ArticlePDF Available

Repurposing INCI-registered compounds as skin prebiotics for probiotic Staphylococcus epidermidis against UV-B

Springer Nature
Scientific Reports
Authors:

Abstract and Figures

Repurposing existing compounds for new indications may facilitate the discovery of skin prebiotics which have not been well defined. Four compounds that have been registered by the International Nomenclature of Cosmetic Ingredients (INCI) were included to study their abilities to induce the fermentation of Staphylococcusepidermidis (S. epidermidis), a bacterial species abundant in the human skin. Liquid coco-caprylate/caprate (LCC), originally used as an emollient, effectively initiated the fermentation of S. epidermidis ATCC 12228, produced short-chain fatty acids (SCFAs), and provoked robust electricity. Application of LCC plus electrogenic S. epidermidis ATCC 12228 on mouse skin significantly reduced ultraviolet B (UV-B)-induced injuries which were evaluated by the formation of 4-hydroxynonenal (4-HNE), cyclobutane pyrimidine dimers (CPD), and skin lesions. A S. epidermidis S2 isolate with low expressions of genes encoding pyruvate dehydrogenase (pdh), and phosphate acetyltransferase (pta) was found to be poorly electrogenic. The protective action of electrogenic S. epidermidis against UV-B-induced skin injuries was considerably suppressed when mouse skin was applied with LCC in combination with a poorly electrogenic S. epidermidis S2 isolate. Exploring new indication of LCC for promoting S. epidermidis against UV-B provided an example of repurposing INCI-registered compounds as skin prebiotics.
Content may be subject to copyright.

Scientic Reports | (2020) 10:21585 | 
www.nature.com/scientificreports
Repurposing INCI‑registered
compounds as skin prebiotics
for probiotic Staphylococcus
epidermidis against UV‑B
Arun Balasubramaniam1, Prakoso Adi1, Do Thi Tra My1, Sunita Keshari2, Raman Sankar3,
Chien‑Lung Chen4 & Chun‑Ming Huang1*
Repurposing existing compounds for new indications may facilitate the discovery of skin prebiotics
which have not been well dened. Four compounds that have been registered by the International
Nomenclature of Cosmetic Ingredients (INCI) were included to study their abilities to induce the
fermentation of Staphylococcus epidermidis (S. epidermidis), a bacterial species abundant in the
human skin. Liquid coco‑caprylate/caprate (LCC), originally used as an emollient, eectively initiated
the fermentation of S. epidermidis ATCC 12228, produced short‑chain fatty acids (SCFAs), and
provoked robust electricity. Application of LCC plus electrogenic S. epidermidis ATCC 12228 on mouse
skin signicantly reduced ultraviolet B (UV‑B)‑induced injuries which were evaluated by the formation
of 4‑hydroxynonenal (4‑HNE), cyclobutane pyrimidine dimers (CPD), and skin lesions. A S. epidermidis
S2 isolate with low expressions of genes encoding pyruvate dehydrogenase (pdh), and phosphate
acetyltransferase (pta) was found to be poorly electrogenic. The protective action of electrogenic S.
epidermidis against UV‑B‑induced skin injuries was considerably suppressed when mouse skin was
applied with LCC in combination with a poorly electrogenic S. epidermidis S2 isolate. Exploring new
indication of LCC for promoting S. epidermidis against UV‑B provided an example of repurposing INCI‑
registered compounds as skin prebiotics.
Drug repurposing or repositioning is an eective approach to rapidly identify novel indications from knowncom-
pounds1,2. ere have been numerous successful cases of repurposed drugs, including sildenal citrate (Viagra)
as a medicine for erectile dysfunction and pulmonary arterial hypertension and raloxifene hydrochloride (Evista)
as a treatment for osteoporosis in postmenopausal women3. Repurposing drugs, including Remdesivir4 and
Dexamethasone5, to discover potential forms of treatment for severe acute respiratory syndrome coronavirus 2
(SARS-CoV-2) is actively ongoing. Skin diseasesaect half of the worlds population and novel drugs for treat-
ments of skin diseases are in high demand. eskin isthe largest organ in humans and continuously protects
the humans from the harmful environment6,7. Over 100 distinct species, which contribute to making up a total
of 1 million microbes in the skin microbiome, conquer every square centimeter of human skin6,8. Our previous
studies have demonstrated that fermentation of skin probiotic bacteria generated benecial metabolites, such
as short-chain fatty acids (SCFAs), that can attenuate skin disorders9. For example, Staphylococcus epidermidis
(S. epidermidis), a common member in the human skin microbiome, can fermentatively metabolize carbon-rich
molecules as prebiotics to yield SCFAs against pathogenic Staphylococcus aureus (S. aureus)10.
Prebiotics were originally dened by Gibson and Roberfroid in 1995 as nondigestible food ingredients11. is
denition was later revised in12 and13 as “Glucose-based dietary bers and non-carbohydrate substances including
polyunsaturated fatty acid (PUFA) have been used as prebiotics for gut bacteria”. Prebiotics for bacteria in the
skin and other human organs are not yet dened. Prebiotics can provide probiotic bacteria as carbon sources
to initiate the fermentation and produce SCFAs as acetate and butyrate14. It has been reported that oxidation of
acetate or butyrate served as an electron donor to discharge electron to electron acceptors15. e gene-encoding
proteins in the extracellular electron transfer (EET) family of homologs are believed to be present in both Gram-
negative and Gram-positive bacteria16. Unlike Gram-negative bacteria, Gram-positive bacteria, not having an
OPEN
Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan. Department
of Life Sciences, National Central University, Taoyuan, Taiwan. Institute of Physics, Academia Sinica, Nankang,
Taipei, Taiwan. Division of Nephrology, Landseed International Hospital, Taoyuan, Taiwan. *email: chunming@
ncu.edu.tw
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol:.(1234567890)
Scientic Reports | (2020) 10:21585 | 
www.nature.com/scientificreports/
outer membrane, carry a cell envelope with a textured peptidoglycan layer and teichoic acids that are thought
to be poorly electrogenic17. However, a avin-based EET process has been recently identied in Gram-positive
bacteria to produce electricity16 by activation of type II NADH hydrogenase, which can catalyze electron exchange
from cytosolic NADH to a quinone derivative such as quinone demethylmenaquinone (DMK)16. Our recent
studies demonstrated that S. epidermidis can mediate glycerol as a source to generate electricity and enhanced
bacterial resistance to UV-B18.
