ArticlePDF Available

E-cigarettes and flavorings induce inflammatory and pro-senescence responses in oral epithelial cells and periodontal fibroblasts

Authors:

Abstract and Figures

Electronic-cigarettes (e-cigs) represent a significant and increasing proportion of tobacco product consumption, which may pose an oral health concern. Oxidative/carbonyl stress via protein carbonylation is an important factor in causing inflammation and DNA damage. This results in stress-induced premature senescence (a state of irreversible growth arrest which re-enforces chronic inflammation) in gingival epithelium, which may contribute to the pathogenesis of oral diseases. We show that e-cigs with flavorings cause increased oxidative/carbonyl stress and inflammatory cytokine release in human periodontal ligament fibroblasts, Human Gingival Epithelium Progenitors pooled (HGEPp), and epigingival 3D epithelium. We further show increased levels of prostaglandin-E2 and cycloxygenase-2 are associated with upregulation of the receptor for advanced glycation end products (RAGE) by e-cig exposure-mediated carbonyl stress in gingival epithelium/tissue. Further, e-cigs cause increased oxidative/carbonyl and inflammatory responses, and DNA damage along with histone deacetylase 2 (HDAC2) reduction via RAGE-dependent mechanisms in gingival epithelium. A greater response is elicited by flavored e-cigs. Increased oxidative stress, pro-inflammatory and pro-senescence responses (DNA damage and HDAC2 reduction) can result in dysregulated repair due to proinflammatory and pro-senescence responses in periodontal cells. These data highlight the pathologic role of e-cig aerosol and its flavoring to cells and tissues of the oral cavity in compromised oral health.
Content may be subject to copyright.
Oncotarget1
www.impactjournals.com/oncotarget
www.impactjournals.com/oncotarget/ Oncotarget, Advance Publications 2016
E-cigarettes and avorings induce inammatory and pro-
senescence responses in oral epithelial cells and periodontal
broblasts
Isaac K. Sundar1, Fawad Javed2, Georgios E. Romanos3, Irfan Rahman1
1Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY
2Department of General Dentistry, Eastman Institute for Oral Health University of Rochester, Rochester, NY
3Department of Periodontology, School of Dental Medicine, Stony Brook University, Stony Brook, NY
Correspondence to: Irfan Rahman, email: irfan_rahman@urmc.rochester.edu
Keywords: e-cigarettes, e-juices, RAGE, COX2, PGE2
Received: August 02, 2016 Accepted: October 14, 2016 Published: October 24, 2016
ABSTRACT
Electronic-cigarettes (e-cigs) represent a signicant and increasing proportion
of tobacco product consumption, which may pose an oral health concern. Oxidative/
carbonyl stress via protein carbonylation is an important factor in causing inammation
and DNA damage. This results in stress-induced premature senescence (a state of
irreversible growth arrest which re-enforces chronic inammation) in gingival
epithelium, which may contribute to the pathogenesis of oral diseases. We show that
e-cigs with avorings cause increased oxidative/carbonyl stress and inammatory
cytokine release in human periodontal ligament broblasts, Human Gingival Epithelium
Progenitors pooled (HGEPp), and epigingival 3D epithelium. We further show increased
levels of prostaglandin-E2 and cycloxygenase-2 are associated with upregulation of
the receptor for advanced glycation end products (RAGE) by e-cig exposure-mediated
carbonyl stress in gingival epithelium/tissue. Further, e-cigs cause increased oxidative/
carbonyl and inammatory responses, and DNA damage along with histone deacetylase
2 (HDAC2) reduction via RAGE-dependent mechanisms in gingival epithelium. A greater
response is elicited by avored e-cigs. Increased oxidative stress, pro-inammatory
and pro-senescence responses (DNA damage and HDAC2 reduction) can result
in dysregulated repair due to proinammatory and pro-senescence responses in
periodontal cells. These data highlight the pathologic role of e-cig aerosol and its
avoring to cells and tissues of the oral cavity in compromised oral health.
INTRODUCTION
The use of electronic-cigarettes (e-cigs) is increasing
in the United States, which may pose oral health concerns
[1]. E-cigs are battery operated devices, which consist
of a metal heating element in a stainless steel shell, a
cartridge, an atomizer and a battery. The heating element
vaporizes a solution containing a mixture of chemicals
including nicotine and other additives/humectants, such as
base/carrying agents propylene glycol, glycerin/glycerol,
and avoring agents including fruit and candy avors.
Apart from inhaled nicotine, variable levels of aldehydes
and carbonyls are detected in e-cig aerosols during
vaporizations [2, 3]. Aldehyde causes carbonyl/oxidative
stress, DNA adducts/damage, as well as stress-induced
cellular senescence (a state of irreversible growth arrest
which re-enforces chronic inammation) [4, 5] leading to
oral health problems [6–8].
Periodontal disease is characterized by chronic
inammation of the supporting tissues of the teeth.
Periodontal ligament and gingival broblasts as well as
epithelial cells are the most abundant structural cells in
periodontal tissue. Upon stimulation or stress, these cells
are able to incite and maintain inammatory responses
[9]. There is an association between smoking and tooth
loss, periodontal attachment level, deeper periodontal
pockets, and more extensive alveolar bone loss along with
the destruction of connective tissue and matrix, leading
Oncotarget2
www.impactjournals.com/oncotarget
to increased risk of periodontitis [10]. Clinical studies
[11, 12] have also shown that habitual tobacco smokers
exhibit a greater number of sites with plaque accumulation,
clinical attachment loss and probing depth (≥ 4 mm) as
compared to individuals who had never used tobacco in
any form. It is important to mention that bleeding upon
probing (a classical marker of periodontal disease activity)
is masked in tobacco smokers than non-smokers [11, 12].
This most likely occurs as a result of the vasoconstrictive
effect of nicotine (nicotine is the main component in
e-cigs) on gingival blood vessels. Therefore, tobacco
smokers may remain unaware of ongoing periodontal
destruction until the inammatory process reaches a stage
where tooth mobility becomes evident.
We have recently shown the comparable oxidants/
reactive oxygen species (ROS) reactivity in e-cig
aerosols compared to conventional cigarette smoke
[13, 14]. Smoking tobacco contributes to the progression
of periodontal disease [10]. However, there is no
information available regarding the e-cig aerosols vaping
on periodontal/gingival oral health effects, especially in
response to e-cig avoring agents and nicotine. Periodontal/
gingival cells in the oral cavity are the rst targets by
aerosols of e-cigs. Furthermore, the effects of e-cig aerosols
on carbonyl stress, inammation, and pro-senescence have
not been studied on oral health. Here, we have determined
the mechanism of gingival epithelial inammation and pro-
senescence by e-cig aerosols with avorings in human oral
epithelial cells and periodontal ligament broblasts.
RESULTS
E-cigarette exposure in human periodontal
ligament broblasts (HPdLFs) and human
gingival epithelium progenitors, pooled
(HGEPp) increases protein carbonylation and
pro-inammatory responses
It is possible that ROS produced by e-cig vapors
in cultured cells can augment protein carbonylation. The
oxyblot assay was performed for immunodetection of the
carbonyl groups that were introduced into protein side
chains by e-cig-induced oxidative stress. We found that
e-cig avoring (BLU® Classic Tobacco, 16 mg nicotine
and Magnicent Menthol, zero nicotine) had different
levels of protein oxidation as conrmed by OxyBlot.
E-cigarette vapors from BLU® Classic Tobacco showed
signicant increase in protein carbonylation compared
to air exposed controls. E-cigarette vapors from BLU®
Magnicent Menthol avor also showed a trend towards
increased protein carbonylation but not signicant
compared to the control (Figure 1A–1B).
HPdLFs and HGEPp were exposed to BLU®
e-cig vapors (Classic Tobacco, 16 mg nicotine; and
Magnicent Menthol, zero nicotine) using ALI system
for 15 min. Conditioned medium was collected from
both air exposed (control) and e-cig vapor exposed cells
24 hrs post ALI exposure. IL-8 secretion in conditioned
medium 24 hrs after ALI exposure was signicantly
increased in both BLU® e-cig vapors. PGE2 secretion
showed a trend of incremental increase, but was not
signicant compared to air exposed controls (Figure
1C). IL-8 release was signicantly higher in both BLU
®
Classic Tobacco (16 mg nicotine) and Magnicent
Menthol (zero nicotine) e-cig vapor exposed HPdLF cells
compared to the controls. PGE
2
release was signicantly
higher in BLU® Magnicent Menthol (zero nicotine)
e-cig vapor exposed cells compared to the controls
(Figure 1C). Similarly, HGEPp cells also showed an
increased trend in IL-8 release in BLU® Classic Tobacco
(16 mg nicotine) e-cig vapor exposed cells compared to
the controls. We did not see a signicant increase in IL-8
release in HGEPp cells exposed to BLU® Magnicent
Menthol (zero nicotine) e-cig vapor compared to control
(Figure 1D). Overall, e-cig vapors with avoring induce
protein carbonylation and increase in pro-inammatory
cytokines release which are indicative of oxidative stress
and inammatory response cause by e-cig vapors in
HPdLF and HGEPp.
