placebo-controlled,
experimental design, 
patients with  chronic
diabetic leg ulcers
Inclusion criteria: () diagnosis of type II diabetes independent of
glycemic control with neuropathic or mixed (venous and arterial)
ulcers, () ulcer located on the lower extremity, () ulcer present
for a minimum of weeks during which it has been either stable or
worsening, () willingness to participate in the study and
commitment to the follow-up protocol, and () signed written
consent. Ulcers were cleaned with .% physiological saline and
dried before phototherapy was applied twice per week for a
maximum of  days. Ulcers were dressed with % silver
sulfadiazine cream covered with gauze and bandaged. Ulcers in the
irradiated group were treated with  and  nm probes (LEDs)
 s per point until the entire ulcer surface was treated.
Laser irradiation using a combination of  and nm
promoted tissue granulation and rapid healing of diabetic ulcers
that failed to respond to other forms of treatment.
Minatel et al. []
Double-blind, randomized
controlled clinical trial, 
patients with chronic
diabetic ulcers
Patients having a diabetic foot ulcer for a minimum of  weeks
with ulcer stages I and II who were capable of giving informed
consent, understanding instructions, and cooperating with study
protocol were enrolled. Patients were divided into laser treated and
conventional therapy or conventional therapy alone (placebo
group). Ulcers were treated x/week for two successive weeks and
then every other day up to complete healing. Ulcers were treated
with a  nm laser at a dose of J/cm. Patients in the placebo
treatment group received sham irradiation.
Laser irradiation increased wound healing. Four weeks aer
beginning treatment, the size of ulcers was signicantly decreased
and by  weeks a greater number of patients in the irradiated
group showed complete healing than in the placebo group, and the
mean time of healing was lower.
Kaviani et al. []
 e Scientic World Journal
for diabetic wounds. Chung et al. [] treated full-thickness
circular wounds ( mm) in diabetic mice (type diabetes)
with a  nm laser to various uencies (– J/cm2)for
seven consecutive days. Wounds were splintered to minimize
contraction; the main process of healing would thus be
epithelization and granulation. On day  mice were eutha-
nized and the wound excised. At a uence between . and
 J/cm2, splinted irradiated wounds responded better and
healed quicker than nonirradiated splintered wounds. Santos
et al. [] irradiated cutaneous aps with poor circulation
in diabetic Wistar rats either with  or  nm (.J/cm2
per point). It was shown that angiogenesis was increased in
irradiated rats compared to control, non-irradiated rats, more
so when a wavelength of  nm was used.
Irradiation of diabetic male Wistar rats reduced the
expression of MMP and accelerated collagen production [].
Firat and colleagues []irradiatedfullthicknesswounds
in diabetic male Wistar rats with a nm diode laser
( J/cm2). Histopathological analysis revealed that there
was a decrease in inammatory cells and an increase in
collagen and vascularization. Blood tests showed that there
was a decrease in oxidative stress. LILI has been found to
promote healing in both so and hard tissue. Irradiation at
 nm has been shown to stimulate wound healing and
the formation of new bone []. Peplow and colleagues
[] demonstrated that the eect of irradiation is due to
cellular and biochemical changes in the wound environment,
rather than a hypoglycemic eect. One of the long-term
complications of diabetes includes musculoskeletal abnor-
malities, and it is a source of disability in these patients. Laser
irradiation ( nm, J/cm2) of cryoinjured diabetic male
Wistar rats showed improved muscle repair, with enhanced
reorganization of the myobers and the perimysium and
reduced brosis [].
Caetano et al. []conductedarandomizedplacebo-
controlled double-blind study on  patients with a total
of  chronic venous ulcers. Patients were divided into
three groups: in group one (placebo), ulcers were cleaned
with saline and treated with % silver sulfadiazine (SDZ)
cream and patients received a placebo phototherapy; in
group two ulcers were treated similarly and patients received
phototherapy (combined  nm and  nm at a uence of
J/cm
2); and in group three (controls) ulcers were treated
similarly and received no phototherapy. Patients that received
phototherapy responded to the laser treatment, and ulcers
healed signicantly faster than the control and placebo group,
particularly larger ulcers. Minatel et al. []showedthatcom-
bined irradiation with  and  nm promoted granulation
and healing of diabetic ulcers that failed to respond to other
forms of treatment. Kaviani et al. []performedarandom-
ized study on diabetic patients with foot ulcers that would
not respond to other treatments. Patients were divided into
two groups: group one received conventional treatment and
placebo irradiation, while group two received conventional
treatment and phototherapy ( nm,  J/cm2). e size of
theulcersinthephototherapytreatedgroupwassignicantly
smaller and the average time of healing was  weeks as
opposedtoweeksasobservedintheplacebogroup.
Infectionindiabeticwoundsisamajorproblem,and
eradication with antibiotics proves dicult due to decreased
blood ow. Laser irradiation has also been shown to inhibit
bacteria. Enwemeka et al. [] showed a dose-dependent
decrease in the number of methicillin-resistant Staphylo-
coccus aureus (MRSA) treated in vitro at a wavelength of
 nm (blue light). Irradiation at a wavelength of  nm was
suggested by Ankri and colleagues []intreatinginfected
wounds to clear the infection, followed by irradiation at
nmtospeedupthehealingprocess.isisanimportant
breakthrough as combined irradiation with visible red and
blue light can potentially be used to treat infected diabetic
ulcers.
3. Conclusion
DM is the leading cause for lower limb amputations. Current
treatments are challenging, lengthy, costly, and associated
with failure to heal and relapse. e patients quality of life
is aected, and a burden is placed on both patients and
caregivers. ere is a need to develop additional therapies
to treat diabetic ulcers. Due to its stimulatory eect and
no reported sideeects, laser therapy has been used to treat
chronic wounds, including diabetic ulcers. Phototherapy
has been shown to be benecial in treating diabetic ulcers
which are unresponsive to conventional treatments. is has
ledtoanimprovementinthequalityofpatientslives.By
studying the eects of LILI in vitro, the underlying mech-
anisms are being identied. e number of clinical studies
in DM is limited, and there is methodological heterogeneity
which explains the varied results seen. Better designed, well-
controlled, randomized, and double-blind studies are needed
for this type of therapy to become accepted and used as
an adjuvant therapy for the treatment of diabetic ulcers.