In this study, we selected four carbon-rich molecules that have been listed on the International Nomencla-
ture of Cosmetic Ingredients (INCI) to examine their prebiotic activities. e liquid coco-caprylate/caprate
(LCC) with C8-10 fatty acid connected to C12-C18 fatty alcohols is currently used as an emollient on ultraviolet
(UV) lter absorbance19. Isononyl isononanoate (ININ) (C18H36O2) is an ingredient in cosmetics and personal
care products as an emollient, texture enhancer, and plasticizer20. Polyethylene glycol (PEG)-150 distearate
(PDS) [(C2H4O)n.C36H70O3] is a thickening agent for shampoo products21. PEG-150 pentaerythrityl tetrastearate
(PETIS) (C77H148O8) has been used for increasing viscosity of an aqueous agent in cosmetics22. To screen these
four carbon-rich molecules for repurposing, we examined their prebiotic activities for induction of fermenta-
tion of S. epidermidis. It has been documented that S. epidermidis can mediate fermentation to down-regulate
UV-B-induced inammation in mouse skin9. We thus further explored the mechanism by which repurposing
carbon-rich molecules as skin prebiotics inuence the skin damage induced by UV-B. UV radiation which
provokes free radical formation, making it a primary risk factor for skincancer23. It has been reported that UV,
in particular UV-B, radiation can up-regulate the local neuroendocrine axes, induce the release of hormones to
circulation, activate the central hypothalamic–pituitary–adrenal axis, and reset body hemostasis against skin
disorders including cancers, aging and autoimmune diseases24. Repurposing may facilitate the discovery of new
mechanisms of action for INCI-registered ingredients as skin prebiotics against UV injuries.
Methods
Ethics statement. All animal protocols and experiments have been approved by the National Central
University (NCU), Taiwan. Experiments were conducted in accordance with the protocols (NCU-106-016, 19
December 2017) of the Institutional Animal Care and Use Committee (IACUC) of National Central University
(NCU). Female ICR mice (8–9weeks old) were purchased from the National Laboratory Animal Center Tai-
pei, Taiwan. CO2 sedation was used to sacrice mice in an encased chamber. All human study protocols were
approved by Institutional Review Board (IRB) (No. 19-013-B1, 22 May 2019) and Ethics Committee of Land-
seed International Hospital, Taiwan. e methods followed for skin swab sampling procedure were carried out
in accordance with relevant guidelines and regulations of IRB which was approved by Landseed International
Hospital, Taiwan. Skin swabs were collected from three healthy subjects and informed consent was obtained
from all study participants.
Bacterial fermentation. Staphylococcus epidermidis ATCC 12228 in tryptic soy broth (TSB) (Sigma, St.
Louis, MO, USA) was cultured overnight at 37°C. Bacterial growth was determined at 600nm wavelength
(OD600). e bacterial pellet was collected aer centrifugation at 5,000 × g for 10min, resuspended with 1 × PBS
and diluted to 107CFU/ml before further incubation in rich media [1.5g/l KH2PO4, 10g/l yeast extract (Biokar
Diagnostics, Beauvais, France), 2.5g/l K2HPO4, 3g/l TSB, and 0.002% (w/v) phenol red (Merck, Darmstadt,
Germany)] at 37°C. For fermentation, bacteria in rich media in the presence of 2% LCC, ININ, PDS or PETIS
(TNJC corporation, Chiayi, Taiwan) were incubated for 12h. e color change of phenol red from red to yellow
indicated bacterial fermentation, which was quantied by measurement of OD562. Bacteria alone or rich media
with or without LCC, ININ, PDS or PETIS served as controls. To examine the eect of LCC on the bacterial
growth, S. epidermidis ATCC 12228 [107 colony-forming unit (CFU)/ml)] was incubated with 2% LCC or phos-
phate buer saline (PBS) for 12h at 37°C. Aer incubation, bacteria were serially diluted 1: 100–1:105 in a 96 well
plate. 10μl of serially diluted bacteria were dropped on a TSB agar plate for CFU counts.
Electricity detection. Electricity produced by S. epidermidis was detected in vitro using a chamber
equipped with cathode and anode. A carbon felt (2.5cm × 10cm) and a carbon cloth (10cm × 10cm) (Homy
Tech, Taoyuan, Taiwan) were utilized to fabricate anode and cathode., respectively. e cathode was wrapped up
to a Naon membrane N117 (6cm × 6cm) (Homy Tech), which served as a proton exchange membrane (PEM).
Copper wires were used to connect anode and cathode with external resistance (1,000 Ω)18. S. epidermidis in
the presence or absence of LCC, ININ, PDS or PETIS was pipetted on the surface of the anode. Electricity was
recorded by the changes in voltage (mV) against time (min) using a digital multimeter (Lutron, DM-9962SD,
Sydney, Australia). e recorded voltages in every 10s were used for plotting a graph.
Cyclic voltammetry. e three-electrode autolab potentiostat (PGSTAT 128N, Metrohm Autolab, Utrecht,
Netherland) was used for conducting cyclic voltammetry. e screen-printed carbon electrode (SPCE) (SE-100,
Zensor R&D, Taichung, Taiwan) served as a working electrode with a working area of 5mm. e Ag/AgCl elec-
trode and platinum electrode acted as the reference against applied potential and counter electrode, respectively.
All electrodes were purchased from Metrohm Autolab. S. epidermidis (107CFU/μl) in the presence or absence of
2% of LCC, ININ, PDS or PETIS was drop-coated on the surface of a working electrode. e potential windows
were inspected between −0.8 and 0.2V at 0.005V/s. PBS at 7.4 pH was used as an electrolyte. e potentiostat
was operated using Autolab Nova 2.0 soware (https ://metro hm-autol ab.com/Produ cts/Echem /Sow are/Nova.
html/; Metrohm Autolab, Utrecht, Netherland).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol.:(0123456789)
Scientic Reports | (2020) 10:21585 | 
www.nature.com/scientificreports/
Extraction of bacterial RNA. RNA was extracted from overnight cultured S. epidermidis ATCC 12228
or a S2 isolate (107CFU/ml). e cultured bacteria were centrifuged at 5,000 × g for 10min and the pellet was
collected. RNA in bacterial pellet was extracted using a total RNA mini purication kit (Biokit, Miaoli, Taiwan)
and quantied by UV spectrophotometry in a Synergy HT multi-mode microplate reader (BioTek Instruments
Inc., Highland Park, Winooski, Vermont, USA).
Real‑time qPCR (RT‑qPCR). RT q-PCR was used to analyze the expression of genes encoding pyruvate
dehydrogenase (pdh), phosphate acetyltransferase (pta) and intracellular adhesion A (ica A) in S. epidermidis
ATCC 12228 and S2 isolate. RNA (1ng) was converted to cDNA using an iScript cDNA Synthesis Kit (Bio-Rad,
Hercules, CA, USA). e cDNA was served as a template in StepOnePlus RT PCR System (ermo Fisher Scien-
tic, Waltham, MA, USA), which was executed using Power SYBR Green and PCR Master Mix (ermo Fisher
Scientic). e primer-Blast tool (https ://blast .ncbi.nlm.nih.gov/Blast .cgi/; Rockville Pike, Bethesda MD, USA)
from the National Center for Biotechnology Information (NCBI) was used for designing all primers. Total one
step RT-PCR reaction condition was xed for 40 cycles as follows: 95°C for 10min followed by 95°C for 15s,
60°C for 60s, and 72°C for 30s. Gene expression was normalized with the 16S rRNA gene. e cycle threshold
(2−ΔΔCt) was implemented to analyze the relative expression of genes. e designed primers for all genes were
shown in TableS2.
