ArticlePDF AvailableLiterature Review

Abstract and Figures

Skin fibrosis, also known as skin scarring, is an important global health problem that affects an estimated 100 million persons per year worldwide. Current therapies are associated with significant side effects and even with combination therapy, progression, and recurrence is common. Our goal is to review the available published data available on light-emitting diode-generated (LED) red light phototherapy for treatment of skin fibrosis. A search of the published literature from 1 January 2000 to present on the effects of visible red light on skin fibrosis, and related pathways was performed in January 2016. A search of PubMed and EMBASE was completed using specific keywords and MeSH terms. "Fibrosis" OR "skin fibrosis" OR "collagen" was combined with ("light emitting diode," "LED," "laser," or "red light"). The articles that were original research studies investigating the use of visible red light to treat skin fibrosis or related pathways were selected for inclusion. Our systematic search returned a total of 1376 articles. Duplicate articles were removed resulting in 1189 unique articles, and 133 non-English articles were excluded. From these articles, we identified six articles related to LED effects on skin fibrosis and dermal fibroblasts. We augmented our discussion with additional in vitro data on related pathways. LED phototherapy is an emerging therapeutic modality for treatment of skin fibrosis. There is a growing body of evidence demonstrating that visible LED light, especially in the red spectrum, is capable of modulating key cellular characteristic associated with skin fibrosis. We anticipate that as the understanding of LED-RL's biochemical mechanisms and clinical effects continue to advance, additional therapeutic targets in related pathways may emerge. We believe that the use of LED-RL, in combination with existing and new therapies, has the potential to alter the current treatment paradigm of skin fibrosis. There is a current lack of clinical trials investigating the efficacy of LED-RL to treat skin fibrosis. Randomized clinical trials are needed to demonstrate visible red light's clinical efficacy on different types of skin fibrosis.
Theoretical mechanism of LED red light photobiomodulation. 1 Light has optimal tissue penetration when its wavelength is within the “optical window,” (600–1070 nm). Red light (620–750 nm) takes advantage of this penetration window [15]. 2 LED-RL stimulates the photo-acceptor copper complex in cytochrome C oxidase, stimulating the photodissociation of nitric oxide (NO), leading to upregulation of the electron transport chain [15, 16]. 3 This stimulation of the electron transport leads to the following intramitochondrial changes: increased generation of ATP and reactive oxygen species (ROS), increased intramitochondrial calcium concentration, and an increase in the mitochondrial membrane potential [16]. 4 TGF-Betas are secreted associated with latency-associated peptide (LAP). These associated latency peptides determine the activity TGF-Beta subtypes [17]. Reactive oxygen species have been shown to trigger a conformational change in LAP, thus freeing TGF-Beta 1 from its latency peptide [17–20]. It has been suggested that activation of TGF-Beta 3 may function by a similar ROS-induced release of LAP [20]. 5 TGF-B1, upon being released from its latency complex, binds to the TGF-Beta receptor II (TGF-BRII) stimulating activation [21]. TGF-B3 inhibits activation of the TGF-BRII and antagonizes TGF-B1 signaling [21]. 6 TGF-BRII then forms a heteromeric complex with TGF-BRI, causing the phosphorylation of specific serine residues [21, 22]. 7 When activated, the intracytoplasmic domain of the TGF-Beta receptor complex phosphorylates SMAD proteins. Once phosphorylated, pSMAD2/3 has the ability to migrate to the nucleus and associates with DNA-binding partners to cause changes in target gene expression [22]. SMAD7 activation functions as a negative feedback loop, inhibiting TGF-B1 signaling [23]. 8 SMAD signaling alters COL1A1 gene expression leading to changes in extracellular collagen deposition and changes in fibroblast proliferation [24]. In addition, SMAD signaling contributes to the collagen deposition and pathogenesis of skin fibrosis [25]. These pathways, and related pathways such as Akt, are believed to contribute to skin fibrosis through modulation of fibroblast proliferation and migration speed [26]. 9 TGF-B1 has been shown to increase cell proliferation at low levels; however, high levels of TGF-B1 inhibits dermal fibroblast proliferation, supporting the idea that modulation of levels of TGF-B1 may contribute to LED-RL’s modulation of proliferation [27]. In addition, the release of TGF-B1 or TGF-B3 from their LAP has been suggested as a possible mechanism behind the photobiomodulation of LED-RL [20]. Figure legend: ψm, mitochondrial membrane potential; [Ca]m, intramitochondrial calcium concentration; ATP, adenosine triphosphate; ROS, reactive oxygen species; LAP/LTBP, latency-associate peptide/latent transforming growth factor beta binding protein; TGF-B1, transforming growth factor beta-1; TGF-B3, transforming growth factor beta-3; BR2, transforming growth factor receptor II; BR1, transforming growth factor receptor I; miRNA, microRNA
… 
This content is subject to copyright. Terms and conditions apply.
LASER THERAPY (J JAGDEO, SECTION EDITOR)
Visible Red Light Emitting Diode Photobiomodulation for Skin
Fibrosis: Key Molecular Pathways
Andrew Mamalis
1,2
&Daniel Siegel
3
&Jared Jagdeo
1,2,3
Published online: 16 April 2016
#The Author(s) 2016. This article is published with open access at Springerlink.com
Abstract Skin fibrosis, also known as skin scarring, is an
important global health problem that affects an estimated
100 million persons per year worldwide. Current therapies
are associated with significant side effects and even with com-
bination therapy, progression, and recurrence is common. Our
goal is to review the available published data available on
light-emitting diode-generated (LED) red light phototherapy
for treatment of skin fibrosis. A search of the published liter-
ature from 1 January 2000 to present on the effects of visible
red light on skin fibrosis, and related pathways was performed
in January 2016. A search of PubMed and EMBASE was
completed using specific keywords and MeSH terms.
BFibrosis^OR Bskin fibrosis^OR Bcollagen^was combined
with (Blight emitting diode,^BLED,^Blaser,^or Bred light^).
The articles that were original research studies investigating
the use of visible red light to treat skin fibrosis or related
pathways were selected for inclusion. Our systematic search
returned a total of 1376 articles. Duplicate articles were re-
moved resulting in 1189 unique articles, and 133 non-English
articles were excluded. From these articles, we identified six
articles related to LED effects on skin fibrosis and dermal
fibroblasts. We augmented our discussion with additional
in vitro data on related pathways. LED phototherapy is an
emerging therapeutic modality for treatment of skin fibrosis.
There is a growing body of evidence demonstrating that vis-
ible LED light, especially in the red spectrum, is capable of
modulating key cellular characteristic associated with skin
fibrosis. We anticipate that as the understanding of LED-
RLs biochemical mechanisms and clinical effects continue
to advance, additional therapeutic targets in related pathways
may emerge. We believe that the use of LED-RL, in combi-
nation with existing and new therapies, has the potential to
alter the current treatment paradigm of skin fibrosis. There is a
current lack of clinical trials investigating the efficacy of LED-
RL to treat skin fibrosis. Randomized clinical trials are needed
to demonstrate visible red lights clinical efficacy on different
types of skin fibrosis.
Keywords Skin fibrosis .LED .Visible lig ht .Red light .
Fibroblast .Low level light therapy .Photobiomodulation .
Reactive oxygen species .Collagen
Introduction
Skin fibrosis, also known as skin scarring, is a significant
international health problem with an estimated incidence of
greater than 100 million persons affected per annum world-
wide [1,2]. Skin fibrosis is the key clinical characteristic of
several diseases including systemic sclerosis, morphea, ke-
loids, hypertrophic scars, chronic graft versus host disease,
and gadolinium-induced nephrogenic systemic fibrosis. Skin
fibrosis often results from chronic tissue injury, infection, in-
flammation, or immune response leading to fibroblast activa-
tion. The hallmarks of skin fibrosis are increased fibroblast
proliferation, increased collagen production, increased extra-
cellular matrix (ECM) deposition, and upregulation of pro-
fibrotic signaling pathways (Fig. 1). Despite the morbidity
This article is part of the Topical Collection on Laser Therapy
*Jared Jagdeo
jrjagdeo@gmail.com
1
Department of Dermatology, University of California at Davis,
Sacramento, CA, USA
2
Dermatology Service, Sacramento VA Medical Center, Mather, CA,
USA
3
Department of Dermatology, SUNY Downstate, Brooklyn, NY, USA
Curr Derm Rep (2016) 5:121128
DOI 10.1007/s13671-016-0141-x
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
122 Curr Derm Rep (2016) 5:121128
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
and socioeconomic burdens associated with skin fibrosis,
there are limited effective therapeutic options for skin fibrosis.