Phototherapy can be an important tool in speeding up the
healing process as well as alleviating pain and inammation.
ere is also a need to inform clinicians and other health care
providers of the benecial eects of phototherapy.
Conflict of Interests
e material in this research paper neither has been published
nor is being considered elsewhere for publication.
References
[] S. P. Pendsey, “Understanding diabetic foot, Inter national
JournalofDiabetesinDevelopingCountries,vol.,no.,pp.
–, .
[]N.Singh,D.G.Armstrong,andB.A.Lipsky,“Preventing
foot ulcers in patients with diabetes, Journal of the American
Medical Association,vol.,no.,pp.,.
[]A.L.Carrington,C.A.Abbott,J.Grithsetal.,“Afootcare
program for diabetic unilateral lower-limb amputees, Diabetes
Care,vol.,no.,pp.,.
[]A.Schindl,M.Schindl,H.Sch
¨
on,R.Knobler,L.Havelec,
and L. Schindl, “Low-intensity laser irradiation improves skin
circulation in patients with diabetic microangiopathy, Diabetes
Care, vol. , no. , pp. –, .
e Scientic World Journal 
[] G. E. Reiber, “Epidemiology of foot ulcers and amputations in
the diabetic foot, in e Diabetic Foot,J.H.BowkerandM.A.
Pfeifer,Eds.,pp.,Mosby,St.Louis,Mo,USA,.
[] World Health Organisation, “Diabetes, Fact sheet , ,
http://www.who.int/mediacentre/factsheets/fs/en/.
[] C.K.Sen,G.M.Gordillo,S.Royetal.,“Humanskinwounds:a
major and snowballing threat to public health and the economy:
perspective article, Wound Repair and Regeneration,vol.,no.
, pp. –, .
[] M.Peppa,P.Stavroulakis,andS.A.Raptis,“Advancedglycoxi-
dation products and impaired diabetic wound healing, Wo u nd
Repair and Regeneration,vol.,no.,pp.,.
[] K.N.Francis-Goforth,A.H.Harken,andJ.D.Saba,“Normal-
ization of diabetic wound healing, Surgery,vol.,no.,pp.
–, .
[] J. Chen, M. Kasper, T. Heck et al., “Tissue factor as a link
between wounding and tissue repair,” Diabetes,vol.,no.,
pp.,.
[] S. Guo and L. A. DiPietro, Critical review in oral biology &
medicine: factors aecting wound healing, Journal of Dental
Research,vol.,no.,pp.,.
[] D.OReilly,R.Linden,L.Fedorkoetal.,“Aprospective,double-
blind, randomized, controlled clinical trial comparing stan-
dard wound care with adjunctive hyperbaric oxygen therapy
(HBOT) to standard wound care only for the treatment of
chronic, non-healing ulcers of the lower limb in patients with
diabetes mellitus: a study protocol, Tri als,vol.,article,
.
[] N. J. Trengove, M. C. Stacey, S. Macauley et al., Analysis of the
acute and chronic wound environments: the role of proteases
and their inhibitors, Wound Repair and Regeneration,vol.,no.
, pp. –, .
[] J. L. Richard, A. Sotto, and J. P. Lavigne, New insights in
diabetic foot infection, Wound J o ur nal of Dia b e t es,vol.,no.
, pp. –, .
[]A.M.Eskes,D.T.Ubbink,M.J.Lubbers,C.Lucas,andH.
Vermeulen, “Hyperbaric oxygen therapy: solution for dicult
to heal acute wounds? Systematic review, Wor l d Jour n a l o f
Surgery, vol. , no. , pp. –, .
[] A. Kaya, F. Aydin, T. Altay, L. Karapinar, H. Ozturk, and C.
Karakuzu, “Can major amputation rates be decreased in dia-
betic foot ulcers with hyperbaric oxygen therapy?” Inter national
Orthopaedics,vol.,no.,pp.,.
[] A. J. Nemeth, “Las ers and wound healing,” Dermatologic Clinics,
vol. , no. , pp. –, .
[] T. Karu, “Primary and secondary mechanisms of action of
visible to near-IR radiation on cells, JournalofPhotochemistry
and Photobiology B,vol.,no.,pp.,.
[] J. Tuner and L. Hode, Laser erapy. Clinical Practice and
Scientic Background,PrimaBooks,Gr
¨
angesberg, Sweden,
.
[] A. L. Barbosa Pinheiro, S. Carneiro Nascimento, A. L. De Barros
Vieira et al., “Eects of low-level laser therapy on malignant
cells: in vitro study,” Journal of Clinical Laser Medicine and
Surgery,vol.,no.,pp.,.
[] M. Mati´
c, B. Lazeti´
c, M. Poljacki, V. Duran, and M. Ivkov-Simi´
c,
“Low level laser irradiation and its eect on repair processes in
the skin, Medicinski Pregled,vol.,no.-,pp.,.
[] S. Takac and S. Stojanovi´
c, “Diagnostic and biostimulating
lasers, Medicinski Pregled,vol.,no.-,pp.,.
[] T. I. Karu, “Primary and secondary mechanisms of the action of
monochromatic visible and near infrared radiation on cells, in
e Science of Low-Power Laser erapy,T.I.Karu,Ed.,pp.
, Gordon and Breach Science, Amsterdam, e Netherlands,
.
[] R. Lubart, H. Friedmann, I. Peled, and N. Grossman, Light
eect on broblast proliferaton,” Laser erapy,vol.,no.,pp.
–, .
[] P.C.L.Silveira,L.A.D.Silva,D.B.Fraga,T.P.Freitas,E.L.