E-cigarette exposure in human periodontal
ligament broblasts (HPdLFs) increases
inammation and DNA damage markers
HPdLFs exposed to e-cig vapor from BLU® e-cig
(Classic Tobacco, 16 mg nicotine; and Magnicent
Menthol, zero nicotine) for 15 min using ALI system and
incubated for 24 h. Then, we measured several markers of
inammation such as PGE2-mediated COX-2 induction,
HDAC2, S100A8, RAGE and phosphorylated γH2A.X
(Ser139) in whole cell lysates. We found E-cig avoring
BLU® Classic Tobacco (16 mg nicotine) and Magnicent
Menthol (zero nicotine) showed a differential effect on
levels of COX-2, HDAC2, S100A8, RAGE and γH2A.X
in HPdLFs in vitro (Figure 2A). HPdLFs exposed to
BLU® e-cig vapors (Magnicent Menthol, zero nicotine)
showed a signicant increase in all the inammatory
markers (COX-2, S100A8, RAGE), a trend towards
reduced HDAC2 and increased phosphorylated γH2A.X
(Ser139), a DNA damage marker compared to control.
HPdLFs exposed to BLU® e-cig (Classic Tobacco, 16 mg
nicotine) also showed a signicant increase in S100A8 and
γH2A.X, and an increased trend in COX-2 compared to
the controls. We did not observe any signicant increase in
RAGE or a signicant decrease in HDAC2 levels in BLU
®
E-cig vapor (Classic Tobacco, 16 mg nicotine) exposed
HPdLFs compared to the controls (Figure 2A). Likewise,
the HPdLFs exposed to e-cig vapor from BLU® e-cig
(Classic Tobacco, 16 mg nicotine) showed a signicant
increase in DNA damage as measured by the percentage of
DNA in tail using the Comet assay (Figure 2B). Overall,
we showed that e-cig vapors with avoring differentially
Oncotarget3
www.impactjournals.com/oncotarget
affects inammatory response and DNA damage markers
as a result of oxidative stress and inammatory response
caused by e-cig vapors in HPdLFs.
E-cigarette exposure in human gingival
epithelium progenitors, pooled (HGEPp)
increases inammation and DNA damage
markers
We found E-cig avoring BLU® Classic Tobacco
(16 mg nicotine) and Magnicent Menthol (zero
nicotine) showed differential effects on levels of COX-
2, S100A8, RAGE and γH2A.X in HGEPp cells in
vitro (Figure 2A). HGEPp cells exposed to BLU® e-cig
vapors (Classic Tobacco, 16 mg nicotine and Magnicent
Menthol, zero nicotine) showed a signicant increase
in the inammatory markers (COX-2 and RAGE), and
DNA damage marker (phosphorylated γH2A.X Ser139)
compared to the controls. The effect of BLU
®
e-cig vapors
(Magnicent Menthol, zero nicotine) on HGEPp cells
was signicantly higher compared to BLU
®
e-cig vapors
Classic Tobacco (16 mg nicotine) and the controls. We
did not observe any signicant increase in COX-2 and
S100A8 levels in BLU
®
e-cig vapor from Classic Tobacco
(16 mg nicotine) as well as S100A8 levels in Magnicent
Menthol (zero nicotine) exposed HGEPp cells compared
to the controls (Figure 3). Overall, we conrmed that
e-cig vapors with avoring affects inammatory response
and DNA damage markers as a result of oxidative stress
and inammatory response caused by e-cig vapors in
HGEPp cells.
E-cigarette exposure in human 3D in vitro model
of EpiGingival tissue increases inammation and
DNA damage markers
Human 3D model of EpiGingival tissues were
exposed to e-cig vapor from BLU
®
(Classic Tobacco, 16 mg
nicotine; and Magnicent Menthol, zero nicotine) for 15 min
using the modied ALI system without culture media. The
human 3D EpiGingival tissue insert was contained within
35 mm culture dishes along with 900 μl culture medium
during e-cig vapor exposure, and incubated for 24 h. Then,
we measured pro-inammatory cytokines IL-8 and PGE2
Figure 1: E-cig aerosol resulted in increased protein carbonylation and inammatory responses in human periodontal
ligament broblasts (HPdLFs) and human gingival epithelium progenitors, pooled (HGEPp). HPdLFs (AC) and HGEPp
(D) were exposed to aerosols from BLU® e-cig (Classic Tobacco, 16 mg nicotine; and Magnicent Menthol, ‘zero’ nicotine) (2 puffs/min;
4–5 sec/puff every 25 sec) using air-liquid interface system for 15 min, and then incubated at 37°C and 5% CO2 for 24 h. (A, B) Protein
carbonylation in cell lysates was determined by the oxyblots. Gel pictures shown are representative of at least 3 separate experiments for
protein carbonylation. (C, D) Levels of IL-8 and PGE2 in supernatants were determined by ELISA. Data are means ± SE (n = 3–6/group)
and signicance determined using 1-way ANOVA. *P < 0.05, vs. air. MWM: Molecular weight markers; Negative: without derivitizing
reagent 2,4-dinitrophenylhydrazine (DNPH).
Oncotarget4
www.impactjournals.com/oncotarget
in conditioned medium collected 24 h post last exposure.
Markers of inammation and DNA damage, such as RAGE,
PGE2-mediated COX-2 induction and γH2A.X were
measured using whole tissue lysates from 3D EpiGingival
cultures. We found e-cig avoring BLU® Classic Tobacco
(16 mg nicotine) and Magnicent Menthol (zero nicotine)
showed an increasing trend in the levels of IL-8 release
and a signicant increase in the levels of PGE2 release
compared to the controls (Figure 4A). Additionally, only
e-cig avoring BLU
®
Magnicent Menthol (zero nicotine)
showed an increasing trend in RAGE, COX-2, and γH2A.X
(immunoblot analysis and immunohistochemistry) in 3D
EpiGingival tissues in vitro compared to the controls (Figure
4B–4C). The effect of BLU® e-cig vapors (Magnicent
Menthol, zero nicotine) on 3D EpiGingival tissues was
signicantly higher compared to BLU
®
e-cig vapors Classic
Tobacco (16 mg nicotine) and the controls. Overall, we
reproduced the above ndings in 3D models that e-cig
vapors with avoring differentially affects inammatory
response and DNA damage markers as a result of oxidative
stress and inammatory response caused by e-cig vapors in
3D in vitro model of EpiGingival tissues.
DISCUSSION
In this study, we show that e-cigs with avorings
cause increased oxidative/carbonyl stress and inammatory
cytokine release in human periodontal ligament broblasts,
Human Gingival Epithelium Progenitors pooled (HGEPp),
and EpiGingival 3D epithelium. We further show increased
levels of prostaglandin-E2 and cycloxygenase-2 were
associated with upregulation of the receptor for advanced
glycation end products (RAGE) by e-cig exposure-
mediated carbonyl stress in gingival epithelium/tissue.
Further, e-cigs cause increased oxidative/carbonyl stress
and inammatory responses, and DNA damage along with
histone deacetylase 2 (HDAC2) reduction via RAGE-
dependent mechanisms in gingival epithelium, with
greater response by avored e-cigs. Increased oxidative
stress, pro-inammatory and pro-senescence responses
(DNA damage and HDAC2 reduction) can result in
dysregulated repair due to proinammatory and pro-
senescence responses in periodontal cells. Our data also
implicate that e-cig affects the regenerative potential of
human progenitor cells due to increased inammatory and
DNA damage responses. It is well known that the mTOR
pathway is activated by most oncogenes that induce
cellular senescence (i.e., by Ras, Raf, MEK, and Akt).
Further, the mechanistic target of rapamycin (mTOR) is
generally activated in proliferating cells. During acute
DNA damage, mTOR induces DNA damage response
(DDR) and cell cycle arrest (induction of p21 and 16) [15].