Gas chromatography‑mass spectrometry (GC–MS) analysis. Staphylococcus epidermidis ATCC
12228 (107CFU/ml) with 2% LCC in rich media was cultured for 24h and then centrifuged at 5,000 × g for
10min. Supernatants of bacterial culture were collected and ltered through 0.22µm lters. e GC–MS proto-
col for SCFAs detection were obtained following the method25.
UV‑B exposure. Mouse hair in the dorsal skin was removed using Nair cream (Church and Dwight, Ewing
Township, NJ, USA) one day before experiments started. e dorsal skin of each mouse was exposed to 100mJ/
cm2 UV-B irradiation using a UV lamp (Model EB-280C, Spectronics Corp., Westbury, NY, USA) twice a week,
followed by subsequent application of 107CFU/ml S. epidermidis with or without 2% LCC three times per week
for two weeks. e images of mouse skin were captured on day 0, 7 and 14. Skin lysates and sections from dorsal
skin (1cm2) was prepared. Skin sections were stained with hematoxylin and eosin (H&E) and visualized using
the Olympus BX63 microscope (Olympus, Tokyo, Japan).
Western blotting. Tissue Protein Extraction Reagent (T-PER) (ermo Fisher Scientic) was used for pre-
paring the skin lysates. Protein concentrations of skin lysates were measured by a bicinchoninic acid (BCA)
assay (Bio-Rad). Skin lysates (30μg) were loaded to a 10% sodium dodecyl sulphate–polyacrylamide gel elec-
trophoresis (SDS-PAGE) gel and transferred to a PVDF membrane (Millipore Sigma, Burlington, MA, USA).
e membrane was blocked with 5% (w/v) non-fat milk, and incubated with primary antibodies to cyclobutane
pyrimidine dimer (CPD) (1:1,000; Cosmo Bio, Tokyo, Japan), 4-hydroxynonenal (4-HNE) (1:2,000; Abcam,
Cambridge, MA, USA), or β-actin (1:5,000; ACE Biolabs, Taoyuan, Taiwan) overnight at 4°C. e membrane
was subsequently incubated with secondary antibodies goat anti-rabbit or anti-mouse IgG (H + L) horseradish
peroxidase (HRP) (1:5000; ACE Biolabs) for 1h. Protein bands in membranes were developed using chemilu-
minescent detection reagent (ermo Fisher Scientic) and visualized by an Omega Lum C Imaging System
(Gel Co., San Francisco, CA, USA). ImageJ soware (https ://image j.nih.gov/ij/index .html/; National Institutes
of Health, Bethesda, MD, USA) was employed to quantify the intensities of protein bands.
Statistical analysis. GraphPad Prism 8 (https ://www.graph pad.com/; GraphPad Soware, La Jolla, CA,
USA) soware was employed for data analysis by unpaired t-test. e signicant dierence was considered by
P-values observation as follows: P-values of < 0.05 (*), < 0.01 (**), and < 0.001 (***). e mean ± standard devia-
tion (SD) was obtained from at least three separate experiments.
Results
INCI‑registered compounds function as skin probiotics for induction of SCFA production and
bacterial electricity. Our previous studies have demonstrated the fermentation and electrogenic activities
of S. epidermidis in the presence of glycerol as a carbon source18. To examine if INCI-registered compounds can
act as skin prebiotics that provides carbon sources to induce fermentation of skin bacteria, we cultured S. epider-
midis ATCC 12228 (107CFU/ml), a non-biolm forming skin bacterium, in rich media containing phenol red
with 2% each individual INCI-registered compound including LCC, ININ, PDS, and PETIS for 12h. Media with
bacteria alone served as controls. A change in color of phenol red from red to yellow and a signicant reduction
of the optical density of 562nm (OD562) due to low pH values26 in the culture media of the S. epidermidis in
the presence of 2% each individual INCI-registered compound served as indications of bacterial fermentation.
As shown in Fig.1A, 2% LCC and ININ, compared to PDS and PETIS, induced signicant fermentation of S.
epidermidis by detection of yellowish media and OD562 reduction. e reduction of OD562 induced by LCC was
greater than that by ININ. To validate the occurrence of fermentation, six SCFAs including acetate, butyrate,
hexanoate, isobutyrate, isovalerate, and propionate in the media of LCC fermentation were detected by GC–MS
analysis (Fig.1B). A high amount of acetate (> 15mM) was produced by LCC fermentation of S. epidermidis.
SCFAs, especially acetate and butyrate, have been proved as potent electron donors during the EET process in
bacteria2729. We thus investigated the electrogenic activity of S. epidermidis in the presence of INCI-registered
compounds. Changes in voltages in an invitro chamber and current values detected by cyclic voltammetry30
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol:.(1234567890)
Scientic Reports | (2020) 10:21585 | 
www.nature.com/scientificreports/
were used to monitor bacterial electricity. Compared to media with S. epidermidis alone, media with S. epider-
midis plus ININ, PDS or PETIS did not elicit high voltage changes and currents. By contrast, a robust increase
in voltage changes with a peak voltage of approximately 6mV and currents was detected in the media with S.
epidermidis plus LCC (Fig.2). e result provided evidence for repurposing INCI-registered LCC as a prebiotic
to provoke the fermentation and electricity production of skin S. epidermidis bacteria.
Figure1. LCC fermentation of S. epidermidis. (A) S. epidermidis ATCC 12228 (107CFU/ml; B) was incubated
for 12h in rich media in the presence or absence of 2% LCC, ININ, PDS or PETIS. e prevalence of
fermentation was indicated by colour change of phenol red from red to yellow and quantied by OD562. (B) S.
epidermidis ATCC 12228 (107CFU/ml) in the presence of 2% LCC in rich media was cultured for 24h. e
levels (mM) of six SCFAs (acetate, butyrate, hexanoate, isobutyrate, isovalerate and propionate) in fermentation
media were quantied. Data are the mean ± SD from three separate experiments. ***P < 0.001 (two-tailed t-test).
Figure2. Electricity production by LCC fermentation of S. epidermidis. (A) e electricity measured by voltage
changes (mV) was recorded for 20min aer pipetting S. epidermidis alone or with LCC (B/LCC), ININ (B/
ININ), PDS (B/PDS) or PETIS (B/PETIS) on the surface of an anode. (B) Cyclic voltammetry was employed to
measure the current (µA) generated by various experimental conditions above. Data presented the mean ± SD
from three separate experiments. ***P < 0.001 (two-tailed t-test).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol.:(0123456789)
Scientic Reports | (2020) 10:21585 | 
www.nature.com/scientificreports/
Topical application of S. epidermidis plus LCC reduced UV‑B‑induced the formation of 4‑HNE
and CPD. It has been documented that UV-B-induced free radicals can cause skin hyperplasia31, lipid
peroxidation32 and the CPD formation33. We next assessed the inuence of fermentation and electricity pro-
duced by S. epidermidis plus LCC on the UV-B-induced skin injuries. e recurrent exposure of UV-B signi-
cantly fostered the formation of 4-HNE and CPD on mouse skin topically applied with LCC or S. epidermidis
alone. In addition, UV-B-induced epidermal hyperplasia, as characterized by an increase in epidermal thickness
(Figure S1) and lesions (Fig.3C), can be detected on mouse skin topically applied with LCC or S. epidermidis
alone. However, the UV-B-induced the formation of 4-HNE, CPD, epidermal hyperplasia and lesions were con-
siderably attenuated when mouse skin was topically applied with S. epidermidis plus LCC (Fig.3, Figure S1).