Current therapies are associated with significant side effects
and even with combination therapy, progression, and recur-
rence often occurs [3,4].
Ultraviolet (UV) phototherapy is a non-invasive modality
that has been used to treat several diseases associated with
skin fibrosis including morphea, systemic sclerosis, chronic
graft versus host disease, and nephrogenic systemic fibrosis
[57]. However, UV phototherapy causes thymidine dimer
DNA damage that is associated with an increased incidence
of skin cancers and premature photoaging [810]. In addition
to these safety concerns, UV phototherapy units are often
prohibitively expensive for home use and require fluorescent
or incandescent bulbs that limit portability. Therefore, UV
phototherapy requires frequent office visits that patients often
find burdensome [11,12]. In contrast, light-emitting diode-
generated red light (LED-RL) phototherapy is a safe, non-
invasive, inexpensive, and portable treatment that may be
combined with existing treatment modalities. Furthermore,
the visible red light spectrum has superior depth of penetra-
tion, when compared to UV light, that allows it to penetrate
the epidermis and reach the dermis to affect fibroblast function
[13]. LED-RL is not known to cause thymidine dimer DNA
damage or to be associated with an increased incidence of skin
cancer [14]. However, the underlying biochemical mecha-
nisms and clinical effects of visible light photobiomodulation
of skin fibrosis are not well characterized.
The purpose of this review is to review the available evi-
dence on LED-RL phototherapy for the treatment of skin fi-
brosis, witha special emphasis on the key molecular pathways
involved. Herein, we also highlight several strengths and lim-
itations of visible red light phototherapy and suggest enhance-
ments and futuredirections to evaluate their clinical utility. We
anticipate that as the understanding of LED-RLsbiochemical
mechanisms and clinical effects continue to advance, addi-
tional therapeutic targets in related pathways may emerge.
We believe that the use of LED-RL, in combination with
existing and new therapies, has the potential to alter the cur-
rent treatment paradigm of skin fibrosis.
Methods
A search of the published literature from 1 January 2000 to
present on the effects of visible red light on skin fibrosis and
related pathways was performed in January 2016. A search of
PubMed and EMBASE was done using specific keywords
and MeSH terms. BFibrosis^OR Bskin fibrosis^was com-
bined with (Blight emitting diode,^BLED,^Blaser,^or Bred
light^). The articles that were original research studies that
investigated the use of visible red light to treat skin fibrosis
or related pathways were selected for inclusion. Non-English
articles were excluded.
Results
A schematic of our search strategy is outlined in Fig. 2.Our
systematic search returned a total of 1376 articles. Duplicate
articles were removed resulting in 1189 unique articles, and
Fig. 1 a Normal fibroblast function. Fibroblasts are the primary resident
cell in the dermis and are the major contributor to skin fibrosis.
Fibroblasts typically proliferate and produce collagen at a basal rate to
maintain dermal integrity. bAbnormal fibroblast function increases
proliferation and collagen production leading to skin fibrosis.
Fibroblasts contributing to skin fibrosis have an increased proliferation
rate and an increased collagen production and deposition rate. These
cellular alterations are the hallmark of skin fibrosis, and thus are targets
of therapeutic interest. cLight-emitting diode-generated red light (LED-
RL) reduces fibroblast proliferation and collagen production. LED-RL
alters fibroblast function leading to decreased collagen production and
fibroblast proliferation. If LED-RL is capable of returning fibroblast ac-
tivity to basal levels, LED-RL may be a therapeutic option for the pre-
vention or treatment of skin fibrosis
Fig. 2 Schematic of the search
strategy listing the number of
articles matching inclusion or
exclusion criteria
Curr Derm Rep (2016) 5:121128 123
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
133 non-English articles were excluded. From these articles,
we identified six articles related to LED effects on skin fibro-
sis and dermal fibroblasts. We augmented our discussion with
additional in vitro data on related pathways.
Discussion
Molecular Mechanism of Red Light Photobiomodulation
Although visible light makes up 44 % of the total solar energy
in our environment, its effect on cellular function and physiol-
ogy are not fully established [14]. There is emerging in vitro
mechanistic data demonstrating red light may be an effective
treatment for skin fibrosis; however, there is a paucity of evi-
dence for red lights clinical effects. Visible light may represent
a safer therapeutic modality compared to UV light as it does
not generate DNA damage associated with skin cancer [14].
A potential mechanistic pathway demonstrating the cellular
effects of red light photobiomodulation in skin fibrosis is
diagrammed in Fig. 3. Key downstream targets for the modu-
lation of skin fibrosis include reducing cellular fibroblast pro-
liferation and migration speed, inhibiting pro-fibrotic cyto-
kines and their related pathways such as the transforming growth
factor-beta (TGF-beta) pathway, and decreasing synthesis and
deposition of collagen.
Mitochondrial Signaling
The molecular mechanism behind LED-RLs
photobiomodulatory effects appearstoinitiateinthemitochon-
dria [28,29]. Red light stimulates the copper/heme-iron centers
on cytochrome C oxidase (CCO), an intramitochondrial com-
ponent of the electron transport chain (Fig. 3)[15,16]. CCO is a
protein complex containing two copper and heme-iron groups
that play a key role in the electron transport within the mito-
chondrial. LED-RL photostimulation of CCO directly influ-
ences reactive oxygen species (ROS) and adenosine triphos-
phate (ATP) production and results in increased ROS and
ATP lev e ls (Fi g . 3).
Red light has also been shown to modulate a number of
other mitochondrial functions, including increased
intramitochondrial calcium concentration, and alterations in
mitochondrial membrane potential, which may also play a role
in mediating downstream effects (Fig. 3)[16]. Furthermore,
while there is substantial data demonstrating the effects of
redox mechanisms on cellular health and function, there is a
paucity of data on how specific mitochondrial measures, such
as calcium concentration, might lead to downstream cellular
effects. The absence of precise mechanistic links between
these other mitochondrial alterations has led many researchers
to focus on studying the downstream cellular effects of
LED-RL and investigating the role of visible light-
associated alterations in ROS levels.
ROS-Related Intracellular Signaling and Transcriptional
Changes
For instance, altering ROS levels can release TGF-beta 1 and
TGF-beta 3 from their associated latency binding proteins
(Fig. 3). The interaction of these cytokines with their associ-
ated receptors is critical in the pathogenesis or prevention of
skin fibrosis. Altering the levels of TGF-beta 1 versus TGF-
Beta 3 bound to the TGF-beta receptor complex modulates the
pro-fibrotic cascade that leads to downstream activation of
key signaling molecules called SMADs and numerous growth
factors that ultimately result in fibroblast proliferation and
collagen biosynthesis [17,18,21,22].
It is believed that visible light-associated increases in ROS
levels within the cell trigger redox-sensitive transcription
factors such as AP-1, NF-kB, p53, and hypoxia inducible
factor 1 (HIF-1) [30]. Cellular redox changes also modulate
insulin-like growth factors (IGFs), Akt/PKB, and
phosphoinositide 3-kinase (PI3K) pathways, and activate
mammalian target of rapamycin (mTOR) [31]. ROS-initiated
alterations in these pathways often contribute to the
downstream effects on transcription, cellular proliferation,
migration speed, and extracellular matrix production. This
suggests that ROS may be the mechanistic link between
the mitochondrial effects of LED-RL and the resulting
downstream transcriptional and cellular effects.