Such arrested cells where mTOR is active play an
important role in geroconversion (converts quiescence into
senescence) to their pro-senescent phenotype [15, 16]. In
case of oncogene-induced senescence, DDR causes cell
Figure 2: E-cig vapor exposure caused inammatory responses and DNA damage in human periodontal ligament
broblasts (HPdLFs). (A) HPdLFs were exposed to aerosols from BLU® e-cig (Classic Tobacco, 16 mg nicotine; and Magnicent
Menthol, ‘zero’ nicotine) for 15 min (2 puffs/min; 4–5 sec/puff every 25 sec) using air-liquid interface system, and then incubated at 37°C
and 5% CO2 for 24 h. (A) Levels of COX-2, S100A8, RAGE, γH2AX, and HDAC2 in cell lysates were measured by Western blotting. (B)
DNA damage, indicated by an increase in uorescent tail length, was measured by the Comet assay. Dash lines indicate DNA in tails. Data
are means ± SE (n = 3–6/group) and signicance determined using 1-way ANOVA or 2-tail t-test. *P < 0.05, **P < 0.01, ***P < 0.001
vs. air.
Oncotarget5
www.impactjournals.com/oncotarget
cycle arrest, leading to cell senescence [16]. This may
be one of the mechanisms of e-cigarette induce DDR
response via mTOR activation.
Conventional cigarette smoke has been shown to
cause deleterious effects on oral and periodontal health
[10]. However, the role of e-cig vaping and its association
with carbonyl stress, inammation, and DNA damage-
triggered senescence on oral/periodontal epithelium
remains unknown. Periodontal ligament and gingival
broblasts as well as epithelial cells are the most abundant
structural cells in periodontal tissue, and are the direct
targets of e-cigs upon vaping. Upon stimulation or stress,
these cells are able to incite and maintain inammatory
responses [9]. There is an association between smoking
and tooth loss, periodontal attachment loss, deeper
periodontal pockets, and more extensive alveolar bone
loss along with the destruction of connective tissue and
matrix , leading to an increased risk of periodontitis [10].
However, no studies are available to document the effects
of e-cig vaping especially in response to e-cig avoring
agents on periodontal health in terms of oxidative/
carbonyl stress and inammation in human gingival
epithelial cells in vitro.
We have recently shown oxidants/ROS reactivity
from e-cig aerosols is comparable to conventional
cigarette smoke [13, 14]. We show that e-cig avoring
caused protein oxidation as reected in increased protein
carbonylation. This was associated with increased IL-8
and PGE2 secretion from HPdLFs and HGEPp cells
upon exposure to e-cig aerosols. Direct exposure to
e-liquids (this is not the case when users vape e-cigs i.e.
users do not consume e-liquids) has also been shown
to produce harmful effects in periodontal ligament
and gingival broblasts in culture [17, 18]. While the
contribution of smoking tobacco to the progression
of periodontal disease and other adverse oral health
outcomes is well described [10, 19], no information is
available regarding the impact of e-cig aerosols vaping
on periodontal/gingival oral health effects, especially in
response to e-cig avoring agents. We determined the
effect of avoring on oxidative and pro-inammatory
responses, and upon exposure of periodontal ligament
broblasts, Human Gingival Epithelium Progenitors
pooled (HGEPp), and epigingival 3D epithelium to
menthol avoring agent resulted in increased oxidative/
carbonyl stress, and IL-8 release. Menthol acts on
transient receptor potential ankyrin 1 (TRPA1) receptors
to activate inammatory responses [20, 21]. It may be
conceived that activation of TRPA receptors by e-cig
aerosols will drive COX-PGE2 mediated responses
in periodontal tissues, leading to augmentation of
inammatory and pro-brotic and pro-carcinogenic
responses. However, further studies are required to
understand the augmented response by BLU® menthol
avoring than BLU® classic tobacco.
Protein carbonylation leads to autoantibody
production which may lead to destruction of matrix
and bone loss during periodontitis [6, 7]. Further, it is
possible that carbonyls/aldehydes play an important
role in e-cig aerosol-induced oral toxicity. Conventional
tobacco smoke is known to cause oxidative burden
leading to DNA damage and inammatory responses
[22]. The RAGE is a pattern-recognition receptor
implicated in immune and inammatory diseases
including dental pulp inammation and periodontitis
[23–27]. RAGE is involved in smoking-related disorders
and known to cause cellular senescence via oxidant stress
[28, 29]. However, the mechanism of RAGE-mediated
Figure 3: E-cig vapor exposure caused inammatory responses and DNA damage in human gingival epithelium
progenitors, pooled cells (HGEPp). (A) HGEPp cells were exposed to aerosols from BLU® e-cig (Classic Tobacco, and Magnicent
Menthol) (2 puffs/min; 4–5 sec/puff every 25 sec) using air-liquid interface system for 15 min, and then incubated at 37°C and 5% CO2 for
24 h. Levels of COX-2, S100A8, RAGE, and γH2AX in cell lysates were measured by Western blotting. Data are means ± SE (n = 3–6/
group) and signicance determined using 1-way ANOVA. *P < 0.05, ***P < 0.001, vs. air.
Oncotarget6
www.impactjournals.com/oncotarget
Figure 4: E-cig vapor caused inammatory responses and DNA damage in normal human 3D in vitro model of
EpiGingival tissues. Normal human 3D in vitro model of EpiGingival tissue (Cat#: GIN-100, MatTek) were exposed to aerosols from
BLU® e-cig (Classic Tobacco, and Magnicent Menthol) using air-liquid interface system for 15 min, and then incubated at 37°C and 5%
CO2 for 24 h. (A) Levels of IL-8 and PGE2 in culture media were determined by ELISA. (B) Levels of RAGE, COX-2, and γH2AX in tissue
lysates were measured by Western blotting. (C) Representative images of EpiGingival tissues used for ALI exposures stained with H&E
(showing histological features) and γH2AX staining 24 h post exposure to control (air) and avored e-cig aerosols. Immunohistochemistry
revealed a distinct staining in both the avored BLU e-cig exposed EpiGingival tissues for γH2AX. Data are means ± SE (n = 4–6/group)
and signicance determined using 1-way ANOVA. ***P < 0.001,vs. air.
Oncotarget7
www.impactjournals.com/oncotarget
signaling especially via its ligand S100A8 as a
susceptible factor in inducing gingival epithelial
inammation and senescence by e-cigs is not known.
E-cig vapor exhibited signicant inammatory response
(COX2, RAGE S100A8), DNA damage as determined
by the Comet assay and γ-H2AX levels (a marker of
DNA damage) in gingival epithelium/tissue. Our data
showing inammatory and pro-DNA damage responses
are unique in light of primary cells and 3D tissue culture
models which mimic closely with users vaping of e-cigs.
Our results further attest the pro-oxidant, DNA damaging
and pro-inammatory effects of e-cig vapor exposure.
The increased levels of pro-inammatory mediators IL-8
and PGE2 would cause remodeling of the ECM during
periodontitis by e-cig due to cellular senescence, where
these cells have secretory phenotype to perpetuate the
inammatory responses.
It has been reported that outcomes of periodontal
therapy are compromised in smokers compared with
non-smokers [30, 31]. A variety of mechanisms have
been proposed in this regard. For example, increased
expression of RAGE occurs in gingival epithelial cells
of smokers as compared to non-smokers. Furthermore,
it has been reported that nornicotine (a metabolite of
nicotine), upregulates RAGE expression in the gingivae
of smokers and elicits a proinammatory response by
stimulating the secretion of cytokines and ROS which
are involved in destruction of the periodontal tissue
[32]. The vasoconstrictive effects of nicotine increase
platelet adhesiveness, increases the risk of microvascular
occlusion and causes tissue ischemia [32]. Furthermore,
tobacco smoking is also associated with catecholamines
release resulting in vasoconstriction and decreased
tissue perfusion [33]. Therefore, it is hypothesized that
the outcomes of periodontal surgery are compromised
in e-cig users compared with non-smokers/non-users
through the mechanisms comparable to those stated above.
However, further studies are needed to test this contention
in a clinical cohort of users and non-users of e-cigs vs
conventional smokers.
In conclusion, our data showed that e-cig aerosol
cause increased oxidative/carbonyl stress and inammatory
responses, and cellular senescence associated with
persistent DNA damage via RAGE-HDAC2-dependent
mechanisms in gingival epithelium, with greater response
by avored e-cigs. Further understanding of the chronic
effect of vaping could lead to molecular mechanisms for
susceptibility (inammatory, DNA damage and senescence
responses) to the development of periodontitis, and
therapeutic targets or biomarkers in determining vaping-
avoring mediated oral complications in cells and tissues
of the oral cavity. Our data also implicate that e-cig affects
the regenerative potential of human progenitor cells due
to increased inammatory and DNA damage responses.
Overall, our data suggest the pathogenic role of e-cig
aerosol to cells and tissues of the oral cavity, leading to
compromised periodontal health.