Topical application of PBS or LCC alone without S. epidermidis on mouse skin before UV-B irradiation exhibited
the same levels of 4-HNE and CPD as well as skin lesions (Figure S2), indicating that LCC itself was insucient
to impede the UV-B-induced skin injuries. Data above dened a novel function for the repurposed LCC in con-
junction with S. epidermidis for suppressing UV-B-induced skin injuries.
The S. epidermidis S2 isolate with low expression of pdh and pta genes was poorly electro‑
genic. Since LCC eectively induced S. epidermidis to undergo fermentation and yield electricity, we next
determined the expression of genes related to fermentation, acetate production, and biolm formation in S.
epidermidis ATCC 12228 and a S. epidermidis S2 strain isolated from human skin. e pdh gene encodes for
pyruvate dehydrogenase, which catalyzes pyruvate to acetyl coenzyme A (acetyl-CoA) at the upstream site of the
fermentation pathway34. e pta gene encoding for phosphate acetyltransferase is involved in the conversion of
acetyl-CoA to acetate35. e icaA gene encoding for intracellular adhesion A has been well characterized in the
engagement of the biolm formation in S. epidermidis36,37. e 16s rRNA sequence of S. epidermidis S2 isolate
shared 99.8% identity to that of S. epidermidis ATCC 12228 (TableS1). However, the expressions of pdh and pta
genes in S. epidermidis S2 isolate were much lower than those in S. epidermidis ATCC 12228 (Fig.4A). Further-
more, the activity of LCC fermentation monitored by OD562 reduction of phenol red-containing rich media for
S. epidermidis S2 isolate was relatively low compared to that in S. epidermidis ATCC 12228 (Fig.4B). e high
expression of icaA gene (Fig.4A) and obvious biolms (Fig.4D) were detected in S. epidermidis S2 isolate, but
not in the non-biolm forming bacterial strain, S. epidermidis ATCC 12228. Interestingly, unlike S. epidermidis
ATCC 12228, the S. epidermidis S2 isolate that expressed low levels of pdh and pta genes was poorly electrogenic.
As shown in Fig.4C, consistent with Fig.2, a peak voltage of approximately 6mV was detected in media with S.
epidermidis ATCC 12228 plus LCC whereas little or no voltage change was measured in S. epidermidis S2 isolate
Figure3. Eect of S. epidermidis in the presence of LCC on the UV-B-induced formation of 4-HNE, CPD and
lesions. e dorsal skin of ICR mice topically applied with LCC alone, S. epidermidis ATCC 12228 alone (B), or
S. epidermidis plus LCC (B/LCC) was irradiated with (+ UV) or without (− UV) UV-B. e images of protein
bands of (A) 4-HNE or (B) CPD analyzed by western blot were displayed. e ratio of intensities of protein
bands of 4-HNE or CPD normalized to β-actin was shown. (C) Morphologies of mouse skin irradiated with
(+ UV) or without (− UV) 100mJ/cm2 UV-B 0, 7, and 14days aer irradiation were shown. Skin lesions were
indicated by white arrows. Data are the mean ± SD from three independent experiments. ***P < 0.001 (two-tailed
t-test). ns = non-signicant.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol:.(1234567890)
Scientic Reports | (2020) 10:21585 | 
www.nature.com/scientificreports/
plus LCC. Collectively, the pdh and pta genes may participate in fermentation and electricity production of S.
epidermidis triggered by LCC.
LCC plus S. epidermidis S2 isolate did not confer protection against UV‑B‑induced skin inju‑
ries. To conrm the critical roles of pdh and pta genes in LCC fermentation of S. epidermidis against UV-B
invivo, dorsal skin of ICR mice was topically applied with S. epidermidis ATCC 12228 or S2 isolate in the pres-
ence of LCC before UV-B irradiation. e levels of 4-HNE and CPD in mouse skin were measured by western
blot. In agreement with data in Fig.3, aer UV-B irradiation, the formation of 4-HNE, CPD and skin lesions
were detected at low levels and moderate on mouse skin topically applied with S. epidermidis plus LCC. How-
ever, when mouse skin was topically applied with S. epidermidis S2 isolate plus LCC (Fig.5A–C), UV-B induced
signicantly increasing levels of 4-HNE and CPD as well as skin lesions. Since the expressions of pdh and pta
genes in S. epidermidis S2 isolate were much lower than those in S. epidermidis ATCC 12228 (Fig.4), the expres-
sions of pdh and pta genes may mediate the LCC-triggered promotion of S. epidermidis against skin injuries
caused by UV-B.
Discussion
LCC is an ester obtained from the reaction of the coconut alcohol-derived fatty acids with a mixture of caprylic
acid and capric acid38. Both caprylic acid and capric acid, as medium-chain fatty acids, can be extracted from
coconut oil and have been used as ingredients for skincare formulation to protect against UV radiation39,40. It
has been reported that addition of caprylic acid into the fermentation process enhanced acetate production41.
Coconut oil has been used as a fuel for electricity production42. Although many medium-chain fatty acids exhib-
ited potent bactericidal activities43, our result in Figure S3 demonstrated that LCC did not change the growth of
S. epidermidis. Compared to other INCI-registered compounds (ININ, PDS and PETIS), LCC displayed higher
activity in promoting fermentation and electricity production of S. epidermidis (Fig.1). Biolms are electroactive
and can promote the electricity production of bacteria44. By using S. epidermidis ATCC 12228, a non-biolm
forming strain, we demonstrated that skin bacteria can produce electricity without biolm formation on elec-
trodes. Data in our recent publication has revealed that addition of glycerol into the S. epidermidis culture can
induce fermentation and instantly produce detectable electricity, highlighting a possible mechanism that skin
bacteria underwent fermentation to accumulate SCFAs as electron donors to intensify electricity18.