In additional to these canonical transcriptional alterations,
some researchers believe that alterations in microRNA levels
also play a role in LED-RL photobiomodulation (Fig. 3).
However, there is currently a paucity of data investigating
the specific effects of LED-RL on microRNA levels.
Interestingly, research has demonstrated that laser-generated
visible red light leads to specific alterations in microRNA that
are associated with skin fibrosis, including microRNA-7a, 21,
29, 133b, and 192 [31,32]. Further research is warranted to
investigate the role these microRNA play in causing LED-RL-
associated downstream cellular effects.
Effects of Red Light on Cellular Functions Related
to Fibrosis
Cellular Proliferation
Modulation of mitochondrial, intracellular, and nuclear pro-
cesses ultimately alter downstream cellular processes involved
in skin fibroblast proliferation. For instance, fibroblast prolifera-
tion is a key contributor to the initiation and maintenance of
skin fibrosis, and control of fibroblast proliferation is a critical
therapeutic strategy for addressing skin fibrosis [33]. Our
124 Curr Derm Rep (2016) 5:121128
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
group has found that LED-RL is capable of inhibiting fibro-
blast proliferation in a dose-dependent manner [34].
Furthermore, red light does not appear to affect fibroblast
viability, with no increases in apoptosis or necrosis observed
[34,35••]. This suggests that visible red light is likely modu-
lating fibroblast function through means other than direct
cellular cytotoxicity, such as through modulation of the cell
cycle or autophagy.
It is likely that these alterations in proliferation are a result
of alterations in the redox state of fibroblasts treated with
LED-RL. While mild elevations in free radicals have been
shown to increase proliferation, we have found that the
Fig. 3 Theoretical mechanism of LED red light photobiomodulation. 1
Light has optimal tissue penetration when its wavelength is within the
Boptical window,^(6001070 nm). Red light (620750 nm) takes
advantage of this penetration window [15]. 2LED-RL stimulates the
photo-acceptor copper complex in cytochrome C oxidase, stimulating
the photodissociation of nitric oxide (NO), leading to upregulation of
the electron transport chain [15,16]. 3This stimulation of the electron
transport leads to the following intramitochondrial changes: increased
generation of ATP and reactive oxygen species (ROS), increased
intramitochondrial calcium concentration, and an increase in the
mitochondrial membrane potential [16]. 4TGF-Betas are secreted asso-
ciated with latency-associated peptide (LAP). These associated latency
peptides determine the activity TGF-Beta subtypes [17]. Reactive oxygen
species have been shown to trigger a conformational change in LAP, thus
freeing TGF-Beta 1 from its latency peptide [1720]. It has been
suggested that activation of TGF-Beta 3 may function by a similar
ROS-induced release of LAP [20]. 5TGF-B1, upon being released from
its latency complex, binds to the TGF-Beta receptor II (TGF-BRII)
stimulating activation [21]. TGF-B3 inhibits activation of the TGF-BRII
and antagonizes TGF-B1 signaling [21]. 6TGF-BRII then forms a
heteromeric complex with TGF-BRI, causing the phosphorylation of
specific serine residues [21,22]. 7When activated, the intracytoplasmic
domain of the TGF-Beta receptor complex phosphorylates SMAD
proteins. Once phosphorylated, pSMAD2/3 has the ability to migrate to
the nucleus and associates with DNA-binding partners to cause changes
in target gene expression [22]. SMAD7 activation functions as a negative
feedback loop, inhibiting TGF-B1 signaling [23]. 8SMAD signaling
alters COL1A1 gene expression leading to changes in extracellular col-
lagen deposition and changes in fibroblast proliferation [24]. In addition,
SMAD signaling contributes to the collagen deposition and pathogenesis
of skin fibrosis [25]. These pathways, and related pathways such as Akt,
are believed to contribute to skin fibrosis through modulation offibroblast
proliferation and migration speed [26]. 9TGF-B1 has been shown to
increase cell proliferation at low levels; however, high levels of
TGF-B1 inhibits dermal fibroblast proliferation, supporting the idea that
modulation of levels of TGF-B1 may contribute to LED-RLs modulation
of proliferation [27]. In addition, the release of TGF-B1 or TGF-B3 from
their LAP has been suggested as a possible mechanism behind the
photobiomodulation of LED-RL [20]. Figure legend: ψm, mitochondrial
membrane potential; [Ca]m,intramitochondrial calcium concentration;
ATP, adenosine triphosphate; ROS, reactive oxygen species; LAP/LTBP,
latency-associate peptide/latent transforming growth factor beta binding
protein; TGF-B1, transforming growth factor beta-1; TGF-B3,
transforming growth factor beta-3; BR2, transforming growth factor
receptor II; BR1, transforming growth factor receptor I; miRNA,
microRNA
Curr Derm Rep (2016) 5:121128 125
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
dose-dependent decreases in fibroblast proliferation are
associated with a dose-dependent sustained increase in
ROS [35••,36]. Therefore, it is likely that the sustained
alterations in the redox state of fibroblasts treated with
LED-RL and the subsequent redox-initiated alterations
in the TGF-beta pathway and related pathways are con-
tributing to the dose-dependent decrease in fibroblast
proliferation.
Cellular Migration
Furthermore, some believe that cellular migration speed may
play a role in the recruitment of fibroblasts to sites of increased
collagen production [3,4]. This finding is supported by the
fact that fibroblasts derived from skin affected by skin fibrosis
demonstrate increased motility when compared to fibroblasts
derived from normal healthy skin [37,38]. Few studies have
sought to address this potential therapeutic avenue and so the
clinical effect of decreasing fibroblast motility is still unclear.
Researchers have demonstrated that the PI3K/Akt and
MAPK/ERK pathways play crucial roles in the regulation of
fibroblast migration, and that visible light is capable of direct-
ly activating or inhibiting the phosphorylation state of these
key cell signaling molecules [32,3944]. Our group recently
found that LED-RL increased ROS levels and decreased fi-
broblast migration speed in a dose-dependent manner (Fig. 3)
[35••]. Additionally, we found that LED-RL also altered
phospho-Akt levels (unpublished data by Jagdeo Lab).
Furthermore, migration speed returned to control levels when
ROS increases were blocked by the pretreatment of fibroblasts
with the antioxidant resveratrol or when cells were pretreated
with the PI3K/Akt inhibitor LY294002 [35••]. This suggests
that LED-RLs effects on migration may be largely mediated
by increased ROS that lead to modulation of phospho-Akt
levels and subsequent alterations in fibroblast migration
speed. Further research is needed to investigate the role cellu-
lar migration speed plays in the pathogenesis of skin fibrosis
and the clinical effects of therapeutically targeting fibroblast
motility.
Collagen Production
Fundamentally, the pathogenesis of all forms of skin fibrosis
involves an increased deposition of skin collagen [33].
Therefore, suppression of collagen production is a fundamen-
tal component of any effective anti-fibrotic therapy [33].
Several studies support that visible red light is capable of
modulating collagen production in vitro. Our group has dem-
onstrated that LED-RL is capable of suppressing collagen
production in human skin fibroblast cultures. In this study,
fibroblasts were treated with LED-RL, and then collagen
was measured using the collagen stain, picrosirius red (unpub-
lished data by Jagdeo Lab). LED-RL resulted in decreased
collagen production in a dose-dependent manner (Fig. 3).
Furthermore, procollagen 1A1 levels were found to be de-
creased following LED-RL treatment, suggesting that this de-
crease in collagen levels may be due in large part to decreases
in collagen subunits.
Another study investigated the effect of visible red light
generated by a diode laser on murine NIH/3T3 fibroblasts
[45]. They found that red light treatment inhibited TGF-beta
induced fibroblast to myofibroblast differentiation and de-
creased collagen 1 expression. Furthermore, they found that
red light was capable of upregulating matrix metalloprotein-
ases (MMP)-2 and MMP-9, while downregulating tissue in-
hibitor of metalloproteinase (TIMP)-1 and TIMP-2 [45]. This
suggests that red light may not only decrease collagen produc-
tion, but may also change overall extracellular matrix remod-
eling profile. Further studies are needed evaluating the effects
of red light phototherapy on in vivo collagen content and
homeostasis; however, these early in vitro findings are
promising.