Streck, and R. Pinho, “Evaluation of mitochondrial respiratory
chain activity in muscle healing by low-level laser therapy,
Journal of Photochemistry and Photobiology B,vol.,no.,pp.
–, .
[] D. H. Evans and H. Abrahamse, A review of laboratory-based
methods to investigate second messengers in low-level laser
therapy (LLLT), Medical Laser Application,vol.,no.,pp.
–, .
[] T. I. Karu, “Multiple roles of cytochrome c oxidase in mam-
malian cells under action of red and IR-A radiation, IUBMB
Life,vol.,no.,pp.,.
[] T.I.Karu,L.V.Pyatibrat,S.F.Kolyakov,andN.I.Afanasyeva,
Absorptionmeasurementsofcellmonolayersrelevantto
mechanisms of laser phototherapy: reduction or oxidation
of cytochrome c oxidase under laser radiation at .nm,
Photomedicine and Laser Surgery,vol.,no.,pp.,
.
[]P.C.L.Silveira,E.L.Streck,andR.A.Pinho,“Evaluation
of mitochondrial respiratory chain activity in wound healing
by low-level laser therapy,” Journal of Photochemistry and
Photobiology B, vol. , no. , pp. –, .
[] W.-P. Hu, J.-J. Wang, C.-L. Yu, C.-C. E. Lan, G.-S. Chen, and H.-
S. Yu, “Helium-neon laser irradiation stimulates cell prolifera-
tion through photostimulator y eects in mitochondr ia, Journal
of Investigative Dermatology,vol.,no.,pp.,.
[] N. N. Houreld, R. T. Masha, and H. Abrahamse, “Low-intensity
laser irradiation at  nm stimulates cytochrome c oxidase in
stressed broblast cells, Lasers in Surgery and Medicine,vol.,
pp.,.
[] R. T. Masha, N. N. Houreld, and H. Abrahamse, “Low-Intensity
Laser Irradiation at nm Stimulates Transcription of Genes
Involved in the Electron Transport Chain, Photomedicine and
Laser Surgery,vol.,no.,pp.,.
[] T. I. Karu, L. V. Pyatibrat, and N. I. Afanasyeva, Cellular eects
of low power laser therapy can be mediated by nitric oxide,
Lasers in Surgery and Medicine,vol.,no.,pp.,.
[]M.G.Mason,P.Nicholls,M.T.Wilson,andC.E.Cooper,
“Nitric oxide inhibition of respiration involves both competitive
(heme) and noncompetitive (copper) binding to cytochrome c
oxidase, Proceedings of the National Academy of Sciences of the
United States of America,vol.,no.,pp.,.
[] J.-Y. Wu, C.-H. Chen, C.-Z. Wang, M.-L. Ho, M.-L. Yeh,
and Y. -H. Wang, “Low-power laser irradiation suppresses
inammatory response of human adipose-derived stem cells by
modulation intracellular cyclic AMP level and NF-𝜅Bactivity,
PLoS ONE,vol.,no.,ArticleIDe,.
[]K.Choi,B.J.Kang,H.Kimetal.,“Low-levellasertherapy
promotes the osteogenic potential of adipose-derived mes-
enchymal stem cells seeded on an acellular dermal matrix,
Journal of Biomedical Materials Research Part B,vol.,no.,
pp.,.
[] M. Giannelli, F. Chellini, C. Sassoli et al., “Photoactivation
of bone marrow mesenchymal stromal cells with diode laser:
 e Scientic World Journal
eects and mechanisms of action, Journal of Cellular Physiol-
ogy,vol.,no.,pp.,.
[] J. A. de Villiers, N. N. Houreld, and H. Abrahamse, “Inuence
of low intensity laser irradiation on isolated human adipose
derived stem cells over  hours and their dierentiation
potential into smooth muscle cells using retinoic acid, Stem Cell
Reviews and Reports,vol.,no.,pp.,.
[] F.G.Basso,C.F.Oliveira,C.Kurachi,J.Hebling,andC.A.
D. S. Costa, “Biostimulatory eect of low-level laser therapy on
keratinocytes in vitro, Lasers in Medical Science,vol.,no.,
pp. –, .
[] L.Gavish,Y.Asher,Y.Becker,andY.Kleinman,“Lowlevellaser
irradiation stimulates mitochondrial membrane potential and
disperses subnuclear promyelocytic leukemia protein, Lasers in
Surgery and Medicine,vol.,no.,pp.,.
[] F. F. Fathabadie, M. Bayat, A. Amini, M. Bayat, and F. Rezaie,
“Eects of pulsed infra-red low level-laser irradiation on mast
cells number and degranulation in open skin wound healing
of healthy and streptozotocin-induced diabetic rats, Journal of
Cosmetic and Laser erapy,vol.,no.,pp.,.
[] A. Khoshvaghti, M. Zibamanzarmofrad, and M. Bayat, “Eect
of low-level treatment withan -Hz pulsed infrared diode laser
on mast-cell numbers and degranulation in a rat model of third-
degree burn, Photomedicine and Laser Surgery,vol.,no.,pp.
–, .
[] F. G. Basso, T. N. Pansani, A. P. Turrioni, V. S. Bagnato, J.
Hebling, and C. A. de Souza Costa, “In vitro wound healing
improvement by low-level laser therapy application in cultured
gingival broblasts, International Journal of Dentistry,vol.,
ArticleID,pages,.
[] M. Esmaeelinejad, M. Bayat, H. Darbandi, M. Bayat, and N.
Mosaa, “e eects of low-level laser irradiation on cellular
viability and proliferation of human skin broblasts cultured in
high glucose mediums , Lasers in Medical Science.Inpress.
[]S.M.Ayuk,N.N.Houreld,andH.Abrahamse,“Collagen
production in diabetic wounded broblasts in response to low-
intensity laser irradiation at  nm,” Diabetes Technology &
erapeutics, vol. , no. , pp. –, .