MATERIALS AND METHODS
E-cigarettes
BLU® rechargeable e-cigs (Lorillard Technologies,
Inc.) containing two different disposable cartomizers
[Flavors: classic tobacco (16 mg nicotine), and magnicent
menthol (zero nicotine or 13–16 mg nicotine)] with pre-
loaded e-liquid were used. The BLU® e-cigarette device
and disposable cartomizer cartridges were purchased from
local retailers.
Cell culture and air-liquid interface (ALI)
culture/exposures
Clonetics
TM
Human periodontal ligament broblast
(HPdLF, CC-7049; Lonza) were grown at 37ºC in 5% CO
2
incubator to 80–90% conuence in stromal cell medium
(SCGM
TM
BulletKit, Lonza; CC-3205) supplemented with
hFGF-B (0.5 ml), insulin (0.5 ml), FBS (25 ml) and GA-
1000 (0.5 ml) according to manufacturer recommendations.
Human gingival epithelium progenitors, (HGEPp; gingival
epithelial cells) were grown in CnT-Prime medium (CnT-
PR), epithelial culture medium as recommended by
CELLnTEC Advanced Cell Systems. Transwell cultures
were then placed into air-liquid interface (ALI) exposure
chamber [14, 34]. BLU® e-cigarette vapor (Classic Tobacco
containing 16 mg nicotine or Magnicent Menthol avor
containing zero or 13–16 mg nicotine) was drawn into the
ALI exposure chamber (2 puffs every 1 min), 4–5 second
puff followed by 25 second pause for different time
durations 5, 10, and 15 minutes respectively.
Human EpiGingival tissue model
Human gingival tissues (EpiGingival, GIN-100)
were obtained from MatTek Corporation (Ashland,
MA). The 3D tissue model is a reconstructed oral
epithelial tissue that are derived from human primary oral
keratinocytes. This model and allowed to differentiate to
a structure characteristic to that of in vivo. EpiGingival
tissues were exposed to air (control) or BLU
®
e-cigarette
vapors (Classic Tobacco and Magnicent Menthol). After
15 min exposure, the conditioned medium was collected to
measure pro-inammatory cytokines and tissues harvested
for Western blotting and immunohistochemistry according
to the recommendations of the manufacturer. The protein
levels were measured using a BCA kit (Pierce, IL, USA).
Comet assay
Comet assay was performed as per the instructions
of the manufacturer (Trevigen, Gaithersburg, MD) [34].
Comet images were captured using a Nikon ECLIPSE Ni
uorescent microscope. The images were analyzed using
OpenComet software. The extent of DNA damage was
expressed as a measure of percentage of DNA in tail.
Oncotarget8
www.impactjournals.com/oncotarget
Protein carbonylation/oxyblot
Protein oxidation was determined using the
OxyBlot protein oxidation detection kit following the
manufacturer’s instruction (Millipore, S7150). Equal
amount of protein was loaded for oxyblot analysis, and
the results were quantied by densitometry using Image J.
Pro-inammatory cytokine analysis
Following 24 hrs after BLU® e-cigarette vapor
exposure using air-liquid interface approach, conditioned
media was collected and stored at −80ºC for measuring pro-
inammatory mediators. IL-8 (Life Technologies, Carlsbad,
CA) and PGE
2
(Cayman Chemical, Ann Arbor, MI) levels
were measured by enzyme-linked immunosorbent assay
(ELISA) according to manufacturer’s instructions.
Western blotting
For Western blots, 25 μg protein samples were
separated on a 4–15% gradient and 7.5% SDS-PAGE gels.
Then probed with specic primary antibodies (1:1000
dilution in 5% milk in PBS containing 0.1% Tween
20 (v/v), such as anti-COX2 (Cayman chemical; #160112),
HDAC2 (ab32117), S100A8 (ab92331), RAGE (ab37647)
and γH2A.X phospho S139 (ab2893) from Abcam at 4°C
overnight. The bound complexes were detected using
ECL method with the ChemiDocTM MP Imaging System
(Bio-Rad). Equal loading of the samples was determined
by quantitation of proteins and by stripping and re-
probing membranes for β-actin or GAPDH (Santa Cruz
Biotechnology, sc-1616 and sc-365062) and the results
were quantied by densitometry using Image J.
Statistical analysis
Statistical analysis of signicance was calculated
using unpaired Student’s t-test for comparison between
two groups control vs. e-cigarette. Probability of
signicance compared to control for more than
two treatment groups (different e-cigarette avors)
was analyzed by 1-way ANOVA (Tukey’s multiple
comparisons test) using GraphPad Prism 6 as indicated in
gure legends. The results are shown as the mean ± SEM
unless otherwise indicated. P < 0.05 is considered as
statistically signicant.
ACKNOWLEDGMENTS
We thank Ms. Janice Gerloff for technical assistance.
CONFLICTS OF INTEREST
The authors have declared that no conicts of
interests exists.
GRANT SUPPORT
This study was supported by the
NIH 2R01HL085613 and 3R01HL085613-07S2 (to I.R.).
Authors’ contributions
IKS, IR: Conceived and designed the experiments;
IKS: Performed the experiments; IKS: Analyzed the data;
FJ, GER: Provided inputs and thoughtful discussion as
well as edited the manuscript; IKS, IR: Wrote and edited
the manuscript.
REFERENCES
1. Regan AK, Promoff G, Dube SR, Arrazola R. Electronic
nicotine delivery systems: adult use and awareness of the
‘e-cigarette’ in the USA. Tobacco control. 2013; 22:19–23.
2. Cheng T. Chemical evaluation of electronic cigarettes.
Tobacco control. 2014; 23:11–17.
3. Kosmider L, Sobczak A, Fik M, Knysak J, Zaciera M, Kurek
J, Goniewicz ML. Carbonyl compounds in electronic cigarette
vapors: effects of nicotine solvent and battery output voltage.
Nicotine & tobacco research : ofcial journal of the Society
for Research on Nicotine and Tobacco. 2014; 16:1319–1326.
4. Baraibar MA, Liu L, Ahmed EK, Friguet B. Protein
oxidative damage at the crossroads of cellular senescence,
aging, and age-related diseases. Oxidative medicine and
cellular longevity. 2012; 2012:919832.
5. Satoh R, Kishino K, Morshed SR, Takayama F, Otsuki S,
Suzuki F, Hashimoto K, Kikuchi H, Nishikawa H, Yasui T,
Sakagami H. Changes in uoride sensitivity during in vitro
senescence of normal human oral cells. Anticancer research.
2005; 25:2085–2090.
6. Pradeep AR, Ramchandraprasad MV, Bajaj P, Rao NS,
Agarwal E. Protein carbonyl: An oxidative stress marker in
gingival crevicular uid in healthy, gingivitis, and chronic
periodontitis subjects. Contemporary clinical dentistry. 2013;
4:27–31.
7. Baltacioglu E, Akalin FA, Alver A, Deger O, Karabulut E.
Protein carbonyl levels in serum and gingival crevicular
uid in patients with chronic periodontitis. Archives of oral
biology. 2008; 53:716–722.
8. Canakci CF, Tatar A, Canakci V, Cicek Y, Oztas S, Orbak
R. New evidence of premature oxidative DNA damage:
mitochondrial DNA deletion in gingival tissue of patients with
periodontitis. Journal of periodontology. 2006; 77:1894–1900.
9. Ara T, Kurata K, Hirai K, Uchihashi T, Uematsu T, Imamura Y,
Furusawa K, Kurihara S, Wang PL. Human gingival broblasts
are critical in sustaining inammation in periodontal disease.
Journal of periodontal research. 2009; 44:21–27.
10. Javed F, Bashir Ahmed H, Romanos GE. Association
between environmental tobacco smoke and periodontal
disease: a systematic review. Environmental research. 2014;
133:117–122.
Oncotarget9
www.impactjournals.com/oncotarget
11. Javed F, Al-Askar M, Samaranayake LP, Al-Hezaimi K.
Periodontal disease in habitual cigarette smokers and
nonsmokers with and without prediabetes. The American
journal of the medical sciences. 2013; 345:94–98.
12. Javed F, Nasstrom K, Benchimol D, Altamash M,
Klinge B, Engstrom PE. Comparison of periodontal
and socioeconomic status between subjects with type 2
diabetes mellitus and non-diabetic controls. Journal of
periodontology. 2007; 78:2112–2119.
13. Lerner CA, Sundar IK, Watson RM, Elder A, Jones R,
Done D, Kurtzman R, Ossip DJ, Robinson R, McIntosh S,
Rahman I. Environmental health hazards of e-cigarettes
and their components: Oxidants and copper in e-cigarette
aerosols. Environmental pollution. 2015; 198C:100–107.