Results in Fig.1 demonstrated that the electricity measured by changes in voltage and currents was con-
siderably produced when the anode was pipetted with S. epidermidis ATCC 12228 plus 2% LCC. However, the
electricity produced by bacteria plus LCC was largely reduced in the S. epidermidis S2 isolate which expressed
Figure4. e gene (pdh, pta, and icaA) expression, electricity and biolm formation in S. epidermidis ATCC
12228 and S2 isolate. (A) Relative expression of pdh, pta, and icaA genes normalized to 16S rRNA was analyzed
by the RT-qPCR. (B) LCC fermentation for 12h in rich media was quantied by measurement of OD562. (C)
Electricity production of bacteria in the presence of 2% LCC was analyzed by voltage changes (mV) in an
invitro chamber. (D) Bacterial biolms were stained by crystal violet aer culture of bacteria in TSB on a
24-well plate for 48h. B/LCC: S. epidermidis ATCC 12228 plus 2% LCC; S2/LCC: S. epidermidis S2 isolate plus
2% LCC. Scale bars = 1cm. Data are the mean ± SD from three separate experiments. **P < 0.01; ***P < 0.001
(two-tailed t-test).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol.:(0123456789)
Scientic Reports | (2020) 10:21585 | 
www.nature.com/scientificreports/
lower levels of pdh and pta genes (Fig.3). e data indicated that proteins corresponding to pdh and pta genes
may play a role in electricity production of S. epidermidis in the presence of LCC. Electrons are derived by the
reduction reaction of NAD+ to NADH in the metabolic pathway of bacterial fermentation45. e conversion of
NAD+ to NADH is involved in pyruvate dehydrogenase (pdh), which initiates the process of electron transport
chain45,46. Previous studies have shown that electricity production of Shewanella oneidensis MR-1, a representa-
tive electrochemically active bacterium (EAB) extensively studied in the laboratory, was mediated by activation
of NAD+-linked PDH47. Phosphate acetyltransferase (pta) can catalyze the conversion of acetyl-CoA to acetate,
a known electron donor. A pta knockout strain of Shewanella oneidensis strain has been used to study the eect
of electricity on bacterial fermentation48. e protective eect of bacterial fermentation on the suppression of
the UV-B-induced formation of 4-HNE and CPD was remarkably diminished when mouse skin was topically
applied with LCC and S. epidermidis S2 isolate which yielded low electricity and expressed lower levels of pdh and
pta genes (Fig.5). e data suggested that SCFAs and electricity induced by LCC fermentation of S. epidermidis
may synergistically provide mice protection from UV-B injuries. Future studies will include the construction
of a pdh knockout S. epidermidis strain and investigation of the essential role of pdh gene in S. epidermidis for
production of electricity against UV-B injuries.
4-HNE is known to be genotoxic and damages DNA by producing bulky 1,N2-propano-2-deoxyguanosine
adducts49. Here, we showed that 4-HNE and CPD were formed when mouse skin was constantly bombarded
with UV-B. Results in our previous studies have demonstrated that butyrate produced by glycerol fermentation
of S. epidermidis can down-regulate UV-B-induced pro-inammatory interleukin (IL)-6 cytokines through SCFA
receptor 2 (FFAR2)50. us, skin bacteria may take advantage of endogenous glycerol as a carbon source to pro-
voke fermentation and simultaneously produced SCFAs and electrons. Besides being electron donors, SCFAs may
regulate FFAR2 and/or histone deacetylases (HDAC)51 to mitigate the inammation induced by UV irradiation.
Electrons may play a key role in neutralizing free radicals generated by UV irradiation. It has been shown that UV
light can mediate nitrate, a constituent of sweat in skin, to generate free radicals52, which can subsequently induce
lipid peroxidation to produce 4-HNE53. However, commensal bacteria can utilize nitrate as an electron acceptor
Figure5. e UV-B-induced formation of 4-HNE, CPD and lesions in mouse skin applied with LCC in
combination with S. epidermidis S2 isolate. e dorsal skin of ICR mice was topically applied with S. epidermidis
ATCC 12228 (B/LCC) or S. epidermidis S2 (S2/LCC) in the presence of 2% LCC. e levels of (A) 4-HNE, (B)
CPD related to β-actin in western blot analysis and (C) lesions (at 0, 7, and 14days post-irradiation) on mouse
skin irradiated with 100mJ/cm2 UV-B (+ UV) were shown. Skin lesions were indicated by white arrows. Data
are the mean ± SD from three separate experiments. **P < 0.01; ***P < 0.001 (two-tailed t-test).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol:.(1234567890)
Scientic Reports | (2020) 10:21585 | 
www.nature.com/scientificreports/
for energy transduction54. For example, under anoxic or oxygen-depleted conduction, Pseudomonas species can
mediate denitrication by using nitrate as an electron acceptor to convert nitrate to nitrogenous gases55. us,
a possible mechanism behind the protective eect of S. epidermidis plus LCC on UV-B skin injuries is that LCC
triggers bacteria to produce electrons, activating bacterial denitrication to reduce UV-B-induced free radicals.
In summary, prebiotics for skin probiotic bacteria have been not dened by the Food and Drug Administra-
tion (FAD) even though they may become novel therapeutics for treatments of skin disorders via induction of
fermentation of skin bacteria. Our study here demonstrated an approach by repurposing INCI-registered com-
pounds as skin prebiotics and revealed their capabilities of generating electricity from skin bacteria to combat
UV-B-induced skin damages.
Received: 6 October 2020; Accepted: 12 November 2020
References
1. Sleigh, S. H. & Barton, C. L. Repurposing strategies for therapeutics. Pharm. Med. 24, 151–159. https ://doi.org/10.1007/BF032
56811 (2010).
2. Ashburn, T. T. & or, K. B. Drug repositioning: identifying and developing new uses for existing drugs. Nat. Rev. Drug Discovery
3, 673–683. https ://doi.org/10.1038/nrd14 68 (2004).
3. Tartaglia, L. A. Complementary new approaches enable repositioning of failed drug candidates. Expert Opin. Investig. Drugs 15,
1295–1298. https ://doi.org/10.1517/13543 784.15.11.1295 (2006).
4 . Beigel, J. H. et al. Remdesivir for the Treatment of Covid-19—Preliminary Report. https ://doi.org/10.1056/NEJM o a2007 764 (2020).
5. Tomazini, B. M. et al. COVID-19-associated ARDS treated with DEXamethasone (CoDEX): study design and rationale for a
randomized trial. Rev. Bras. Terapia Intens. https ://doi.org/10.1101/2020.06.24.20139 303 (2020).
6. Zeeuwen, P. L., Kleerebezem, M., Timmerman, H. M. & Schalkwijk, J. Microbiome and skin diseases. Curr. Opin. Allergy Clin.
Immunol. 13, 514–520 (2013).
7. Edmonds-Wilson, S. L., Nurinova, N. I., Zapka, C. A., Fierer, N. & Wilson, M. Review of human hand microbiome research. J.
Dermatol. Sci. 80, 3–12. https ://doi.org/10.1016/j.jderm sci.2015.07.006 (2015).
8. Grice, E. A. et al. A diversity prole of the human skin microbiota. Genome Res. 18, 1043–1050. https ://doi.org/10.1101/gr.07554
9.107 (2008).
9. Keshari, S. et al. Butyric acid from probiotic staphylococcus epidermidis in the skin microbiome down-regulates the ultraviolet-
induced pro-inammatory IL-6 cytokine via short-chain fatty acid receptor. Int. J. Mol. Sci. 20, 4477. https ://doi.org/10.3390/ijms2
01844 77 (2019).