[] P. R. Sekhejane, N. N. Houreld, and H. Abrahamse, “Irradiation
at  nm positively aects diabetic wounded and hypoxic cells
in vitro, Photomedicine and Laser Surgery,vol.,no.,pp.
, .
[] N. Houreld and H. Abrahamse, “In vitro exposure of wounded
diabetic broblast cells to a helium-neon laser at and J/cm2
,” Photomedicine and Laser Surgery,vol.,no.,pp.,
.
[] I. L. Zungu, D. Hawkins Evans, N. Houreld, and H. Abrahamse,
“Biological responses of injured human skin broblasts toass ess
the ecacy of in vitro models for cell stress studies, Alliance
Journal of Business Research,vol.,no.,pp.,.
[] L. Gavish, L. Perez, and S. D. Gertz, “Low-level laser irradiation
modulates matrix metalloproteinase activity and gene expres-
sioninporcineaorticsmoothmusclecells,Lasers in Surgery
and Medicine, vol. , no. , pp. –, .
[] S. Hamajima, K. Hiratsuka, M. Kiyama-Kishikawa et al., “Eect
of low-level laser irradiation on osteoglycin gene expression in
osteoblasts, Lasers in Medical Science,vol.,no.,pp.,
.
[] S.-J. Pyo, W.-W. Song, I.-R. Kim et al., “Low-level laser therapy
induces the expressions of BMP-, osteocalcin, and TGF-𝛽in
hypoxic-cultured human osteoblasts, Lasers in Medical Science,
vol. , no. , pp. –, .
[] S. O. Yazdani, A. F. Golestaneh, A. Shaee, M. Hazi, H.-A. G.
Omrani, and M. Soleimani, “Eects of low level laser therapy
on proliferation and neurotrophic factor gene expression of
human schwann cells in vitro, Journal of Photochemistry and
Photobiology B,vol.,no.,pp.,.
[] N. Houreld and H. Abrahamse, “Irradiation with a . nm
helium-neon laser with J/cm2stimulates proliferation and
expression of interleukin- in diabetic wounded broblast
cells, Diabetes Technology and erapeutics,vol.,no.,pp.
–, .
[] A. N. Pereira, C. De Paula Eduardo, E. Matson, and M. M.
Marques, “Eect of low-power laser irradiation on cell growth
and procollagen synthesis of cultured broblasts, Lasers in
Surgery and Medicine, vol. , no. , pp. –, .
[] L. Frigo, G. M. F´
avero, H. J. Campos Lima et al., “Low-
level laser irradiation (InGaAIP- nm) increases broblast
cell proliferation and reduces cell death in a dose-dependent
manner, Photomedicine and Laser Surgery,vol.,no.,pp.
S–S, .
[] D. Taniguchi, P. Dai, T. Hojo, Y. Yamaoka, T. Kubo, and T.
Takamatsu, “Low-energy laser irradiation promotes synovial
broblast proliferation by modulating p subcellular localiza-
tion, Lasers in Surgery and Medicine,vol.,no.,pp.,
.
[] S. S. Hakki and S. B. Bozkurt, “Eects of dierent setting of
diode laser on the mRNA expression of growth factors and
type I collagen of human gingival broblasts, Lasers in Medical
Science,vol.,no.,pp.,.
[] P.-J. Huang, Y.-C. Huang, M.-F. Su, T.-Y. Yang, J.-R. Huang, and
C.-P. Jiang, “In vitro observations on the inuence of copper
peptide aids for the LED photoirradiation of broblast collagen
synthesis, Photomedicine and Laser Surgery,vol.,no.,pp.
–, .
[] D. H. McDaniel, R. A. Weiss, R. G. Geronemus, C. Mazur, S.
Wilson, and M. A. Weiss, “Varying ratios of wavelengths in
dual wavelength LED photomodulation alters gene expression
proles in human skin broblasts,” Lasers in Surgery and
Medicine,vol.,no.,pp.,.
[] N. N. Houreld, P. R. Sekhejane, and H. Abrahamse, “Irradiation
at  nm stimulates nitric oxide production and inhibits pro-
inammatory cytokines in diabetic wounded broblast cells,
Lasers in Surgery and Medicine,vol.,no.,pp.,.
[] I. Saygun, S. Karacay, M. Serdar, A. U. Ural, M. Sencimen,
and B. Kurtis, “Eects of laser irradiation on the release of
basic broblast growth factor (bFGF), insulin like growth
factor- (IGF-), and receptor of IGF- (IGFBP) from gingival
broblasts, Lasers in Medical Science,vol.,no.,pp.,
.
[] C. A. Damante, G. De Micheli, S. P. H. Miyagi, I. S. Feist, and
M. M. Marques, “Eect of laser phototherapy on the release of
broblast growth factors by human gingival broblasts, Lasers
in Medical Science,vol.,no.,pp.,.
[] A. C.-H. Chen, P. R. Arany, Y.-Y. Huang et al., “Low-Level
laser therapy activates NF-kB via generation of reactive oxygen
species in mouse embryonic broblasts, PLoS ONE,vol.,no.
, Article ID e, .
[] I. L. Zungu, D. Hawkins Evans, and H. Abrahamse, “Mitochon-
drial responses of normal and injured human skin broblasts
following low level laser irradiation—an in vitro study,” Photo-
chemistry and Photobiology,vol.,no.,pp.,.
[] W. B. Lim, J. S. Kim, Y. J. Ko et al., “Eects of nm light-
emitting diode irradiation on angiogenesis in CoCl2-exposed
e Scientic World Journal 
HUVECs, Lasers in Surgery and Medicine,vol.,no.,pp.
–, .
[] M. M. Marques, A. N. Pereira, N. A. Fujihara, F. N. Nogueira,
andC.P.Eduardo,“Eectoflow-powerlaserirradiation
on protein synthesis and ultrastructure of human gingival
broblasts, Lasers in Surgery and Medicine,vol.,no.,pp.
–, .