Limitations and Future Directions
However, red light phototherapy does possess several limita-
tions. First, the current understanding of the biochemical
mechanisms underlying visible light photobiomodulation is
limited. More laboratory research is needed to characterize
the key pathways involved in initiating the downstream cellu-
lar effects observed. Another limitation of the field of visible
light phototherapy is that many in vitro studies are done on
cultured skin fibroblasts. Fibroblast monocultures do not
completely recapitulate the complex fibroblast phenotype or
the extracellular milieu that contributes to skin fibrosis pathol-
ogy. Thus, randomized clinical trials are needed to demon-
strate visible red lightseffectonskinfibrosis.
Perhaps, one of the most critical challenges facing visible
light phototherapy is the selection of appropriate dosimetry.
Visible light does not have sufficient measures for evaluating
the pharmacokinetics of light or its effect on in vivo tissue.
Therefore, many dosing protocols are based upon observed
effects. However, the fluence delivered depends on the dura-
tion of treatment, the power density of the light source, and the
distance of the source from its target tissue. Differing any one
variable can at times lead to different photobiomodulatory
effects. For instance, while red light at fluences above 320 J/
cm2 inhibit fibroblast proliferation, red light at fluences below
50 J/cm2 often promote fibroblast proliferation. Therefore,
establishing standardized dosing ranges and thresholds for
future basic science and clinical research studies may improve
the comparability of different clinical studies.
We believe the use of commercially available LEDs as a
visible light source is an exciting avenue of future research.
LED-RL devices are safe, economic, and portable, and we
126 Curr Derm Rep (2016) 5:121128
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
believe are the optimal devices for future research and clinical
use of visible red light.
Conclusions
Visible light phototherapy is an emerging therapeutic modal-
ity for treatment of skin fibrosis. There is a growing body of
evidence demonstrating that visible red light is capable of
modulating key cellular characteristic associated with skin
fibrosis. We believe that further laboratory research may elu-
cidate the underlying mechanisms and effects involved in vis-
ible light photobiomodulation. LED-based devices are the op-
timal devices for red visible light phototherapy. There is a
current lack of clinical trials investigating the efficacy of
LED-RL to treat skin fibrosis. Randomized clinical trials are
needed to demonstrate visible red lightsclinicalefficacyon
different types of skin fibrosis.
Acknowledgments This study was funded by the National Center for
Advancing Translational Sciences, National Institutes of Health, through
grant number UL1 TR000002 and linked awards TL1 TR000133 and
KL2 TR000134.
Compliance with Ethical Standards
Conflict of Interest The authors declare that they have no conflict of
interest.
Human and Animal Rights and Informed Consent This article does
not contain any studies with human or animal subjects performed by any
of the authors.
Disclaimer The contents herein do not represent the views of the US
Department of Veterans Affairs or the US Government. The content is
solely the responsibility of the authors and does not necessarily represent
the official views of the National Institutes of Health.
Open Access This article is distributed under the terms of the Creative
Commons Attribution 4.0 International License (http://
creativecommons.org/licenses/by/4.0/), which permits unrestricted use,
distribution, and reproduction in any medium, provided you give appro-
priate credit to the original author(s) and the source, provide a link to the
Creative Commons license, and indicate if changes were made.
References
Papers of particular interest, published recently, have been
highlighted as:
•• Of major importance
1. Bayat A, McGrouther DA, Ferguson MW. Skin scarring. BMJ.
2003;326:8892.
2. Lim AF, Weintraub J, Kaplan EN, et al. The embrace device sig-
nificantly decreases scarring following scar revision surgery in a
randomized controlled trial. Plast Reconstr Surg. 2014;133:398405.
3. Mamalis AD, Lev-Tov H, Nguyen DH, et al. Laser and light-based
treatment of keloidsa review. J Eur Acad Dermatol Venereol.
2013;28(6):68999.
4. Vitiello M, Abuchar A, Santana N, et al. An update on the treatment
of the cutaneous manifestations of systemic sclerosis: the dermatol-
ogists point of view. J Clin Aesthet Dermatol. 2012;5:3343.
5. Tran KT, Prather HB, Cockerell CJ, et al. UV-A1 therapy for
nephrogenic systemic fibrosis. Arch Dermatol. 2009;145:11704.
6. Connolly KL, Griffith JL, McEvoy M, et al. Ultraviolet A1 photo-
therapy beyond morphea: experience in 83 patients. Photodermatol
Photoimmunol Photomed. 2015;31:28995.
7. Sunderkotter C, Kuhn A, Hunzelmann N, et al. Phototherapy: a
promising treatment option for skin sclerosis in scleroderma?
Rheumatology (Oxford). 2006;45 Suppl 3:iii52iii4.
8. Berking C, Takemoto R, Satyamoorthy K, et al. Induction of mel-
anoma phenotypes in human skin by growth factors and ultraviolet
B. Cancer Res. 2004;64:80711.
9. Cleaver JE, Crowley E. UV damage, DNA repair and skin carcino-
genesis. Front Biosci. 2002;7:d102443.
10. Matsumura Y, Ananthaswamy HN. Short-term and long-term cel-
lular and molecular events following UV irradiation of skin: implica-
tions for molecular medicine. Expert Rev Mol Med. 2002;4:122.
11. Koek MB, Sigurdsson V, van Weelden H, et al. Cost effectiveness
of home ultraviolet B phototherapy for psoriasis: economic evalu-
ation of a randomised controlled trial (PLUTO study). BMJ.
2010;340:c1490.
12. NPF. National Psoriasis Foundation policy brief: phototherapy
copayments impact access to treatment 2010 [cited 2015 10-10-
2015]. Available from: http://www.psoriasis.org/document.doc?
id=1387.
13. Jagdeo JR, Adams LE, Brody NI, et al. Transcranial red and near
infrared light transmission in a cadaveric model. PLoS One. 2012;7:
e47460.
14. Liebel F, Kaur S, Ruvolo E, et al. Irradiation of skin with visible
light induces reactive oxygen species and matrix-degrading en-
zymes. J Investig Dermatol. 2012;132:19017.
15. Chung H, Dai T, Sharma SK, et al. The nuts and bolts of low-level
laser (light) therapy. Ann Biomed Eng. 2012;40:51633.
16. Karu TI. Mitochondrial signaling in mammalian cells activated by
red and near-IR radiation. Photochem Photobiol. 2008;84:10919.
17. Barcellos-Hoff MH, Dix TA. Redox-mediated activation of latent
transforming growth factor-beta 1. Mol Endocrinol. 1996;10:
107783.
18. Jobling MF, Mott JD, Finnegan MT, et al. Isoform-specific activa-
tion of latent transforming growth factor beta (LTGF-beta) by reac-
tive oxygen species. Radiat Res. 2006;166:83948.
19. Lorda-Diez CI, Montero JA, Garcia-Porrero JA, et al. Tgfbeta2 and
3 are coexpressed with their extracellular regulator Ltbp1 in the
early limb bud and modulate mesodermal outgrowth and BMP
signaling in chicken embryos. BMC Dev Biol. 2010;10:69.
20. Waynant R, Tata D, Arany P. Photobiomodulation by low power
laser irradiation involves activation of latent TGF-β1. Proceedings of
light-activated tissue regeneration and therapy conference. Lecture
notes in electrical engineering. 12: Springer US; 2008. p. 20712.
21. Massague J. TGFbeta signaling: receptors, transducers, and Mad
proteins. Cell. 1996;85:94750.
22. Chin GS, Liu W, Peled Z, et al. Differential expression of
transforming growth factor-beta receptors I and II and activation of
Smad 3 in keloid fibroblasts. Plast Reconstr Surg. 2001;108:4239.