14. Lerner CA, Rutagarama P, Ahmad T, Sundar IK, Elder A,
Rahman I. Electronic cigarette aerosols and copper
nanoparticles induce mitochondrial stress and promote
DNA fragmentation in lung broblasts. Biochemical and
biophysical research communications. 2016; 477:620–625.
15. Blagosklonny MV. Cell cycle arrest is not yet senescence,
which is not just cell cycle arrest: terminology for TOR-
driven aging. Aging (Albany NY). 2012; 4:159–165.
16. Blagosklonny MV. Molecular damage in cancer: an
argument for mTOR-driven aging. Aging (Albany NY).
2011; 3:1130–1141.
17. Sancilio S, Gallorini M, Cataldi A and di Giacomo V.
Cytotoxicity and apoptosis induction by e-cigarette uids
in human gingival broblasts. Clinical oral investigations.
2016; 20:477–483.
18. Willershausen I, Wolf T, Weyer V, Sader R, Ghanaati S,
Willershausen B. Inuence of E-smoking liquids on human
periodontal ligament broblasts. Head & face medicine.
2014; 10:39.
19. Dodani K, Anumala N, Avula H, Reddy K, Varre S,
Kalakonda BB, Arora N, Suri C, Avula JK. Periodontal
ndings in patients with oral submucous brosis and
comet assay of affected gingival epithelial cells. Journal of
periodontology. 2012; 83:1038–1047.
20. Willis DN, Liu B, Ha MA, Jordt SE, Morris JB. Menthol
attenuates respiratory irritation responses to multiple cigarette
smoke irritants. FASEB journal. 2011; 25:4434–4444.
21. Ha MA, Smith GJ, Cichocki JA, Fan L, Liu YS, Caceres
AI, Jordt SE, Morris JB. Menthol attenuates respiratory
irritation and elevates blood cotinine in cigarette smoke
exposed mice. PloS one. 2015; 10:e0117128.
22. Kode A, Yang SR, Rahman I. Differential effects of
cigarette smoke on oxidative stress and proinammatory
cytokine release in primary human airway epithelial cells
and in a variety of transformed alveolar epithelial cells.
Respiratory research. 2006; 7:132.
23. Tancharoen S, Tengrungsun T, Suddhasthira T, Kikuchi K,
Vechvongvan N, Tokuda M, Maruyama I. Overexpression
of receptor for advanced glycation end products and
high-mobility group box 1 in human dental pulp inammation.
Mediators of inammation. 2014; 2014:754069.
24. Lalla E, Lamster IB, Schmidt AM. Enhanced interaction of
advanced glycation end products with their cellular receptor
RAGE: implications for the pathogenesis of accelerated
periodontal disease in diabetes. Annals of periodontology /
the American Academy of Periodontology. 1998; 3:13–19.
25. Lalla E, Lamster IB, Feit M, Huang L, Spessot A, Qu W,
Kislinger T, Lu Y, Stern DM, Schmidt AM. Blockade of
RAGE suppresses periodontitis-associated bone loss in
diabetic mice. The Journal of clinical investigation. 2000;
105:1117–1124.
26. Ito Y, Bhawal UK, Sasahira T, Toyama T, Sato T, Matsuda
D, Nishikiori H, Kobayashi M, Sugiyama M, Hamada N,
Arakawa H, Kuniyasu H. Involvement of HMGB1 and
RAGE in IL-1beta-induced gingival inammation. Archives
of oral biology. 2012; 57:73–80.
27. Xie J, Mendez JD, Mendez-Valenzuela V, Aguilar-
Hernandez MM. Cellular signalling of the receptor for
advanced glycation end products (RAGE). Cellular
signalling. 2013; 25:2185–2197.
28. Liu J, Huang K, Cai GY, Chen XM, Yang JR, Lin LR,
Yang J, Huo BG, Zhan J, He YN. Receptor for advanced
glycation end-products promotes premature senescence
of proximal tubular epithelial cells via activation of
endoplasmic reticulum stress-dependent p21 signaling.
Cellular signalling. 2014; 26:110–121.
29. Fang M, Wang J, Li S, Guo Y. Advanced glycation end-
products accelerate the cardiac aging process through the
receptor for advanced glycation end-products/transforming
growth factor-beta-Smad signaling pathway in cardiac
broblasts. Geriatrics & gerontology international. 2015.
30. Kotsakis GA, Javed F, Hinrichs JE, Karoussis IK,
Romanos GE. Impact of cigarette smoking on clinical
outcomes of periodontal ap surgical procedures: a systematic
review and meta-analysis. Journal of periodontology. 2015;
86:254–263.
31. Javed F, Al-Rasheed A, Almas K, Romanos GE, Al-Hezaimi
K. Effect of cigarette smoking on the clinical outcomes of
periodontal surgical procedures. The American journal of
the medical sciences. 2012; 343:78–84.
32. Katz J, Caudle RM, Bhattacharyya I, Stewart CM, Cohen
DM. Receptor for advanced glycation end product (RAGE)
upregulation in human gingival broblasts incubated with
nornicotine. Journal of periodontology. 2005; 76:1171–1174.
33. Balaji SM. Tobacco smoking and surgical healing of oral
tissues: a review. Indian journal of dental research : ofcial
publication of Indian Society for Dental Research. 2008;
19:344–348.
34. Lerner CA, Sundar IK, Yao H, Gerloff J, Ossip DJ,
McIntosh S, Robinson R, Rahman I. Vapors produced by
electronic cigarettes and e-juices with avorings induce
toxicity, oxidative stress, and inammatory response in
lung epithelial cells and in mouse lung. PloS one. 2015;
10:e0116732.
... Electronic cigarette products deliver stimulant nicotine to the users in aerosol state contributing to the chemical part of the addiction and at the same time they offer sensory and motor stimuli resembling smoking, but without the occurrence of the tobacco-burning process (Isik Andrikopoulos et al. 2019). Although e-cigs were developed and marketed as a healthier alternative to smoking tobacco products, there is a growing body of evidence proving that their aerosols contain In studies comparing e-cigs aerosol to tobacco smoke, lower levels (9-to 450-fold lower) of potentially toxic compounds (e.g., formaldehyde, acetaldehyde, acrolein, and toluene) but considerable levels of potential carcinogens including toxic metals (aluminum, cadmium, chromium, copper, lead, magnesium, manganese, nickel, and zinc), a number of organic compounds including carbonyls (e.g., acrolein from glycerol/glycerine), and potentially harmful compounds such as silicate beads, tin, and flavorings as well as propylene oxide (from propylene glycol) that are not present in traditional tobacco cigarettes are found in these aerosols (Williams et al. 2013;Murakami et al. 2013;D'Ruiz et al. 2015;Sundar et al. 2016). ...
... Cyclooxygenase-2 (COX-2) plays a crucial role in periodontitis by mediating inflammatory reactions in periodontal tissues, a process closely related to the production of prostaglandin E2 (PGE2) (Schaefer et al. 2010;Yucel-Lindberg and Båge 2013). PGE2, derived from COX-2, is linked to periodontal tissue damage as it is a potent stimulator of bone resorption (Kim et al. 2012;Sundar et al. 2016). It has been demonstrated that treating periodontitis in rats with COX-2 inhibitors results in significantly reduced alveolar bone loss and decreased PGE2 expression compared to the untreated periodontitis group (Moro et al. 2019). ...
... It can be seen that the high concentration of nicotine in saliva plays an important role in periodontitis. Enhance the inflammatory response significantly Kim et al. (2012); Sundar et al. (2016) Oxidative stress induced biomolecular damage in saliva ...
Article
Full-text available
Tobacco smoking involves the use of devices such as pipes, cigars, or cigarettes to inhale and exhale smoke from burning tobacco leaves, primarily to ingest nicotine and other substances. The impact of oxidative stress from smoking on periodontitis and its underlying mechanisms remains poorly understood. Previous research has shown that smoking activates oxidative stress responses, generating harmful oxidative substances and free radicals that induce periodontitis. Although traditionally recognized as a key pathway, recent studies suggest additional mechanisms are involved. The study aims to conduct a comprehensive review of the literature on periodontitis induced by oxidative stress from smoking, exploring potential mechanisms involving microorganisms, inflammation, immunity, cellular responses, and saliva. The primary objective is to compare and elucidate the various mechanisms by which traditional tobacco smoke and electronic cigarettes induce oxidative stress and lead to periodontitis and summarize the similarities or differences between the two. In addition, this article explores the different effects of smoking on oxidative stress and periodontitis under different conditions of nicotine presence and nicotine content. This comprehensive review contributes to our evolving understanding of how traditional tobacco smoke and electronic cigarettes affect periodontitis through different pathways and components, emphasizing that oxidative stress is an important factor in smoking-induced periodontitis. The insights gained from this study may help develop targeted interventions for the different pathways of the impact of traditional tobacco smoke and electronic cigarettes in clinical practice, to prevent or treat smoking-induced periodontitis and ultimately safeguard public oral health.