[] C. Webb and M. Dyson, “e eect of  nm low level laser
energy on human broblast cell numbers: a possible role in
hypertrophic wound healing, Journal of Photochemistry and
Photobiology B,vol.,no.,pp.,.
[] L. O. Pereira, J. P. F. Longo, and R. B. Azevedo, Laser irradiation
did not increase the proliferation or the dierentiation of stem
cells from normal and inamed dental pulp, Archives of Oral
Biology,vol.,no.,pp.,.
[] H. O. Schwartz-Filho, A. C. Reimer, C. Marcantonio, E. Mar-
cantonioJr.,andR.A.C.Marcantonio,“Eectsoflow-levellaser
therapy ( nm) at dierent doses in osteogenic cell cultures,
Lasers in Medical Science,vol.,no.,pp.,.
[] N. N. Houreld and H. Abrahamse, “Eectiveness of helium-
neon laser irradiation on viability and cytotoxicity of diabetic-
wounded broblast cells, Photomedicine and Laser Surgery,vol.
, no. , pp. –, .
[] D. Hawkins and H. Abrahamse, “Eect of multiple exposures
of low-level laser therapy on the cellular responses of wounded
human skin broblasts,” Photomedicine and Laser Surgery,vol.
, no. , pp. –, .
[] D. H. Hawkins and H. Abrahamse, “e role of laser uence in
cell viability, proliferation, and membrane integrity of wounded
human skin broblasts following Helium-Neon laser irradia-
tion, Lasers in Surgery and Medicine,vol.,no.,pp.,
.
[] A. Gupta, T. Dai, and M. R. Hamblin, “Eect of red and near-
infrared wavelengths on low-level laser (light) therapy-induced
healing in partial-thickness dermal abrasion in mice , Lasers in
Medical Science.Inpress.
[] F. A. H. Al-Watban, “Laser therapy converts diabetic wound
healingtonormalhealing,Photomedicine and Laser Surgery,
vol. , no. , pp. –, .
[]T.-Y.Chung,P.V.Peplow,andG.D.Baxter,“Laserphoto-
biostimulation of wound healing: dening a dose response
for splinted wounds in diabetic mice, Lasers in Surgery and
Medicine,vol.,no.,pp.,.
[] N. R. S. Santos, J. N. Dos Santos, J. A. Dos Reis et al., Inuence
of the use of laser phototherapy (𝜆 or  nm) on the
survivalofcutaneousapsondiabeticrats,Photomedicine and
Laser Surgery, vol. , no. , pp. –, .
[] A. Aparecida Da Silva, E. C. Leal-Junior, A. C. Alves et al.,
“Wound-healing eects of low-level laser therapy in diabetic
rats involve the modulation of MMP- and MMP- and the
reduction of collagen types I and III, Journal of Cosmetic and
Laser erapy,vol.,no.,pp.,.
[] E. T. Firat, A. Da˘
g, A. G˝
unay et al., “e eects of low-level laser
therapy on palatal mucoperiosteal wound healing and oxidative
stress status in experimental diabetic rats, Photomedicine and
Laser Surgery, vol. , no. , pp. –, .
[] J. J. Park and K. L. Kang, “Eect of -nm GaAIAs diode laser
irradiation on healing of extraction sockets in streptozotocin-
induced diabetic rats, a pilot study, Lasers in Medical Science,
vol. , pp. –, .
[] C. M. Franc¸a,C.deLouraSantana,C.B.Takahashietal.,“Eect
of laser therapy on skeletal muscle repair process in diabetic
rats, Lasers in Medical Science,vol.,no.,pp.,.
[] K. S. Caetano, M. A. C. Frade, D. G. Minatel, L. ´
A. Santana, and
C. S. Enwemeka, “Phototherapy improves healing of chronic
venous ulcers, Photomedicine and Laser Surgery,vol.,no.,
pp. –, .
[] D. G. Minatel, M. A. C. Frade, S. C. Franc¸a, and C. S. Enwemeka,
“Phototherapy promotes healing of chronic diabetic leg ulcers
that failed to respond to other therapies, Lasers in Surgery and
Medicine,vol.,no.,pp.,.
[] A. Kaviani, G. E. Djavid, L. Ataie-Fashtami et al., A random-
ized clinical trial on the eect of low-level laser therapy on
chronic diabetic foot wound healing: a preliminary report,
Photomedicine and Laser Surgery,vol.,no.,pp.–,.
[] P. V. Peplow, T.-Y. Chung, and G. D. Baxter, “Laser photo-
stimulation ( nm) of wound healing in diabetic mice is not
brought about by ameliorating diabetes, Lasers in Surgery and
Medicine,vol.,no.,pp.,.
[]C.S.Enwemeka,D.Williams,S.K.Enwemeka,S.Hollosi,
and D. Yens, “Blue -nm light kills Methicillin-Resistant
Staphylococcus aureus (MRSA) in vitro,” Photomedicine and
Laser Surgery, vol. , no. , pp. –, .
[] R. Ankri, R. Lubart, and H. Taitelbaum, “Estimation of the
optimal wavelengths for laser-induced wound healing, Lasers
in Surgery and Medicine,vol.,no.,pp.,.
... Wound healing involves a wide variety of cellular and molecular activities that are aimed at repairing the damaged tissue. Delayed wound healing is linked to impaired biological mechanisms which reduce cellular migration and proliferation [9]. Several signalling pathways such as PI3K/AKT/mTOR and growth factors, including vascular endothelial growth factor (VEGF) are involved in the process of wound healing. ...
... Photobiomodulation (PBM) refers to a form of therapy that is based on the use of light at a specific wavelength. PBM delivers photon energy that has therapeutic benefits such as accelerating the healing of diabetic wounds, decreasing inflammation and oedema, and reducing pain [9]. PBM has demonstrated to be the most effective in the visible red and near-infrared (NIR) electromagnetic spectrum, however the precise underlying cellular and molecular mechanisms of action leading to the observed effects is not entirely understood [13]. ...