23. Yan X, Liu Z, Chen Y. Regulation of TGF-beta signaling by Smad7.
Acta Biochim Biophys Sin Shanghai. 2009;41:26372.
24. Penn JW, Grobbelaar AO, Rolfe KJ. The role of the TGF-beta
family in wound healing, burns and scarring: a review. Int J Burns
Trauma. 2012;2:1828.
25. Khoo YT, Ong CT, Mukhopadhyay A, et al. Upregulation of secre-
tory connective tissue growth factor (CTGF) in keratinocyte-
Curr Derm Rep (2016) 5:121128 127
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
fibroblast coculture contributes to keloid pathogenesis. J Cell
Physiol. 2006;208:33643.
26. Cutroneo KR. TGF-beta-induced fibrosis and SMAD signaling:
oligo decoys as natural therapeutics for inhibition of tissue fibrosis
and scarring. Wound Repair Regen. 2007;15 Suppl 1:S5460.
27. Ghahary A, Tredget EE, Ghahary A, et al. Cell proliferating effect
of latent transforming growth factor-beta1 is cell membrane depen-
dent. Wound Repair Regen. 2002;10:32835.
28. Greco M, Guida G, Perlino E, et al. Increase in RNA and protein
synthesis by mitochondria irradiated with helium-neon laser.
Biochem Biophys Res Commun. 1989;163:142834.
29. Karu TI, Pyatibrat LV, Kalendo GS. Photobiological modulation of
cell attachment via cytochrome c oxidase. Photochem Photobiol
Sci. 2004;3:2116.
30. Chen AC, Arany PR, Huang YY, et al. Low-level laser therapy
activates NF-kB via generation of reactive oxygen species in mouse
embryonic fibroblasts. PLoS One. 2011;6:e22453.
31. Shroff A, Mamalis A, Jagdeo J. Oxidative stress and skin fibrosis.
Curr Pathobiol Rep. 2014;2:25767.
32. Choi H, Lim W, Kim I, et al. Inflammatory cytokines are sup-
pressed by light-emitting diode irradiation of P. gingivalis LPS-
treated human gingival fibroblasts: inflammatory cytokine changes
by LED irradiation. Lasers Med Sci. 2012;27:45967.
33. Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic
translation for fibrotic disease. Nat Med. 2012;18:102840.
34. Lev-Tov H, Mamalis A, Brody N, et al. Inhibition of fibroblast
proliferation in vitro using red light-emitting diodes. Dermatol
Surg. 2013;39:116770.
35.•• Mamalis A, Koo E, Isseroff RR, et al. Resveratrol prevents high
fluence red light-emitting diode reactive oxygen species-mediated
photoinhibition of human skin fibroblast migration. PLoS One.
2015;10:e0140628. First study demostrating that antioxidants
were capable of reversing the biologic effects of LED red light
on skin fibroblasts.
36. Sarsour EH, Kumar MG, Chaudhuri L, et al. Redox control of the
cell cycle in health and disease. Antioxid Redox Signal. 2009;11:
29853011.
37. Ashcroft KJ, Syed F, Bayat A. Site-specific keloid fibroblasts alter
the behaviour of normal skin and normal scar fibroblasts through
paracrine signalling. PLoS One. 2013;8:e75600.
38. Wu CS, Wu PH, Fang AH, et al. FK506 inhibits the enhancing effects
of transforming growth factor (TGF)-beta1 on collagen expression
and TGF-beta/Smad signalling in keloid fibroblasts: implication for
new therapeutic approach. Br J Dermatol. 2012;167:53241.
39. Guo A, Song B, Reid B, etal. Effects of physiological electric fields
on migration of human dermal fibroblasts. J Investig Dermatol.
2010;130:23207.
40. Huang C, Jacobson K, Schaller MD. MAP kinases and cell migra-
tion. J Cell Sci. 2004;117:461928.
41. Kanazawa S, Fujiwara T, Matsuzaki S, et al. bFGF regulates PI3-
kinase-Rac1-JNK pathway and promotes fibroblast migration in
wound healing. PLoS One. 2010;5:e12228.
42. Li W, Fan J, Chen M, et al. Mechanism of human dermal fibroblast
migration driven by type I collagen and platelet-derived growth
factor-BB. Mol Biol Cell. 2004;15:294309.
43. Sepe L, Ferrari MC, Cantarella C, et al. Ras activated ERK and
PI3K pathways differentially affect directional movement of cul-
tured fibroblasts. Cell Physiol Biochem. 2013;31:12342.
44. Ong WK, Chen HF, Tsai CT, et al. The activation of directional
stem cell motility by green light-emitting diode irradiation.
Biomaterials. 2013;34:191120.
45. Sassoli C, Chellini F, Squecco R, et al. Low intensity 635 nm diode
laser irradiation inhibits fibroblast-myofibroblast transition reduc-
ing TRPC1 channel expression/activity: new perspectives for tissue
fibrosis treatment. Lasers Surg Med. 2015;48(3):31832.
128 Curr Derm Rep (2016) 5:121128
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Although the broad range of visible light (400-700 nm, blue, green, and red) could induce ROS generation that has been linked to various cellular mechanisms [7,[9][10][11], the individual effects of cell viability in specific light ranges is not yet fully understood. ...
... Therefore, it is noteworthy to examine the effect of LED light on the skin and investigate the specific mechanisms. Recently accumulated evidence has suggested that while red LED light increased cell proliferation, blue LED light decreased cell viability in various cell types via altered mitochondrial functions including aberrant ROS formation [7][8][9][10][11][12]. ...
... FAK activity rapidly and robustly increased in the order of blue, green, and red LED lights. Recent studies have suggested that red LED light stimulated ERK activation and promoted cell proliferation, while blue LED light induced the inflammation pathway in various cell types [7,8,10,11]. Upon red LED light exposure, phosphorylation of ERK was rapidly increased at 10 min and then decreased after 30 min. ...
Article
Full-text available
In this study, changes in cell signaling mechanisms in skin cells induced by various wavelengths and intensities of light-emitting diodes (LED) were investigated, focusing on the activity of focal adhesion kinase (FAK) in particular. We examined the effect of LED irradiation on cell survival, the generation of intracellular reactive oxygen species (ROS), and the activity of various cell-signaling proteins. Red LED light increased cell viability at all intensities, whereas strong green and blue LED light reduced cell viability, and this effect was reversed by NAC or DPI treatment. Red LED light caused an increase in ROS formation according to the increase in the intensity of the LED light, and green and blue LED lights led to sharp increases in ROS formation. In the initial reaction to LEDs, red LED light only increased the phosphorylation of FAK and extracellular-signal regulated protein kinase (ERK), whereas green and blue LED lights increased the phosphorylation of inhibitory-κB Kinase α (IKKα), c-jun N-terminal kinase (JNK), and p38. The phosphorylation of these intracellular proteins was reduced via FAK inhibitor, NAC, and DPI treatments. Even after 24 h of LED irradiation, the activity of FAK and ERK appeared in cells treated with red LED light but did not appear in cells treated with green and blue LED lights. Furthermore, the activity of caspase-3 was confirmed along with cell detachment. Therefore, our results suggest that red LED light induced mitogenic effects via low levels of ROS–FAK–ERK, while green and blue LED lights induced cytotoxic effects via cellular stress and apoptosis signaling resulting from high levels of ROS.
... Fibroblastic proliferation and excess collagen deposition are associated with imbalanced healing and scarring [3,51,52]. Interestingly, studies have shown that light within the red part of the visual spectrum suppresses collagen production in human skin fibroblast cultures [51] and inhibits type I collagen expression as well as TGF-βinduced fibroblast to myofibroblast differentiation [53]. Previous studies focusing on FLE have investigated collagen production early after treatment showing that FLE can modulate collagen production both in vitro and in vivo [12,16]. ...