... Although EoE can compromise mucosal integrity, the abrupt onset of symptoms following vaping, combined with negative infectious workup and the severity of endoscopic findings, supports a toxic/chemical etiology rather than an EoE flare. Reviews and similar cases have suggested that heated chemical aerosols in ecigarette vapor can directly injure the esophageal epithelium [2,3,7,8]. ...
... However, it is hypothesized that the inhalation of heated chemicals and flavoring agents in e-cigarette aerosols may lead to direct mucosal irritation and inflammation. Additionally, nicotine exposure has been shown to alter esophageal pH and motility, potentially exacerbating mucosal damage [2,[8][9][10]. ...
... Reactive Oxygen Species (ROS) E-cigarette vapor led to a significant increase in ROS after 24 hours it reaches a peak and decreases after 48 hours, especially with nicotine-containing vapor (25). ROS production was linked to eCV's harmful chemical emissions, such as formaldehyde, heavy metals, diacetyl, carbonyls, and flavoring chemicals (26,27). ...
... Nicotine and flavor additives in e-liquids exacerbated these effects, although some research showed that there was no significant impact (2,17,22). The adverse cellular responses to e-cigarette aerosols may include proliferation and transition to cancer in some tissue types (25,26). E-cigarette vapor was also found to upregulate genes like TP53, linked to cancer risk, especially in cells exposed to higher puff volumes of eCV (15,17,22). ...
... The chemical composition of e-liquids and their aerosols varies widely, with many formulations containing potentially harmful substances such as propylene glycol, glycerin, and flavoring agents. While these compounds are generally considered less toxic than those found in combusted tobacco, some studies have shown that they can exert proinflammatory and oxidative effects, particularly with prolonged exposure [16]. Furthermore, nicotine delivered via vaping may still induce subtle but clinically relevant In contrast, vaporized liquid users demonstrated the least severe capillary alterations, suggesting a relatively lower impact on oral microcirculation. ...
... The chemical composition of e-liquids and their aerosols varies widely, with many formulations containing potentially harmful substances such as propylene glycol, glycerin, and flavoring agents. While these compounds are generally considered less toxic than those found in combusted tobacco, some studies have shown that they can exert pro-inflammatory and oxidative effects, particularly with prolonged exposure [16]. Furthermore, nicotine delivered via vaping may still induce subtle but clinically relevant changes in the capillary network over time. ...
Article
Full-text available
The habit of smoking in its various forms represents a significant public health concern due to its wide range of pathological effects, included the oral cavity. In recent years, alternatives to traditional cigarettes, such as heated tobacco products and electronic cigarettes, have gained popularity and are often marketed as potentially less harmful options. This study seeks to evaluate and compare the morphometric characteristics of oral mucosal capillaries in individuals who consume combusted tobacco, heated tobacco, vaporized liquid, and non-smokers. Using videocapillaroscopy, we assessed both parametric and non-parametric data from 60 patients, divided into four groups according to their smoking habits. The analysis revealed significant differences in capillary morphology among the groups. Users of combusted tobacco exhibited pronounced reductions in capillary diameter, alongside increased tortuosity and the presence of microaneurysms. These alterations are indicative of chronic inflammation and vasoconstriction, likely driven by exposure to nicotine and the high temperatures associated with combustion. Conversely, users of heated tobacco and vaporized liquid exhibited comparatively fewer vascular abnormalities, although angiogenic effects attributable to nicotine were still observable. These findings suggest that alternative tobacco products may have a comparatively lesser impact on the oral microcirculation when compared to traditional smoking. However, the potential long-term effects of these products remain unclear. Further longitudinal research is required to fully understand the risks associated with prolonged use of heated tobacco and electronic cigarettes.
... Reactive Oxygen Species (ROS) E-cigarette vapor led to a significant increase in ROS after 24 hours it reaches a peak and decreases after 48 hours, especially with nicotine-containing vapor (25). ROS production was linked to eCV's harmful chemical emissions, such as formaldehyde, heavy metals, diacetyl, carbonyls, and flavoring chemicals (26,27). ...
... Nicotine and flavor additives in e-liquids exacerbated these effects, although some research showed that there was no significant impact (2,17,22). The adverse cellular responses to e-cigarette aerosols may include proliferation and transition to cancer in some tissue types (25,26). E-cigarette vapor was also found to upregulate genes like TP53, linked to cancer risk, especially in cells exposed to higher puff volumes of eCV (15,17,22). ...
Article
Full-text available
Background Electronic cigarettes (e-cigarettes) are considered relatively safe, thus tobacco and nicotine delivery products have become popular in the last few years. However, the safety of long-term use of these products on oral health is still questionable. E-cigarettes may have potential risks to oral health that can be demonstrated as cellular damage, genetic instability, and mucosal lesions. This review aims to observe the role of e-cigarettes as a risk factor for oral cancer development. Material and Methods This systematic review was conducted following the PRISMA guidelines to provide reliable data on the role of electronic cigarettes as a risk factor for oral cancer development. The research was performed on Pubmed and Scopus by three reviewers from the Oral Pathology Department (Sapienza University of Rome) in May 2024. The search terms included: “e-cigarette”, “oral cancer”, and “risk factor” where 32 articles from PubMed and 75 from Scopus were collected. A total of 12 studies met the eligibility criteria: 6 clinical studies and 6 in vitro. All the included studies were subjected to quality assessment and data extraction processes. The risk of bias assessment of in vitro studies revealed low or unknown risk. None of the studies had any industrial sponsoring and almost all the papers (90%) had the same methods, 64% measured the cell vitality. The qualitative analysis was done for all the included clinical studies using the RoB assessment tool (MINORS). The range of the total RoB score in the comparative studies was between 12 and 23. Results The clinical studies involved a total of 413 participants were also included. Most studies did not specify the age and gender of participants and patients were divided into three main groups based on specific criteria: non-smokers, smokers, and e-cigarette users. These studies highlighted cases of mucosal lesions and genetic instability associated with e-cigarette use. Conclusions However, the limited long-term data and conflicting results emphasize the need for a larger number of studies, such as randomized controlled trials and cohort studies, to acquire more data about the safety and risks associated with e-cigarettes. Key words:E-cigarettes, squamous cell carcinoma, oral oncology, smoking.
... Another limitation is that this study utilized only one rendition of E-liquid (i.e., 50% propylene glycol and 50% glycerol, containing 20 mg/ml of nicotine and no flavors). It is entirely possible that other variations of E-liquids, particularly those containing additional flavorings, could induce more severe outcomes in C. elegans, as shown by others in several human cell lines (Bahl et al., 2012;Leigh et al., 2016;Sundar et al., 2016). It has been shown that when exposing the nematodes directly to E-liquid, nicotine, regardless of solvent, played a role in body size and reproduction (Panitz et al., 2015). ...
Article
Aim: With the invention of electronic cigarettes (ECIG), many questions have been raised regarding their safety as an alternative to smoking conventional cigarettes. Conventional cigarette smoke contains a variety of toxicants including heavy metals. However, ECIG-generated aerosol contains only trace amounts of metals, adding to the argument for it being a safer alternative. In response to heavy metal exposure, metallothioneins are induced in cells to help store the metal, detoxify the body, and are also known responders to oxidative stress. In an attempt to add to the evaluation of the safety of ECIGs, metallothionein expression was quantified using the nematode Caenorhabditis elegans as an assessment of stress induced cellular damage caused by exposure. Methods: Adult nematodes were exposed to either ECIG aerosol or conventional cigarette smoke at doses of 15, 30, and 45 puffs, the equivalent of one, two, and three cigarettes, respectively. Movement, survival, and stress-induced sleep were assessed for up to 24 h after exposure. Relative expression levels for mtl-1 and mtl-2, C. elegans metallothionein genes, were analyzed after 1, 5, and 24 h post exposure using quantitative RT-PCR. Results: Nematodes exposed to conventional cigarette smoke underwent stress-induced sleep in a dose dependent manner with animals recovering to values within the range of air control after 5 h post exposure. Those exposed to ECIG aerosol did not undergo stress-induced sleep and were indistinguishable from controls. The expression of mtl-1 increased in a dose and time dependent manner in C. elegans exposed to conventional cigarette smoke, with a maximum expression observed at 5 h post exposure of 45 puffs. No induction of mtl-2 was observed in any animals. Additionally, ECIG aerosol did not induce expression of mtl-1 and mtl-2 at levels different than those of untreated. Frontiers in Physiology | www.frontiersin.org 1 April 2018 | Volume 9 | Article 426 Cobb et al. Smoke Induces C. elegans Metallothionein Conclusion: ECIG aerosol failed to induce a stress response in C. elegans. In contrast, conventional cigarette smoke induced the production of mtl-1 in a manner that correlates with the induction of stress-induced sleep suggesting a stress response to damage. The lack of cellular stress response to ECIG aerosol suggests it may be a safer alternative to conventional cigarettes.