10. Kao, M.-S. et al. Microbiome precision editing: using PEG as a selective fermentation initiator against methicillin-resistant Staphy-
lococcus aureus. Biotechnol. J. https ://doi.org/10.1002/biot.20160 0399 (2016).
11. Gibson, G. R. & Roberfroid, M. B. Dietary modulation of the human colonic microbiota: introducing the concept of prebiotics. J.
Nutr. 125, 1401–1412. https ://doi.org/10.1093/jn/125.6.1401 (1995).
12. Gibson, G. R., Probert, H. M., Loo, J. V., Rastall, R. A. & Roberfroid, M. B. Dietary modulation of the human colonic microbiota:
updating the concept of prebiotics. Nutr. Res. Rev. 17, 259–275. https ://doi.org/10.1079/nrr20 0479 (2004).
13. Gibson, G. R. et al. Expert consensus document: e International Scientic Association for Probiotics and Prebiotics (ISAPP) con-
sensus statement on the denition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 14, 491–502. htt ps ://doi.org/10.1038/
nrgas tro.2017.75 (2017).
14. Wang, Y. et al. Staphylococcus epidermidis in the human skin microbiome mediates fermentation to inhibit the growth of propi-
onibacterium acnes: implications of probiotics in acne vulgaris. Appl. Microbiol. Biotechnol. 98, 411–424. https ://doi.org/10.1007/
s0025 3-013-5394-8 (2014).
15. Shi, L. et al. Extracellular electron transfer mechanisms between microorganisms and minerals. Nat. Rev. Microbiol. 14, 651–662.
https ://doi.org/10.1038/nrmic ro.2016.93 (2016).
16. Light, S. H. et al. A avin-based extracellular electron transfer mechanism in diverse gram-positive bacteria. Nature 562, 140–144
(2018).
17. Matsuda, S., Liu, H., Kato, S., Hashimoto, K. & Nakanishi, S. Negative faradaic resistance in extracellular electron transfer by
anode-respiring Geobacter sulfurreducens cells. Environ. Sci. Technol. 45, 10163–10169. https ://doi.org/10.1021/es200 834b (2011).
18. Balasubramaniam, A. et al. Skin bacteria mediate glycerol fermentation to produce electricity and resist UV-B. Microorganisms 8,
1092 (2020).
19. Sohn, M. et al. Eect of emollients on UV lter absorbance and sunscreen eciency. J. Photochem. Photobiol. B Biol. 205, 111818
(2020).
20. Subongkot, T., Sirirak, T. J. C. & Biointerfaces, S. B. Development and skin penetration pathway evaluation of microemulsions for
enhancing the dermal delivery of celecoxib. 111103 (2020).
21. Peneld, K. in AIP Conference Proceedings. 899–901 (American Institute of Physics).
22. Johnson, W. Jr. et al. Safety assessment of PEG-150 pentaerythrityl tetrastearate as used in cosmetics. Int. J. Toxicol. 37, 5s–9s. https
://doi.org/10.1177/10915 81818 79445 7 (2018).
23. Jurkiewicz, B. A. & Buettner, G. R. Ultraviolet light-induced free radical formation in skin: an electron paramagnetic resonance
study. Photochem. Photobiol. 59, 1–4. https ://doi.org/10.1111/j.1751-1097.1994.tb049 93.x (1994).
24. Slominski, A. T., Zmijewski, M. A., Plonka, P. M., Szaarski, J. P. & Paus, R. How UV light touches the brain and endocrine system
through skin, and why. Endocrinology 159, 1992–2007. https ://doi.org/10.1210/en.2017-03230 %JEndo crino logy (2018).
25. Kao, M.-S. et al. Microbiome precision editing: using PEG as a selective fermentat ion initiator against methicillin-resistant Staphy-
lococcus aureus. Biotechnol. J. https ://doi.org/10.1002/biot.20160 0399 (2017).
26. Kao, M. S. et al. Microbiome precision editing: using PEG as a selective fermentation initiator against methicillin-resistant Staphy-
lococcus aureus. Biotechnol J. https ://doi.org/10.1002/biot.20160 0399 (2017).
27. Finke, N., Vandieken, V. & Jorgensen, B. B. Acetate, lactate, propionate, and isobutyrate as electron donors for iron and sulfate
reduction in Arctic marine sediments, Svalbard. FEMS Microbiol. Ecol. 59, 10–22. https ://doi.o rg/10.1111/j.1574-6941.2006.00214
.x (2007).
28. Chen, C., Shen, Y., An, D. & Voordouw, G. Use of acetate, propionate, and butyrate for reduction of nitrate and sulfate and metha-
nogenesis in microcosms and bioreactors simulating an oil reservoir. Appl Environ Microbiol. https ://doi.org/10.1128/aem.02983
-16 (2017).
29. S orokin, D., Detkova, E. & Muyzer, G. Propionate butyrate dependent bacterial sulfate reduction at extremely haloalkaline condi-
tions description of Desulfobotulus alkaliphilus sp. nov. extremophiles. Extremophiles Life Under Extreme Conditions 14, 71–77.
https ://doi.org/10.1007/s0079 2-009-0288-5 (2009).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Vol.:(0123456789)
Scientic Reports | (2020) 10:21585 | 
www.nature.com/scientificreports/
30. Kumar, S. et al. Improved levulinic acid production from agri-residue biomass in biphasic solvent system through synergistic
catalytic eect of acid and products. Bioresour. Technol. 251, 143–150. https ://doi.org/10.1016/j.biort ech.2017.12.033 (2018).
31. Tyrrell, R. M. Ultraviolet radiation and free radical damage to skin. Biochem. Soc. Symp. 61, 47–53. https ://doi.org/10.1042/bss06
10047 %JBioc hemic alSoc ietyS ympos ia (1995).
32. Halliday, G. M. Inammation, gene mutation and photoimmunosuppression in response to UVR-induced oxidative damage
contributes to photocarcinogenesis. Mutat. Res. 571, 107–120. https ://doi.org/10.1016/j.mrfmm m.2004.09.013 (2005).
33. Bowden, G. T. Prevention of non-melanoma skin cancer by targeting ultraviolet-B-light signalling. Nat. Rev. Cancer 4, 23–35. https
://doi.org/10.1038/nrc12 53 (2004).
34. Zheng, P., Wereath, K., Sun, J., van den Heuvel, J. & Zeng, A.-P. Overexpression of genes of the dha regulon and its eects on cell
growth, glycerol fermentation to 1, 3-propanediol and plasmid stability in Klebsiella pneumoniae. Process Biochem. 41, 2160–2169
(2006).
35. Wolin, M. J. Interactions between H2-producing and methane-producing species. Microbial formation and utilization of gases,
141–150 (1976).
36. Namvar, A. E., Asghari, B., Ezzatifar, F., Azizi, G. & Lari, A. R. Detection of the intercellular adhesion gene cluster (ICA) in clinical
Staphylococcus aureus isolates. GMS Hyg. Infect. Control 8, Doc03–Doc03. https ://doi.org/10.3205/dgkh0 00203 (2013).