... These wounds frequently develop into severe deep chronic ulcers that are difficult to treat and patients may require hospitalisation [35]. This is due to colonisation of bacteria at the wound site resulting in impaired wound healing, and frequently necessitates amputation [9]. Diabetic wounds are mostly characterised by a persistent inflammatory phase, as well as reduced production of granulation tissue and wound tensile strength [33]. ...
Article
Full-text available
Wound healing involves a series of cellular and molecular processes to heal injured tissue. Growth factors such as vascular endothelial growth factor (VEGF), and signalling pathways such as phosphatidylinositol 3-kinase, protein kinase B, and mammalian target of rapamycin (PI3K/AKT/mTOR) are essential in wound healing. VEGF is linked to intracellular signalling pathways including PI3K/AKT/mTOR, which controls cell growth, metabolism, proliferation, apoptosis, and protein synthesis. During photobiomodulation (PBM), low-level light in the visible red and near-infrared (NIR) spectrum is employed to promote healing, and reduce pain, inflammation, and oedema. Several studies demonstrate that PBM enhances cellular survival, proliferation, migration, and viability in vitro, however, the exact cellular and molecular mechanisms responsible for these benefits have not yet been identified. The aim of this review is to explore the effects of PBM on the PI3K/AKT/mTOR signalling pathway in wound healing.
... PBMT stimulates cells with a sufficiently low-energy density to avoid thermal damage to tissues, triggering anti-inflammatory, analgesic, and modulatory effects 12-15 . It is suggested that the effects of laser therapy are predominantly mediated by photoactivation cellular mechanisms, primarily through the absorption of light by mitochondrial cytochrome c oxidase 12,15,16 . This absorption triggers cellular signaling that stimulates protein synthesis and cell proliferation 12,15,16 . ...
... It is suggested that the effects of laser therapy are predominantly mediated by photoactivation cellular mechanisms, primarily through the absorption of light by mitochondrial cytochrome c oxidase 12,15,16 . This absorption triggers cellular signaling that stimulates protein synthesis and cell proliferation 12,15,16 . It is important to note that the underlying mechanisms of the therapeutic effects of laser therapy on analgesia and tissue repair are not fully understood yet, although recent studies have corroborated its efficacy in treating various clinical conditions 17,18 . ...
Article
Full-text available
Purpose: To conduct a systematic review of the mechanisms of photobiomodulation therapy (PBMT) for treating or preventing oral mucositis (OM) caused by antineoplastic therapy. Methods: Following PRISMA 2020 guidelines, a search was conducted in Medline, Latin American and Caribbean Health Sciences Literature (LILACS), Scientific Electronic Library Online (SciELO), and Bibliografia Brasileira de Odontologia from August to September 2023 using descriptors related to OM and laser therapy. Studies on the mechanisms of photobiomodulation in OM were included. Randomized (RCTs) or non-randomized trials from the past 10 years were reviewed. Risk of bias was assessed using RoB 2.0 and ROBINS-I tools. Results: A total of 355 studies was identified. After the screening, seven met the eligibility criteria. The RCTs showed a low risk of bias. PBMT reduced OM incidence in patients undergoing chemotherapy/radiotherapy. PBMT decreased pro-inflammatory cytokines (interleukin-6, tumor necrosis factor-α) and increased anti-inflammatory cytokines (interleukin-4, interleukin-10). It also modulated inflammatory mediators, enhancing the antioxidant enzyme superoxide dismutase and overexpressing genes for keratinocyte differentiation, aiding injury repair. Conclusion: The findings suggested that the mechanism of action of PBMT in OM involves modulation of the inflammatory response, balancing oxygen reactive species generation, and expression of factors related to healing or repair. Further studies are needed to elucidate these mechanisms and optimize treatment protocols. Key words Stomatitis; Low-Level Light Therapy; Drug Therapy; Radiotherapy
... Also, studies have state that, the reconciliation of mitochondrial signaling components like mitochondrial membrane potential (MMP or ΔΨm), mitochondrial ROS and Ca 2+ in under-irradiation cells, and the responses to irradiation in the nucleus (rise in DNA and RNA synthesis, expression of genes) are well documented [19]. Several studies have announced the effects related to enhanced mitochondrial activity, including rising ATP levels, ROS levels, and MMP ensuing the light irradiation [20,21]. ...
Article
Around 7% of the male population in the world are entangle with considerable situation which is known as male infertility. Photobiomodulation therapy (PBMT) is the application of low-level laser radiation, that recently used to increase or promote the various cell functions including, proliferation, differentiation, ATP production, gene expressions, regulation of reactive oxygen spices (ROS), and also boost the tissue healing and reduction of inflammation. This systematic review's main idea is a comprehensive appraisal of the literatures on subjects of PBMT consequences in four light ranges wavelength (blue, green, red, near-infrared (NIR)) on sperm cell characteristics, in vitro and in vivo. In this study, PubMed, Google Scholar, and Scopus databases were used for abstracts and full-text scientific papers published from 2003–2023 that reported the application of PBM on sperm cells. Criteria’s for inclusion and exclusion to review were applied. Finally, the studies that matched with our goals were included, classified, and reported in detail. Also, searched studies were subdivided into the effects of four ranges of light irradiation, including the blue light range (400–500 nm), green light range (500–600 nm), red light range (600–780 nm), and NIR light range (780–3000 nm) of laser irradiation on human or animal sperm cells, in situations of in vitro or in vivo. Searches with our keywords results in 137 papers. After primary analysis, some articles were excluded because they were review articles or incomplete and unrelated studies. Finally, we use the 63 articles for this systematic review. Our category tables were based on the light range of irradiation, source of sperm cells (human or animal cells) and being in vitro or in vivo. Six% of publications reported the effects of blue, 10% green, 53% red and 31% NIR, light on sperm cell. In general, most of these studies showed that PBMT exerted a positive effect on the sperm cell motility. The various effects of PBMT in different wavelength ranges, as mentioned in this review, provide more insights for its potential applications in improving sperm characteristics. PBMT as a treatment method has significant effectiveness for treatment of different medical problems. Due to the lack of reporting data in this field, there is a need for future studies to assessment the biochemical and molecular effects of PBMT on sperm cells for the possible application of this treatment to the human sperm cells before the ART process.