... E-liquids have demonstrated pro-inflammatory effects in human monocytes, and display toxic effects on human stem cells as well as terminally differentiated human cells (Bahl et al., 2012;Muthumalage et al., 2018;Pushalkar et al., 2020). Among the pulmonary tissue studies, research supports that flavoring agents found in cinnamon, strawberry, blueberry, menthol and tobacco, and not the base humectants (i.e., propylene glycol and/or vegetable glycerin) are responsible for cytokine production and adverse effects such as cell death (Leigh et al., 2016(Leigh et al., , 2018Sundar et al., 2016). Currently, ECIG studies primarily focus on airway tissues. ...
Article
Introduction: Electronic cigarette (ECIG) use or vaping has become popular globally. While the question "Is vaping safer than smoking?" continues, it is becoming clearer that one of the most dangerous components of E-liquids are the flavorings. Since the oral cavity is the first anatomical site to be assaulted by ECIG aerosol, the aim of this study is to test the hypothesis that flavored ECIG aerosols or E-liquids pose a more detrimental effect on the growth of commensal oral streptococcal bacteria compared to flavorless aerosols or E-liquids. Methods: Kirby Bauer assays and 24-h planktonic growth curves were used to compare the effects of flavorless vs. flavored (tobacco, menthol, cinnamon, strawberry and blueberry) ECIG-generated aerosols and E-liquids on the growth of four common strains of oral commensal bacteria (Streptococcus gordonii, Streptococcus intermedius, Streptococcus mitis and Streptococcus oralis). Results: Kirby Bauer assays revealed inhibition of growth for all bacteria tested when exposed to 100% menthol, cinnamon or strawberry flavors. In contrast, 5% flavor in E-liquid had no effect. When exposed to 100 puffs of ECIG-generated aerosol ± flavors (≈ 0.05% flavor in brain heart infusion media) or an equivalent amount of E-liquid ± flavors, twenty-four hour planktonic growth curves indicated no effect on growth for all streptococci tested. Subsequent twenty-four hour planktonic growth curves testing the effects of E-liquid ± flavors (0.0625, 0.125, 0.25, 0.3125, 0.625, and 1.25% flavor in brain heart infusion media) revealed dose-dependent inhibition of growth, particularly for menthol, cinnamon and strawberry), for all bacteria tested. Conclusion: These results support the hypothesis that flavored E-liquids are more detrimental to the growth of oral commensal bacteria than unflavored E-liquids. The streptococci tested in this study are early colonizers and part of the foundation of oral biofilms and dental plaque. Disturbances in the composition and growth of these primary Frontiers in Physiology | www.frontiersin.org 1 November 2020 | Volume 11 | Article 585416 Fischman et al. Flavored E-Liquids and Oral Streptococci colonizers is crucial to the development of a healthy dental plaque and host-bacteria interactions. E-liquids and their aerosols containing flavoring agents alter the growth of these bacteria. Such perturbations of pioneering oral communities pose a potential risk to the health of the oral cavity and, ultimately, health in general.
Article
Full-text available
Os congressos acadêmicos são eventos que promovem uma oportunidade essencial para a formação dos estudantes, pois proporciona um ambiente rico em troca de conhecimentos e experiências, além de exposição às inovações mais recentes da área. Durante o congresso, os alunos podem debater sobre os assuntos que ampliam a compreensão sobre temas complexos e atualizações científicas. Esse contato direto com profissionais e pesquisadores permite que os estudantes desenvolvam um pensamento crítico e reflexivo sobre os avanços médicos e desafios clínicos, estimulando o interesse pela pesquisa e pela prática baseada em evidências. Além disso, os congressos incentivam a criação de redes de contato, facilitando futuras colaborações profissionais e aprimorando o preparo dos futuros médicos para lidar com as demandas e responsabilidades da carreira
Article
Full-text available
Since the oral cavity comes into contact with several xenobiotics (dental materials, oral hygiene formulations, drugs, or tobacco products), it is one major site for toxicity manifestation. Multiple parameters are assessed during toxicity testing (cell viability and proliferation, apoptosis, morphological changes, genotoxicity, oxidative stress, and inflammatory response). Due to the complexity of the oral cavity environment, researchers have made great efforts to design better in vitro models that mimic natural human anatomic and functional features. The present review describes the in vitro methods currently used to investigate the toxic potential of various agents on oral cavity tissues and their evolution from simple 2D cell culture systems to complex organ-a-chip designs.
Thesis
E-Cigarettes were introduced to the United States in the early 2000s and have gained popularity rapidly since. E-Cigarettes aerosolize a liquid, a “refill fluid” that the user then inhales. Although most E-Cigarettes contain nicotine, flavorings in refill fluids are what has drawn youth to use E-Cigarettes. Evidence is growing that flavor chemicals in the refill fluids can cause harm upon inhalation. Seven of the most popular flavor chemicals in refill fluids were chosen to study in this review. Ethyl vanillin, vanillin, cinnamaldehyde, maltol, ethyl maltol, benzyl alcohol, and menthol. Fifty-seven original articles (collected from PubMed, and Google Scholar) on the cytotoxicity of flavor chemicals were collected and critically analyzed. Many of these flavor chemicals are found frequently and are present in high concentrations. The cytotoxic effects of these flavor chemicals reduce mitochondrial reductase activity, decreasing cellular viability, increasing the production of reactive oxygen species, increasing the expression of proinflammatory molecules, disrupt epithelial barriers, and more. More research is needed on the specific effects of these flavor chemicals, as well as the cytotoxic effects of other flavor chemicals that can be found in refill fluids of E-Cigarettes.
Article
Full-text available
Electronic cigarettes (e-cigarettes) are generally acknowledged as a safer alternative to the use of combusted tobacco products. Nevertheless, there are increasing conflicting claims concerning the effect of these novel industrial products on the health of e-cigarettes users. The aim of this work was to investigate the effects of the liquids of e-cigarettes on human gingival fibroblasts (HGFs) and to compare the effects of nicotine-containing fluid to the fluid itself. HGFs were treated with different concentrations (0-5 mg/mL) of fluids of e-cigarettes for different times (0-72 h) and cytotoxicity was analyzed by MTT assay. Fluids were administered also after being vaped (e.g., warmed into the cartomizer). Apoptosis occurrence and Bax expression were evaluated by flow cytometry; ROS production was analyzed by fluorescence optical microscopy. Both nicotine-containing and nicotine-free fluids induced an increased ROS production after 24 h, along with an increased Bax expression, followed by apoptosis occurrence after 48 h of exposure. The cytotoxicity exerted on HGFs by e-cigarettes fluids is not entirely ascribable to nicotine. Since the e-cigarettes are advertised as a safer alternative to traditional ones, especially for the possibility of "smoking" nicotine-free fluids, further studies are necessary to clarify the mechanism involved in the occurrence of cytotoxicity exerted by such compounds. Our results suggest a role for e-cigarette fluids in the pathogenesis of oral diseases, such as periodontitis.
Article
Full-text available
Oxidative stress and inflammatory response are the key events in the pathogenesis of chronic airway diseases. The consumption of electronic cigarettes (e-cigs) with a variety of e-liquids/e-juices is alarmingly increasing without the unrealized potential harmful health effects. We hypothesized that electronic nicotine delivery systems (ENDS)/e-cigs pose health concerns due to oxidative toxicity and inflammatory response in lung cells exposed to their aerosols. The aerosols produced by vaporizing ENDS e-liquids exhibit oxidant reactivity suggesting oxidants or reactive oxygen species (OX/ROS) may be inhaled directly into the lung during a "vaping" session. These OX/ROS are generated through activation of the heating element which is affected by heating element status (new versus used), and occurs during the process of e-liquid vaporization. Unvaporized e-liquids were oxidative in a manner dependent on flavor additives, while flavors containing sweet or fruit flavors were stronger oxidizers than tobacco flavors. In light of OX/ROS generated in ENDS e-liquids and aerosols, the effects of ENDS aerosols on tissues and cells of the lung were measured. Exposure of human airway epithelial cells (H292) in an air-liquid interface to ENDS aerosols from a popular device resulted in increased secretion of inflammatory cytokines, such as IL-6 and IL-8. Furthermore, human lung fibroblasts exhibited stress and morphological change in response to treatment with ENDS/e-liquids. These cells also secrete increased IL-8 in response to a cinnamon flavored e-liquid and are susceptible to loss of cell viability by ENDS e-liquids. Finally, exposure of wild type C57BL/6J mice to aerosols produced from a popular e-cig increase pro-inflammatory cytokines and diminished lung glutathione levels which are critical in maintaining cellular redox balance. Thus, exposure to e-cig aerosols/juices incurs measurable oxidative and inflammatory responses in lung cells and tissues that could lead to unrealized health consequences.