... Fibroblastic proliferation and excess collagen deposition are associated with imbalanced healing and scarring [3,51,52]. Interestingly, studies have shown that light within the red part of the visual spectrum suppresses collagen production in human skin fibroblast cultures [51] and inhibits type I collagen expression as well as TGF-βinduced fibroblast to myofibroblast differentiation [53]. Previous studies focusing on FLE have investigated collagen production early after treatment showing that FLE can modulate collagen production both in vitro and in vivo [12,16]. ...
Article
Full-text available
Skin grafting is often the only treatment for skin trauma when large areas of tissue are affected. This surgical intervention damages the deeper dermal layers of the skin with implications for wound healing and a risk of scar development. Photobiomodulation (PBM) therapy modulates biological processes in different tissues, with a positive effect on many cell types and pathways essential for wound healing. This study investigated the effect of fluorescent light energy (FLE) therapy, a novel type of PBM, on healing after skin grafting in a dermal fibrotic mouse model. Split-thickness human skin grafts were transplanted onto full-thickness excisional wounds on nude mice. Treated wounds were monitored, and excised xenografts were examined to assess healing and pathophysiological processes essential for developing chronic wounds or scarring. Results demonstrated that FLE treatment initially accelerated re-epithelialization and rete ridge formation, while later reduced neovascularization, collagen deposition, myofibroblast and mast cell accumulation, and connective tissue growth factor expression. While there was no visible difference in gross morphology, we found that FLE treatment promoted a balanced collagen remodeling. Collectively, these findings suggest that FLE has a conceivable effect at balancing healing after skin grafting, which reduces the risk of infections, chronic wound development, and fibrotic scarring.
... 162 Several studies have shown the potential of high fluence red light (320-640 J/cm 2 ) to improve skin fibrosis by reducing fibroblast proliferation, collagen deposition, and migration. [163][164][165][166] Transcriptional changes leading to antifibrotic cellular responses such as an increase in MMP-1 have been reported regarding this potential scar treatment modality. 167 In regenerative medicine, the self-assembled skin substitute (SASS) is a method where intact human skin is grown in vitro from primary skin cells (i.e., fibroblasts and keratinocytes), avoiding the use of synthetic or heterologous material [168][169][170][171] and where cells grow their own ECM. ...
Article
Full-text available
Objective: To describe current knowledge regarding established and putative cell signaling pathways involved in skin photobiomodulation. Background: The skin is the largest and most accessible organ of the body. It is the first line of defense against the external environment, including solar radiation. Among solar rays, visible and infrared non-ionizing photons may reach human skin and trigger a cascade of non-thermal cell signaling pathways called photobiomodulation (PBM). The use of PBM using artificial light sources has been known for more than 50 years, but it has not yet been widely accepted due to uncertainty about the cellular mechanisms of action. However, much knowledge has been gained in this field in recent years, which will be summarized in this review. Methods: An extensive literature review was performed using Medline, Embase, and Google Scholar as research databases to acquire relevant publications in this particular field. Results: A comprehensive description of chromophores, primary and secondary effectors is provided in addition to a visual representation of known and putative cell signaling mechanisms involved in such complex light-skin interactions. Also, a summary of clinical indications of skin PBM, key light parameters, and promising skin applications (local and systemic) are mentioned. Conclusions: In PBM, skin cells are the first to absorb photons, triggering specific cell-signaling pathways through primary and secondary effectors, leading to enhanced cell repair and survival, notably in hypoxic or stressed cells. A better understanding of the mechanisms of action will help us optimize known indications and discover new ones.
... Recently accumulated evidence has indicated that red light promotes an increase in cell proliferation, whereas blue light reduces the viability of different cell types by inducing changes in mitochondrial function, including aberrant ROS formation [5][6][7][8]. In particular, blue light has been demonstrated to have a detrimental effect on the skin. ...
Article
Full-text available
Blue light derived from multiple sources, including sunlight, generates reactive oxygen species (ROS) and negatively affects the skin in a manner similar to that of ultraviolet light. Cyanidin-3-O-glucoside (C3OG) and cyanidin-3-O-ruti-noside (C3OR) are anthocyanin antioxidants that have protective effects on various tissues and cell types. However, the effects of anthocyanins on blue light-mediated changes remain unconfirmed. In this study, we determined the protective effects of C3OG and C3OR isolated and purified from waste cherry fruits (Prunus serrulata L. var. tomentella Nakai) against the blue light-induced ROS formation and inflammatory responses in HaCaT cells. It is showed that the treatment of C3OG and C3OR significantly reduced the blue light-induced cytotoxicity and ROS production in a dose dependent manner. Furthermore, we found that focal adhesion kinase (FAK) is a major upstream of blue light-induced expression of inflammatory cytokines (TNF-α, IL-6 and IL-8), and these effects were attenuated by C3OG or C3OR treatment. In the initial reaction, blue lights increased the phosphorylation of inhibitory-κB Kinase α (IKKα), c-jun N-terminal kinase (JNK), and p38. The phosphorylation of these intracellular proteins was reduced via FAK inhibitor, NAC (ROS scavenger), and anthocyanin treatments. After 24 h of blue light irradiation, C3OG or C3OR treatment markedly inhibited caspase-3-mediated apoptosis and cleaved-FAK-mediated anoikis, which is cell detachment-induced apoptosis. Therefore, our results indicate that C3OG and C3OR effectively protected human keratinocytes from harmful blue light-induced cytotoxicity and inflammation.
... Although there are many assumptions about the cellular mechanism of action of PBM, there is a commonly accepted nding that the biochemical reactions start with the absorption of light by the enzyme of cytochrome c oxidase, which is a mitochondrial electron transport chain element, then intracellular reactive oxygen species (ROS) are produced, the mitochondrial membrane potential (MMP) increases and it results in a protein gradient across the cell and mitochondrial membrane. Beside the production of intracellular ROS, there is a release of nitric oxide (NO) and ATP production in the cells that are treated by light at low-level intensities [12,13]. ATP synthesis, intracellular ROS production, and NO release show their effects in the long-term follow-up of the cell behaviors [14,15]. ...
Preprint
Full-text available
Neurodegenerative diseases are the results of irreversible damages in the neuronal cells by affecting vital functions temporarily or even permanently. The use of light for the treatment of these diseases is an emerging promising innovative method. Photobiomodulation (PBM) and Photodynamic Therapy (PDT) are the modalities that have a wide range of use in medicine and have opposite purposes, biostimulation and cell death respectively. In this study, we aimed to compare these two modalities (PDT and PBM) at low-level intensities and create a stimulatory effect on the differentiation of PC12 cells. Three different energy densities (1, 3, and 5 J/cm 2 ) were used in PBM and Chlorin e6-mediated PDT applications upon irradiation with 655-nm laser light.The light-induced differentiation profile of PC12 cells was analyzed by morphological examinations, qRT-PCR, cell viability assay, and some mechanistic approaches such as; the analysis of intracellular ROS production, NO release, and mitochondrial membrane potential change. It has been observed that both of these modalities were successful at neural cell differentiation. PBM at 1 J/cm 2 and low-dose PDT at 3 J/cm 2 energy densities provided the best differentiation profiles which were proved by the over-expressions of SYN-1 and GAP43 genes. It was also observed that intracellular ROS production and NO release had pivotal roles in these mechanisms with more cell differentiation obtained especially in low-dose PDT application. It can be concluded that light-induced mechanisms with properly optimized light parameters have the capacity for neural cell regeneration and thus, can be a successful treatment for incurable neurodegenerative diseases.
... The PBM is an emerging therapeutic modality for the treatment of cutaneous fibrosis. The PBM would act on the cellular plan, as a modulator of reactions, would reduce the speed of proliferation and migration of fibroblasts, would inhibit pro-fibrotic cytokines and their related pathways such as that of TGF-beta and would decrease the synthesis and deposition of collagen [66,67]. ...