... This combination promotes the reduction of free radicals in injured tissue, angiogenesis, and other benefits [5]. Accordingly, it accelerates wound healing, closing lesions faster than conventional treatments [6][7][8][9]. We analyzed the outcomes of this case study using translational research to assess the knowledge generated from basic research and its clinical application until its market availability [10,11]. ...
... Associated with the attenuation of the signaling pathway triggered by IL-1β, studies describe PBM as a therapy capable of modulating the acute inflammatory process by modulating the expression of COX2; in addition, a clinical study reported the effect of the 660-nm laser on diabetic wounds in reducing the gene expression of inflammatory cytokines and consequent epithelial contraction [32]. According to Houreld [33], low-power laser irradiation improves cell viability and proliferation by regulating the expression of genes involved in cell proliferation, migration, and remodeling via PI3K/AKT signaling [34] In this study, there was no significant difference between the groups in terms of NF-κB gene expression, one of many factors that trigger the formation of proinflammatory markers. This finding is possibly related to the activation of other signaling pathways that amplify the inflammatory response in diabetes; this includes the polyol pathway, the hexosamine pathway, the diacylglycerol pathway, the PKC (protein kinase C) pathways and especially the formation of advanced glycation end products, which collectively contribute to disrupting scarring [2,35]. ...
Article
Full-text available
Chronic hyperglycemia caused by diabetes mellitus (DM) slows down the healing process due to prolonged inflammation which impedes the regeneration progression. Photobiomodulation (PBM) is considered a non-pharmacological intervention and has anti-inflammatory and biostimulatory effects that accelerate the healing process. Currently found IL-1β inhibitors are difficult to implement due to their cytotoxic potential, excessive amounts, and invasive administration, and therefore, the application of this peptide in diabetic wounds represents a promising intervention to help resolve the inflammatory response. This study aimed to investigate the effect of an IL-1β inhibitor molecule associated with PBM irradiation in a model of epithelial injury in diabetic mice. After the induction of the DM model with streptozotocin (STZ), the skin lesion model was implemented through surgical excision. Sixty C57BL/6 mice divided into five experimental groups (n = 12) were used: excisional wound (EW), DM + EW, DM + EW + DAP 1–2 (inhibitor peptide), DM + EW + PBM, and DM + EW + PBM + DAP 1–2. Treatment started 12 h after wound induction and was performed daily for 5 days. Twenty-four hours after the last application, the animals were euthanized and the outer edge of the wound was removed. The results obtained demonstrate that the DM + EW + PBM + DAP 1–2 group caused a reduction in the levels of pro-inflammatory cytokines, an increase in anti-inflammatory cytokines, and an increase in TGF-β and maintenance of the cellular redox state with a consequent reduction in levels of inflammatory infiltrate and concomitant stimulation of type III collagen gene expression, as well as a decrease in the size of the wound in square centimeter 6 days after the injury. Only the combination of therapies was able to favor the process of tissue regeneration due to the development of an approach capable of acting at different stages of the regenerative process, through the mechanisms of action of interventions on the inflammatory process by avoiding its stagnation and stimulating progression of regeneration.
... Pequenas feridas podem se desenvolver nos pés de pacientes com DM são comumente rotuladas como úlceras de pé diabéticos e muitas vezes passam despercebidas, na maioria dos casos é resultado de neuropatias e/ou vasculopatias. 1,2 As úlceras venosas são complicações prevalentes do DM e são responsáveis por significativa morbidade, mortalidade e gastos com saúde. A 9ª edição do atlas de DM da International Diabetes Federation mostra que existem cerca de 463 milhões de adultos com DM em todo o mundo, estima-se que 19 a 34% dos pacientes com DM, serão afetados com úlcera de pé diabético durante a vida, e cerca de 9,1 a 26,1 milhões de pessoas desenvolverão úlceras de pé diabético anualmente. ...
Article
Full-text available
Úlceras em pacientes com Diabetes Mellitus são frequentes e consideradas uma das complicações de maior relevância durante a evolução clínica da doença. Analisar a aplicação do light-emitting diode (LED) azul no reparo tecidual de úlcera venosa de membro inferior. Consistiu na aplicação de LED azul, com comprimento de onda em torno de 680 THz e 620 THz no local da ferida, durante 10 minutos, 2 vezes por semana, durante 5 semanas. Foram utilizados instrumentos avaliativos de dor e sintomas neuropáticos, testes de sensibilidade, registro fotográfico diário, mensuração do tamanho das úlceras e índice do tempo de fechamento completo da ferida. Foi observada melhora no reparo tecidual das feridas, atingindo fechamento total em ambos os lados e redução da intensidade de dor referida. O LED como tratamento curativo de úlceras diabéticas crônicas mostrou efetividade na taxa de fechamento total da ferida e melhora de sintomas neuropáticos.