Article
Full-text available
Addition of menthol to cigarettes may be associated with increased initiation of smoking. The potential mechanisms underlying this association are not known. Menthol, likely due to its effects on cold-sensing peripheral sensory neurons, is known to inhibit the sensation of irritation elicited by respiratory irritants. However, it remains unclear whether menthol modulates cigarette smoke irritancy and nicotine absorption during initial exposures to cigarettes, thereby facilitating smoking initiation. Using plethysmography in a C57Bl/6J mouse model, we examined the effects of L-menthol, the menthol isomer added to cigarettes, on the respiratory sensory irritation response to primary smoke irritants (acrolein and cyclohexanone) and smoke of Kentucky reference 2R4 cigarettes. We also studied L-menthol's effect on blood levels of the nicotine metabolite, cotinine, immediately after exposure to cigarette smoke. L-menthol suppressed the irritation response to acrolein with an apparent IC₅₀ of 4 ppm. Suppression was observed even at acrolein levels well above those necessary to produce a maximal response. Cigarette smoke, at exposure levels of 10 mg/m³ or higher, caused an immediate and marked sensory irritation response in mice. This response was significantly suppressed by L-menthol even at smoke concentrations as high as 300 mg/m³. Counterirritation by L-menthol was abolished by treatment with a selective inhibitor of Transient Receptor Potential Melastatin 8 (TRPM8), the neuronal cold/menthol receptor. Inclusion of menthol in the cigarette smoke resulted in roughly a 1.5-fold increase in plasma cotinine levels over those observed in mice exposed to smoke without added menthol. These findings document that, L-menthol, through TRPM8, is a strong suppressor of respiratory irritation responses, even during highly noxious exposures to cigarette smoke or smoke irritants, and increases blood cotinine. Therefore, L-menthol, as a cigarette additive, may promote smoking initiation and nicotine addiction.
Article
Full-text available
Introduction Over the last years, electronic cigarettes (ECs) have become more popular, particularly in individuals who want to give up smoking tobacco. The aim of the present study was to assess the influence of the different e-smoking liquids on the viability and proliferation of human periodontal ligament fibroblasts. Method and materials For this study six test solutions with components from ECs were selected: lime-, hazelnut- and menthol-flavored liquids, nicotine, propylene glycol, and PBS as control group. The fibroblasts were incubated up to 96 h with the different liquids, and cell viability was measured by using the PrestoBlue® reagent, the ATP detection and the migration assay. Fluorescence staining was carried out to visualize cell growth and morphology. Data were statistically analyzed by two-tailed one-way ANOVA. Results The cell viability assay showed that the proliferation rates of the cells incubated with nicotine or the various flavored liquids of the e-cigarettes were reduced in comparison to the controls, though not all reductions were statistically significant. After an incubation of 96 h with the menthol-flavored liquid the fibroblasts were statistically significant reduced (p < 0.001). Similar results were found for the detection of ATP in fibroblasts; the incubation with menthol-flavored liquids (p < 0.001) led to a statistically significant reduction. The cell visualization tests confirmed these findings. Conclusion Within its limits, the present in vitro study demonstrated that menthol additives of e-smoking have a harmful effect on human periodontal ligament fibroblasts. This might indicate that menthol additives should be avoided for e-cigarettes.
Article
Oxidants or nanoparticles have recently been identified as constituents of aerosols released from various styles of electronic cigarettes (E-cigs). Cells in the lung may be directly exposed to these constituents and harbor reactive properties capable of incurring acute cell injury. Our results show mitochondria are sensitive to both E-cig aerosols and aerosol containing copper nanoparticles when exposed to human lung fibroblasts (HFL-1) using an Air-Liquid Interface culture system, evident by elevated levels of mitochondrial ROS (mtROS). Increased mtROS after aerosol exposure is associated with reduced stability of an electron transport chain (ETC) complex IV subunit and nuclear DNA fragmentation. Increased levels of IL-8 and IL-6 in HFL-1 conditioned media were also observed. These findings reveal both mitochondrial, genotoxic, and inflammatory stresses are features of direct cell exposure to E-cig aerosols which are ensued by inflammatory duress, raising a concern on deleterious effect of vaping.
Article
The current study was carried out to evaluate the effect of advanced glycation end-products (AGE) on cardiac aging and to explore its underlying mechanisms. Neonatal rat cardiac fibroblasts were cultured and divided into four groups: control; AGE; AGE + receptor for AGE antibody and AGE + SB431542 (transforming growth factor-β [TGF-β]/Smad signaling pathway inhibitor, 10 μmol/L) group. After being cultured for 48 h, the cells were harvested and the senescence-associated beta-galactosidase expression was analyzed. Then the level of p16, TGF-β, Smad/p-smad and matrix metalloproteinases-2 was evaluated by western blot. Significantly increased senescence-associated beta-galactosidase activity as well as p16 level was observed in the AGE group. Furthermore, AGE also significantly increased the TGF-β1, p-smad2/3 and metalloproteinases-2 expression in cardiac fibroblasts (all P < 0.01). Meanwhile, either pretreatment with receptor for AGE-Ab or SB431542 significantly inhibited the upregulated cardiac senescence (beta-galactosidase activity and P16) and fibrosis-associated (TGF-β1, p-smad2/3 and metalloproteinases-2) markers induced by AGE. Taken together, all these results suggested that AGE are an important factor for cardiac aging and fibrosis, whereas the receptor for AGE and TGF-β/Smad signaling pathway might be involved in the AGE-induced cardiac aging process. Geriatr Gerontol Int 2015; ●●: ●●-●●. © 2015 Japan Geriatrics Society.
Article
To narrow the gap in our understanding of potential oxidative properties associated with Electronic Nicotine Delivery Systems (ENDS) i.e. e-cigarettes, we employed semi-quantitative methods to detect oxidant reactivity in disposable components of ENDS/e-cigarettes (batteries and cartomizers) using a fluorescein indicator. These components exhibit oxidants/reactive oxygen species reactivity similar to used conventional cigarette filters. Oxidants/reactive oxygen species reactivity in e-cigarette aerosols was also similar to oxidant reactivity in cigarette smoke. A cascade particle impactor allowed sieving of a range of particle size distributions between 0.450 and 2.02 μm in aerosols from an e-cigarette. Copper, being among these particles, is 6.1 times higher per puff than reported previously for conventional cigarette smoke. The detection of a potentially cytotoxic metal as well as oxidants from e-cigarette and its components raises concern regarding the safety of e-cigarettes use and the disposal of e-cigarette waste products into the environment.
Article
Objectives: Periodontal flap surgery is often required to achieve adequate reduction in probing depth (PD) in habitual smokers with advanced periodontal disease. Yet, results from clinical studies have reported disadvantageous results for periodontal healing response in smokers as compared to non-smokers. To our knowledge, a meta-analysis of studies assessing the effect of cigarette smoking on periodontal flap procedures has not been previously performed. Thus, the aim of this study was to perform a meta-analysis on controlled, clinical studies comparing periodontal flap surgical procedures in smokers verses non-smokers. Material and Methods: A systematic review of articles relevant to periodontal flap surgical procedures in smokers was performed electronically utilizing the PubMed, MEDLINE (OVID) database, the EMBASE database, the Cochrane Central Register of Controlled Trials (CENTRAL) and the Ovid Medline(R) In-Process database from 1946 up to and including March 2014 utilizing predefined, optimized search strategies. Meta-analyses were conducted separately for each of the two primary outcomes: Changes in clinical attachment levels (CAL) and PD. Results: The initial search yielded 390 titles and abstracts. Following initial screening, 15 out of 390 publications were scrutinized during the second phase of the review. In the second phase, 7 articles were excluded from the analysis and 8 controlled, clinical studies were finally selected. Qualitative assessment of the articles consistently showed an increased treatment effect in non-smokers verses smokers. Meta-analysis on 8 studies reporting on 363 study participants showed an increased reduction in mean [95% Confidence Interval] PD of 0.39mm [0.45, 0.33]. Similar results were found for gain in CAL. Conclusions: Considering the relatively homogeneous available information we concluded that active smokers could be candidates for periodontal flap surgical procedures, but they should be thoroughly informed pre-operatively for the significantly reduced clinical outcomes as compared to non-smokers.