Article
Intrauterine adhesion (IUA), a major cause of uterine infertility, is pathologically characterized by endometrial fibrosis. Current treatments for IUA have poor efficacy with high recurrence rate, and restoring uterine functions is difficult. We aimed to determine the therapeutic efficacy of photobiomodulation (PBM) therapy on IUA and elucidate its underlying mechanisms. A rat IUA model was established via mechanical injury, and PBM was applied intrauterinely. The uterine structure and function were evaluated using ultrasonography, histology, and fertility tests. PBM therapy induced a thicker, more intact, and less fibrotic endometrium. PBM also partly recovered endometrial receptivity and fertility in IUA rats. A cellular fibrosis model was then established with human endometrial stromal cells (ESCs) cultured in the presence of TGF-β1. PBM alleviated TGF-β1-induced fibrosis and triggered cAMP/PKA/CREB signaling in ESCs. Pretreatment with the inhibitors targeting this pathway weakened PBM's protective efficacy in the IUA rats and ESCs. Therefore, we conclude that PBM improved endometrial fibrosis and fertility via activating cAMP/PKA/CREB signaling in IUA uterus. This study sheds more lights on the efficacy of PBM as a potential treatment for IUA.
Article
Full-text available
Disclaimer This article is based on recommendations from the 12 th WALT Congress, Nice, October 3-6, 2018, and a follow-up review of the existing data and the clinical observations of an international multidisciplinary panel of clinicians and researchers with expertise in the area of supportive care in cancer and/or PBM clinical application and dosimetry. This article is informational in nature. As with all clinical materials, this paper should be used with a clear understanding that continued research and practice could result in new insights and recommendations. The review reflects the collective opinion and, as such, does not necessarily represent the opinion of any individual author. In no event shall the authors be liable for any decision made or action taken in reliance on the proposed protocols. Objective This position paper reviews the potential prophylactic and therapeutic effects of photobiomodulation (PBM) on side effects of cancer therapy, including chemotherapy (CT), radiation therapy (RT), and hematopoietic stem cell transplantation (HSCT). Background There is a considerable body of evidence supporting the efficacy of PBM for preventing oral mucositis (OM) in patients undergoing RT for head and neck cancer (HNC), CT, or HSCT. This could enhance patients’ quality of life, adherence to the prescribed cancer therapy, and treatment outcomes while reducing the cost of cancer care. Methods A literature review on PBM effectiveness and dosimetry considerations for managing certain complications of cancer therapy were conducted. A systematic review was conducted when numerous randomized controlled trials were available. Results were presented and discussed at an international consensus meeting at the World Association of photobiomoduLation Therapy (WALT) meeting in 2018 that included world expert oncologists, radiation oncologists, oral oncologists, and oral medicine professionals, physicists, engineers, and oncology researchers. The potential mechanism of action of PBM and evidence of PBM efficacy through reported outcomes for individual indications were assessed. Results There is a large body of evidence demonstrating the efficacy of PBM for preventing OM in certain cancer patient populations, as recently outlined by the Multinational Association for Supportive Care in Cancer/International Society of Oral Oncology (MASCC/ISOO). Building on these, the WALT group outlines evidence and prescribed PBM treatment parameters for prophylactic and therapeutic use in supportive care for radiodermatitis, dysphagia, xerostomia, dysgeusia, trismus, mucosal and bone necrosis, lymphedema, hand-foot syndrome, alopecia, oral and dermatologic chronic graft-versus-host disease, voice/speech alterations, peripheral neuropathy, and late fibrosis amongst cancer survivors. Conclusions There is robust evidence for using PBM to prevent and treat a broad range of complications in cancer care. Specific clinical practice guidelines or evidence-based expert consensus recommendations are provided. These recommendations are aimed at improving the clinical utilization of PBM therapy in supportive cancer care and promoting research in this field. It is anticipated these guidelines will be revised periodically.
Article
Background Neurodegenerative diseases are the results of irreversible damages in the neuronal cells by affecting vital functions temporarily or even permanently. The use of light for the treatment of these diseases is an emerging promising innovative method. Photobiomodulation (PBM) and Photodynamic Therapy (PDT) are the modalities that have a wide range of use in medicine and have opposite purposes, biostimulation and cell death respectively. Methods In this study, we aimed to compare these two modalities (PDT and PBM) at low-level intensities and create a stimulatory effect on the differentiation of PC12 cells. Three different energy densities (1, 3, and 5 J/cm²) were used in PBM and Chlorin e6-mediated PDT applications upon irradiation with 655-nm laser light. The light-induced differentiation profile of PC12 cells was analyzed by morphological examinations, qRT-PCR, cell viability assay, and some mechanistic approaches such as; the analysis of intracellular ROS production, NO release, and mitochondrial membrane potential change. Results It has been observed that both of these modalities were successful at neural cell differentiation. PBM at 1 J/cm² and low-dose PDT at 3 J/cm² energy densities provided the best differentiation profiles which were proved by the over-expressions of SYN-1 and GAP43 genes. It was also observed that intracellular ROS production and NO release had pivotal roles in these mechanisms with more cell differentiation obtained especially in low-dose PDT application. Conclusion It can be concluded that light-induced mechanisms with properly optimized light parameters have the capacity for neural cell regeneration and thus, can be a successful treatment for incurable neurodegenerative diseases.
Article
Full-text available
Background: Skin fibrosis is a significant medical problem that leads to a functional, aesthetic, and psychosocial impact on quality-of-life. Light-emitting diode-generated 633-nm red light (LED-RL) is part of the visible light spectrum that is not known to cause DNA damage and is considered a safe, non-invasive, inexpensive, and portable potential alternative to ultraviolet phototherapy that may change the treatment paradigm of fibrotic skin disease. Objective: The goal of our study was to investigate the how reactive oxygen species (ROS) free radicals generated by high fluence LED-RL inhibit the migration of skin fibroblasts, the main cell type involved in skin fibrosis. Fibroblast migration speed is increased in skin fibrosis, and we studied cellular migration speed of cultured human skin fibroblasts as a surrogate measure of high fluence LED-RL effect on fibroblast function. To ascertain the inhibitory role of LED-RL generated ROS on migration speed, we hypothesized that resveratrol, a potent antioxidant, could prevent the photoinhibitory effects of high fluence LED-RL on fibroblast migration speed. Methods: High fluence LED-RL generated ROS were measured by flow cytometry analysis using dihydrorhodamine (DHR). For purposes of comparison, we assessed the effects of ROS generated by hydrogen peroxide (H2O2) on fibroblast migration speed and the ability of resveratrol, a well known antioxidant, to prevent LED-RL and H2O2 generated ROS-associated changes in fibroblast migration speed. To determine whether resveratrol could prevent the high fluence LED-RL ROS-mediated photoinhibition of human skin fibroblast migration, treated cells were incubated with resveratrol at concentrations of 0.0001% and 0.001% for 24 hours, irradiated with high fluences LED-RL of 480, 640, and 800 J/cm2. Results: High fluence LED-RL increases intracellular fibroblast ROS and decreases fibroblast migration speed. LED-RL at 480, 640 and 800 J/cm2 increased ROS levels to 132.8%, 151.0%, and 158.4% relative to matched controls, respectively. These LED-RL associated increases in ROS were prevented by pretreating cells with 0.0001% or 0.001% resveratrol. Next, we quantified the effect of hydrogen peroxide (H2O2)-associated ROS on fibroblast migration speed, and found that while H2O2-associated ROS significantly decreased relative fibroblast migration speed, pretreatment with 0.0001% or 0.001% resveratrol significantly prevented the decreases in migration speed. Furthermore, we found that LED-RL at 480, 640 and 800 J/cm2 decreased fibroblast migration speed to 83.0%, 74.4%, and 68.6% relative to matched controls, respectively. We hypothesized that these decreases in fibroblast migration speed were due to associated increases in ROS generation. Pretreatment with 0.0001% and 0.001% resveratrol prevented the LED-RL associated decreases in migration speed. Conclusion: High fluence LED-RL increases ROS and is associated with decreased fibroblast migration speed. We provide mechanistic support that the decreased migration speed associated with high fluence LED-RL is mediated by ROS, by demonstrating that resveratrol prevents high fluence LED-RL associated migration speed change. These data lend support to an increasing scientific body of evidence that high fluence LED-RL has anti-fibrotic properties. We hypothesize that our findings may result in a greater understanding of the fundamental mechanisms underlying visible light interaction with skin and we anticipate clinicians and other researchers may utilize these pathways for patient benefit.