Article
Full-text available
Low-level laser (light) therapy (LLLT) promotes wound healing, reduces pain and inflammation, and prevents tissue death. Studies have explored the effects of various radiant exposures on the effect of LLLT; however, studies of wavelength dependency in in vivo models are less common. In the present study, the healing effects of LLLT mediated by different wavelengths of light in the red and near-infrared (NIR) wavelength regions (635, 730, 810, and 980 nm) delivered at constant fluence (4 J/cm(2)) and fluence rate (10 mW/cm(2)) were evaluated in a mouse model of partial-thickness dermal abrasion. Wavelengths of 635 and 810 nm were found to be effective in promoting the healing of dermal abrasions. However, treatment using 730- and 980-nm wavelengths showed no sign of stimulated healing. Healing was maximally augmented in mice treated with an 810-nm wavelength, as evidenced by significant wound area reduction (p < 0.05), enhanced collagen accumulation, and complete re-epithelialization as compared to other wavelengths and non-illuminated controls. Significant acceleration of re-epithelialization and cellular proliferation revealed by immunofluorescence staining for cytokeratin-14 and proliferating cell nuclear antigen (p < 0.05) was evident in the 810-nm wavelength compared with other groups. Photobiomodulation mediated by red (635 nm) and NIR (810 nm) light suggests that the biological response of the wound tissue depends on the wavelength employed. The effectiveness of 810-nm wavelength agrees with previous publications and, together with the partial effectiveness of 635 nm and the ineffectiveness of 730 and 980 nm wavelengths, can be explained by the absorption spectrum of cytochrome c oxidase, the candidate mitochondrial chromophore in LLLT.
Chapter
The first publications about low-power laser therapy (then called laser biostimulation) appeared more than 30 years ago. Since then, approximately 2000 studies have been published on this still controversial topic. In the 1960s and 1970s, doctors in Eastern Europe, and especially in the Soviet Union and Hungary, actively developed laser biostimulation. However, scientists around the world harbored an open skepticism about the credibility of studies stating that low-intensity visible-laser radiation acts directly on an organism at the molecular level. The coherence of laser radiation for achieving stimulative effects on biological objects was more than suspect. Supporters in Western countries, such as Italy, France, and Spain, as well as in Japan and China also adopted and developed this method, but the method was — and still remains — outside mainstream medicine. The controversial points of laser biostimulation, which were topics of great interest at that time, were analyzed in reviews that appeared in the late 1980s.
Article
The healing effect of low energy lasers is generally attributed to enhanced cell proliferation due to the irradiation. As it was not clear whether coherent irradiation is essential, we examined the effect of various wavelengths from non-coherent light sources, on fibroblast proliferation. We found that light at 540 and 600-900 nm significantly accelerates the mitosis of these cells. Moreover, we have found that the effect is not only energy-dose dependent, but depends non-lineraly on the intensity of the light source.
Article
Objective: The biostimulation effects of low-level laser therapy (LLLT) have recently been demonstrated. In this study, we aimed to investigate the effects of LLLT on palatal mucoperiostal wound healing and oxidative stress status in experimental diabetic rats. Materials and methods: Forty-two male Wistar rats that weighed 250-300 g were used in this study. Experimental diabetes was induced in all of the rats using streptozotocin. A standardized full thickness wound was made in the mucoperiosteum of the hard palates of the rats using a 3 mm biopsy punch. The rats were divided into groups: 1 (control group, non- irradiated), and 2 (experimental group, irradiated). Treatment using a GaAlAs laser at a wavelength of 940 nm and at dose of 10 J/cm(2) began after surgery, and was repeated on the 2nd, 4th, and 6th days post-surgery. Seven animals from each group were killed on the 7th, 14th, and 21st day after surgery. Biopsies were performed for the histological analysis and blood samples were collected by cardiac puncture for biochemical analysis. Results: The histopathological findings revealed reduced numbers of inflammatory cells, and increased mitotic activity of fibroblasts, collagen synthesis, and vascularization in rats in group 2. The total oxidative status was significantly decreased in the laser-treated group on the 21st day. Conclusions: LLLT elicits a positive healing effect on palatal mucoperiostal wounds, and modulates the oxidative status in experimental diabetic rats.
Article
This study investigates the feasibility of using an adipose-derived mesenchymal stem cell (ASC)-seeded acellular dermal matrix (ADM) along with low-level laser therapy (LLLT) to repair bone defect in athymic nude mice. Critical-sized calvarial defects were treated either with ADM, ADM/LLLT, ADM/ASCs, or ADM/ASCs/LLLT. In micro-computed tomography scans, the ADM/ASCs and the ADM/ASCs/LLLT groups showed remarkable bone formation after 14 days. Additionally, bone regeneration in the ADM/ASCs/LLLT group was obvious at 28 days, but in the ADM/ASCs group at 56 days. Bone mineral density and bone tissue volume in the ADM/ASCs/LLLT group significantly increased after 7 days, but in the ADM/ASCs group after 14 days. Histological analysis revealed that the defects were repaired in the ADM/ASCs and the ADM/ASCs/LLLT group, while the defects in the ADM and the ADM/LLLT groups exhibited few bone islands at 28 and 56 days. The successful seeding of ASCs onto ADM was confirmed, and LLLT enhanced the proliferation and the survival of ASCs at 14 days. Our results indicate that ASC-seeded grafts promote bone regeneration, and the application of LLLT on ASC-seeded ADM results in rapid bone formation. The implantation of an ASC-seeded ADM combined with LLLT may be used effectively for bone regeneration. © 2013 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 2013.
Article
Introduction: Low-level laser therapy (LLLT) has been reported to be capable of changing mast cell numbers and degranulation in experimental wounds. Objective: We conducted a study on the influence of pulsed LLLT on mast cells in wounds of non-diabetic and diabetic rats. Methods: Thirty-six rats were randomly divided into non-diabetic and diabetic groups. Type 1 diabetes milletes (DM) was induced in rats of the diabetic group by administration of streptozotocin (STZ). We inflicted two wounds in each rat. The first wound in both non-diabetic and diabetic groups was treated with an 890-nm laser, having pulse frequency of 80 Hz and energy density of 0.2 J/cm(2). Mast cell numbers and degranulation of all subgroups were assessed at 4, 7, and 15 days after the infliction of the wounds. Results and conclusion: According to the paired t-test, the total number of laser-treated mast cells was significantly higher than that of the placebos in the non-diabetic groups on days 4 and 15. The total number of laser-treated mast cells was significantly higher than that of the placebos in the diabetic groups on days 4 and 15. The number of granulated mast cells was significantly higher than that of degranulated mast cells for all laser-treated mast cells and placebo mast cells of the non-diabetic and diabetic groups.