37. Kıvanç, S. A., Arık, G., Akova-Budak, B. & Kıvanç, M. Biolm forming capacity and antibiotic susceptibility of Staphylococcus
spp. with the icaA/icaD/bap genotype isolated from ocular surface of patients with diabetes. Malawi Med J 30, 243–249. https ://
doi.org/10.4314/mmj.v30i4 .6 (2018).
38. Fiume, M. M. et al. Safety assessment of alkyl esters as used in cosmetics. Int. J. Toxicol. 34, 5S-69S (2015).
39. Fonseca, B. L. et al. Neuroprotective eects of a new skin care formulation following ultraviolet exposure. Cell Prolif. 45, 48–52.
https ://doi.org/10.1111/j.1365-2184.2011.00795 .x (2012).
40. Bacqueville, D. et al. Ecacy of a dermocosmetic serum combining bakuchiol and vanilla tahitensis extract to prevent skin pho-
toaging invitro and to improve clinical outcomes for naturally aged skin. Clin. Cosmet. Investig. Dermatol. 13, 359–370. https ://
doi.org/10.2147/CCID.S2358 80 (2020).
41. Abel, H., Immig, I. & Harman, E. Eect of adding capr ylic and capric acid to grass on fermentation characteristics during ensiling
and in the articial rumen system RUSITEC. Anim. Feed Sci. Technol. 99, 65–72. https ://doi.org/10.1016/S0377 -8401(02)00084 -6
(2002).
42. Włodarczyk, P., Włodarczyk, B. & Kalinichenko, A. Direct electricity production from coconut oil - the elec trooxidation of coconut
oil in an acid electrolyte. E3S Web Conf. 45, 00103. https ://doi.org/10.1051/e3sco nf/20184 50010 3 (2018).
43. Liu, S.-T., Sugimoto, T., Azakami, H. & Kato, A. Lipophilization of lysozyme by short and middle chain fatty acids. J. Agric. Food
Chem. https ://doi.org/10.1021/jf990 4822 (2000).
44. Reguera, G. Microbial nanowires and electroactive biolms. FEMS Microbiol. Ecol. https ://doi.org/10.1093/femse c/y08 6 (2018).
45. Cahoon, L. A. & Freitag, N. E. e electrifying energy of gut microbes. Nature 562, 43–44. https ://doi.org/10.1038/d4158 6-018-
06180 -z (2018).
46. S erwańska-Leja, K., Czaczyk, K. & Myszka, K. Biotechnological synthesis of 1,3-propanediol using Clostridium ssp. Afr. J. Biotech.
10, 11093–11101. https ://doi.org/10.5897/AJB11 .873 (2011).
47. Madsen, C. S. & TerAvest, M. A. NADH dehydrogenases Nuo and Nqr1 contribute to extracellular electron transfer by Shewanella
oneidensis MR-1 in bioelectrochemical systems. Sci. Rep. 9, 14959. https ://doi.org/10.1038/s4159 8-019-51452 -x (2019).
48. Flynn, J. M., Ross, D. E., Hunt, K. A., Bond, D. R. & Gralnick, J. A. Enabling unbalanced fermentations by using engineered
electrode-interfaced bacteria. mBio https ://doi.org/10.1128/mBio.00190 -10 (2010).
49. Choudhury, S., Pan, J., Amin, S., Chung, F.-L. & Roy, R. Repair kinetics of trans-4-hydroxynonenal-induced cyclic 1, N2-pro-
panodeoxyguanine DNA adducts by human cell nuclear extracts. Biochemistry 43, 7514–7521. https ://doi.org/10.1021/bi049 877r
(2004).
50. Keshari, S. et al. Butyric acid from probiotic Staphylococcus epidermidis in the skin microbiome down-regulates the ultraviolet-
induced pro-inammatory IL-6 cytokine via short-chain fatty acid receptor. Int. J. Mol. Sci. https ://doi.org/10.3390/ijms2 01844
77 (2019).
51. Vinolo, M. A. R., Rodrigues, H. G., Nachbar, R. T. & Curi, R. Regulation of inammation by short chain fatty acids. Nutrients 3,
858–876. https ://doi.org/10.3390/nu310 0858 (2011).
52. Suzuki, T. & Inukai, M. Eects of nitrite and nitrate on DNA damage induced by ultraviolet light. Chem. Res. Toxicol. 19, 457–462.
https ://doi.org/10.1021/tx050 347l (2006).
53. Yang, Y., Sharma, R., Sharma, A., Awasthi, S. & Awasthi, Y. C. Lipid peroxidation and cell cycle signaling: 4-hydroxynonenal, a
key molecule in stress mediated signaling. Acta Biochim. Pol. 50, 319–336 (2003).
54. Wilson, L. P. & Bouwer, E. J. Biodegradation of aromatic compounds under mixed oxygen/denitrifying conditions: a review. J. Ind.
Microbiol. Biotechnol. 18, 116–130. https ://doi.org/10.1038/sj.jim.29002 88 (1997).
55. Borrero-de Acuña, J. M., Timmis, K. N., Jahn, M. & Jahn, D. Protein complex formation during denitrication by Pseudomonas
aeruginosa. Microbial. Biotechnol. 10, 1523–1534. https ://doi.org/10.1111/1751-7915.12851 (2017).
Acknowledgements
e study was supported by the Ministry of Science and Technology (MOST) Grants (108-2622-B-008-001-CC1;
108-2314-B-008-003-MY3, and 107-2923-B-008-001-MY3) and 106/107/108-Landseed Hospital-NCU joint
Grants. We thank Supitchaya Traisaeng at National Central University for assistance at biolm staining.
Author contributions
A.B.: Methodology; A.B., and P.A.: Soware; A.B., P.A., D.T.T.M., S.K., and R.S.: Data curation; A.B.: Writing-
Original dra preparation; C.-M.H. and C.-L.C.: Ethical evaluation; C.-M.H.: Conceptualization, Visualization,
Investigation, Methodology, Supervision, Writing-Reviewing and Editing.
Competing interests
e authors declare no competing interests.
Additional information
Supplementary information is available for this paper at https ://doi.org/10.1038/s4159 8-020-78132 -5.
Correspondence and requests for materials should be addressed to C.-M.H.
Reprints and permissions information is available at www.nature.com/reprints.
Content courtesy of Springer Nature, terms of use apply. Rights reserved

Vol:.(1234567890)
Scientic Reports | (2020) 10:21585 | 
www.nature.com/scientificreports/
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access is article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons licence, and indicate if changes were made. e images or other third party material in this
article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the
material. If material is not included in the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this licence, visit http://creat iveco mmons .org/licen ses/by/4.0/.
© e Author(s) 2020
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
... The S. epidermidis ATCC 12228 is a biofilm-negative strain [52] and carries a truncated copy of the J1 region of SCCmec type I [53]. Mounting research documents the probiotic activity of S. epidermidis ATCC 12228 to produce SCFAs in the presence of various carbon sources [54,55]. Figure 4 illustrated that the values of similarities of the first 20-bp domain at positions 1-400 of 16S rRNA genes varied between three human scalp-isolated S. epidermidis (S1-S3) and five acne lesion-isolated S. epidermidis (Ac1-Ac5). ...