Article
Full-text available
Fibrosis is defined as increased fibroblast proliferation and deposition of extracellular matrix components with potential clinical ramifications including organ dysfunction and failure. Fibrosis is a characteristic finding of various skin diseases which can have life-threatening consequences. These implications call for research into this topic as only a few treatments targeting fibrosis are available. In this review, we discuss oxidative stress and its role in skin fibrosis. Recent studies have implicated the importance of oxidative stress in a variety of cellular pathways directly and indirectly involved in the pathogenesis of skin fibrosis. The cellular pathways by which oxidative stress affects specific fibrotic skin disorders are also reviewed. Finally, we also describe various therapeutic approaches specifically targeting oxidative stress to prevent skin fibrosis. We believe oxidative stress is a relevant target, and understanding the role of oxidative stress in skin fibrosis will enhance knowledge of fibrotic skin diseases and potentially produce targeted therapeutic options.
Article
Full-text available
Keloid disease (KD) is an abnormal cutaneous fibroproliferative disorder of unknown aetiopathogenesis. Keloid fibroblasts (KF) are implicated as mediators of elevated extracellular matrix deposition. Aberrant secretory behaviour by KF relative to normal skin fibroblasts (NF) may influence the disease state. To date, no previous reports exist on the ability of site-specific KF to induce fibrotic-like phenotypic changes in NF or normal scar fibroblasts (NS) by paracrine mechanisms. Therefore, the aim of this study was to investigate the influence of conditioned media from site-specific KF on the cellular and molecular behaviour of both NF and NS enabled by paracrine mechanisms. Conditioned media was collected from cultured primary fibroblasts during a proliferative log phase of growth including: NF, NS, peri-lesional keloid fibroblasts (PKF) and intra-lesional keloid fibroblasts (IKF). Conditioned media was used to grow NF, NS, PKF and IKF cells over 240 hrs. Cellular behavior was monitored through real time cell analysis (RTCA), proliferation rates and migration in a scratch wound assay. Fibrosis-associated marker expression was determined at both protein and gene level. PKF conditioned media treatment of both NF and NS elicited enhanced cell proliferation, spreading and viability as measured in real time over 240 hrs versus control conditioned media. Following PKF and IKF media treatments up to 240 hrs, both NF and NS showed significantly elevated proliferation rates (p
Article
Skin cancer is unique among human cancers in its etiology, accessibility and the volume of detailed knowledge now assembled concerning its molecular mechanisms of origin. The major carcinogenic agent for most skin cancers is well established as solar ultraviolet light. This is absorbed in DNA with the formation of UV-specific dipyrimidine photoproducts. These can be repaired by nucleotide excision repair or replicated by low fidelity class Y polymerases. Insufficient repair followed by errors in replication produce characteristic mutations in dipyrimidine sequences that may represent initiation events in carcinogenesis. Chronic exposure to UVB results in disruption of the epithelial structure and expansion of pre-malignant clones which undergo further genomic changes leading to full malignancy. Genetic diseases in DNA repair, xeroderma pigmentosum, Cockayne syndrome and trichothiodystrophy, show varied elevated symptoms of sun sensitivity involving skin cancers and other symptoms including neurological degeneration and developmental delays. In humans, only xeroderma pigmentosum shows high levels of cancer, but mouse strains, with any of the genes corresponding to these diseases knocked-out, show elevated skin carcinogenesis. The three major skin cancers exhibit characteristic molecular changes defined by certain genes and associated pathways. Squamous cell carcinoma involves mutations in the p53 gene; basal cell carcinoma involves mutations in the PATCHED gene, and melanoma in the p16 gene. The subsequent development of malignant tumors involves many additional genomic changes that have yet to be fully cataloged.
Article
Background and objective: Low-level laser therapy (LLLT) or photobiomodulation therapy is emerging as a promising new therapeutic option for fibrosis in different damaged and/or diseased organs. However, the anti-fibrotic potential of this treatment needs to be elucidated and the cellular and molecular targets of the laser clarified. Here, we investigated the effects of a low intensity 635 ± 5 nm diode laser irradiation on fibroblast-myofibroblast transition, a key event in the onset of fibrosis, and elucidated some of the underlying molecular mechanisms. Materials and methods: NIH/3T3 fibroblasts were cultured in a low serum medium in the presence of transforming growth factor (TGF)-β1 and irradiated with a 635 ± 5 nm diode laser (continuous wave, 89 mW, 0.3 J/cm(2) ). Fibroblast-myofibroblast differentiation was assayed by morphological, biochemical, and electrophysiological approaches. Expression of matrix metalloproteinase (MMP)-2 and MMP-9 and of Tissue inhibitor of MMPs, namely TIMP-1 and TIMP-2, after laser exposure was also evaluated by confocal immunofluorescence analyses. Moreover, the effect of the diode laser on transient receptor potential canonical channel (TRPC) 1/stretch-activated channel (SAC) expression and activity and on TGF-β1/Smad3 signaling was investigated. Results: Diode laser treatment inhibited TGF-β1-induced fibroblast-myofibroblast transition as judged by reduction of stress fibers formation, α-smooth muscle actin (sma) and type-1 collagen expression and by changes in electrophysiological properties such as resting membrane potential, cell capacitance and inwardly rectifying K(+) currents. In addition, the irradiation up-regulated the expression of MMP-2 and MMP-9 and downregulated that of TIMP-1 and TIMP-2 in TGF-β1-treated cells. This laser effect was shown to involve TRPC1/SAC channel functionality. Finally, diode laser stimulation and TRPC1 functionality negatively affected fibroblast-myofibroblast transition by interfering with TGF-β1 signaling, namely reducing the expression of Smad3, the TGF-β1 downstream signaling molecule. Conclusion: Low intensity irradiation with 635 ± 5 nm diode laser inhibited TGF-β1/Smad3-mediated fibroblast-myofibroblast transition and this effect involved the modulation of TRPC1 ion channels. These data contribute to support the potential anti-fibrotic effect of LLLT and may offer further informations for considering this therapy as a promising therapeutic tool for the treatment of tissue fibrosis.
Article
Background/PurposeUltraviolet A1 (UVA1) phototherapy has been used for over 15 years in the United States, primarily for the treatment of localized sclerosis and various sclerosing disorders.The objective was to describe use of UVA1 for dermatoses beyond localized sclerosis at two academic institutions.Methods Data from 83 patients treated with low- (20-40 J/cm2), medium-(>40-80 J/cm2), and high- (>80-120 J/cm2) dose UVA1 phototherapy was retrospectively analyzed. The mean individual treatment dose (J/cm2), the mean number of sessions, and the mean total dose (J/cm2) were evaluated. Effectiveness was assessed by reviewing clinical examination notes from office visits.ResultsGood therapeutic efficacy was seen in patients with systemic sclerosis (SS, 16 patients), graft-versus-host-disease (GVHD, 25 patients), and nephrogenic systemic fibrosis (NSF, 17 patients). A statistically significant a dose-response association was observed in the cases of SS, GVHD and NSF. Likelihood of clinical improvement from UVA1 phototherapy was very likely for medium- and high-dose regimens in SS, while this level of improvement was only observed in GVHD and NSF patients receiving high-dose UVA1.ConclusionUVA1 phototherapy is effective and safe in the treatment of GVHD, NSF, SS, and mast cell disorders. High-dose regimens appear to be more effective than medium and low-dose regimens for NSF and GVHD, while medium- and high-dose regimens outperform low-dose UVA1 in SS.This article is protected by copyright. All rights reserved.
Article
Deciding whether to treat a scar or leave it alone depends on accurate diagnosis of scar type and scar site, symptoms, severity, and stigma