Article
Full-text available
Staphylococcus epidermidis (S. epidermidis) live in different human locations and natural environments. For ribotyping S. epidermidis sub-species, 2507 PCR-amplified reads of 16S rRNA genes of S. epidermidis in a public dataset were used for probabilistic sequence analysis. A sequence probability logo (sequence pLogo) as a reference sequence of 16S rRNA genes of S. epidermidis was constructed. Through implementation of Levenshtein Distance algorithm, two 20-base pairs (bp) motifs, commonly present in 2507 PCR-amplified reads, were identified. The top 38 S. epidermidis isolates, which carried 16S rRNA nucleotide domains that were made of different sequences but have high similarity scores to two 20-bp motifs, were found from 11 human, 8 animal, 9 plant and 10 environmental samples, indicating that these two 20-bp motifs were broadly present in diverse S. epidermidis isolates. Thirty-one PCR-amplified reads of 16S rRNA genes, which were currently not in the dataset, were utilized to verify the feasibility of using two 20-bp motifs for ribotyping S. epidermidis sub-species. S. epidermidis S1, S3, but not S2, isolates on the human scalp carried a 20-bp sequence domain with high similarities to a 20-bp motif in the sequence pLogo. The phylogenetic tree showed that S. epidermidis S1, S2 and S3 were not from a single common ancestor. Two newly identified 20-bp motifs here, thus, provided reference nucleotide residues for ribotyping S. epidermidis.
... Staphylococcus epidermidis merupakan salah satu bakteri gram positif yang dapat ditemukan secara luas di kulit [16]. Kemampuan bakteri ini dalam menghasilkan elektron dan mendonorkannya ke penerima donor elektron yang berada di luar sel telah dilaporkan [17][18][19] c. Uji ferric iron reductase S. epidermidis ATCC 12228 1x10 7 CFU/ml dipipetkan di atas permukaan media agar yang tersusun atas rich medium, (10 ml) [10 g/l yeast extract (Biokar Diagnostics, Beauvais, France), 5 g/l TSB, 2.5 g/l K2HPO4, and 1.5 g/l KH2PO4] yang ditambah dengan gliserol (20g/l) dan 0,1 mg/ml ferric ammonium citrate. ...
Article
Full-text available
The ability of electrogenic bacteria to generate electricity has been widely reported. In some cases of bacteria, the electricity production comes from the bacterial fermentation process of SFI compounds by these bacteria. Later, the resulted electrons are transferred out from inside to extracellular recipient molecules. Among of these studies has shown the ability of gram-positive bacteria S. epidermidis ATCC 12228 in terms of SFI compounds utilization to increase bacterial electron production and its application in the medical field. Based on these studies, the discovery of new SFI compounds becomes interesting to be explored. In this study, a new SFI compound was screened from 24 different compounds. The screen was initiated by testing the ability of these compounds to increase the fermentation activity of S. epidermidis ATCC 12228 in a 96-well plate. Determination of SFI compound was carried out by checking the exclusivity of the compound to increase the fermentation activity of S. epidermidis ATCC 12228. The selected SFI compound was then tested for cytotoxicity against this bacterium and its ability to increase the electron production of S. epidermidis ATCC 12228 using a microbial fuel cell (MCF). This study was successfully demonstrated the non-toxic properties of p-coumaric acid, also the ability of this compound to increase the fermentation activity and electron production of S. epidermidis ATCC 12228. This research is expected to be the first step to find another novel SFI compounds that will be useful in certain fields in the future.
... Significantly reduced UV-B-induced injuries based on the parameters such as formation of 4-hydroxynonenal (4-HNE), cyclobutane pyrimidine dimers (CPD), and skin lesions Balasubramaniam et al. (2020b) 19 ...
Article
Sunlight is pivotal for our survival, and daily UV exposure has impacted the evolutionary course of all forms of life, from microorganisms to humans. Deciphering the role of UVR in regulating the microbial dynamics of environmental and host-associated microbes is crucial. UVR may be responsible for affecting skin pathology by influencing the skin microbiome, both qualitatively and quantitatively, as evident in low-dose narrow-band UVB phototherapy. Some findings have suggested that the skin microbiome has immunomodulatory roles when exposed to UVR; however, its involvement in UV screening or protection has yet to be fully explored. Furthermore, numerous skin disorders are associated not only with an altered skin microbiome but also with an altered gut microbiome. Hence, the skin-gut axis needs to be in physiological homeostasis and immunological harmony. The purpose of this review is to examine the impact of natural UVR on human immunomodulatory mechanisms and the associated cutaneous microbiome, with an emphasis on interactions among UVR, skin homeostasis, vitamin D, and the related skin-gut axis. With the ‘nature as an inspiration’ approach, ongoing research is trying to decipher photoprotective secrets in several microbial-based natural compounds to be used as sunscreens or other topical formulations. In addition, various probiotics have also been shown to have significant antioxidant, antiwrinkle, and antiaging effects that ameliorate UV-induced cellular and molecular damage, as highlighted in the review. These cosmetics, nutricosmetics, and probiotaceuticals will undoubtedly be next-generation solutions against photoaging and maintaining skin health.
Article
The genomic revolution has fueled rapid progress in synthetic and systems biology, opening up new possibilities for using live biotherapeutic products (LBP) to treat, attenuate or prevent human diseases. Among LBP, bacteria-based therapies are particularly promising due to their ability to colonize diverse human tissues, modulate the immune system and secrete or deliver complex biological products. These bacterial LBP include engineered pathogenic species designed to target specific diseases, and microbiota species that promote microbial balance and immune system homeostasis, either through local administration or the gut-body axes. This review focuses on recent advancements in preclinical and clinical trials of bacteria-based LBP, highlighting both on-site and long-reaching strategies.
Article
Objective Oligosaccharides have been shown to enhance the production of short chain fatty acids (SCFAs) by gut probiotics and regulate gut microbiota, to improve intestinal health. Recent research indicates that oligosaccharides may also positively impact skin microbiota by selectively promoting the growth of skin commensal bacteria and inhibiting pathogenic bacteria. However, the specific metabolic and regulatory mechanisms of skin commensal bacteria in response to oligosaccharides remain unclear. This study aims to explore the influence of four oligosaccharides on the growth and metabolism of Staphylococcus epidermidis and further identify skin prebiotics that can enhance its probiotic effects on the skin. Methods Fructooligosaccharides (FOS), isomaltooligosaccharide (IMO), galactooligosaccharides (GOS) and inulin were compared in terms of their impact on cell proliferation, SCFAs production of S . epidermidis CCSM0287 and the biofilm inhibition effect of their fermentation supernatants on Staphylococcus aureus CCSM0424. Furthermore, the effect of FOS on S . epidermidis CCSM0287 was analysed by the transcriptome analysis. Results All four oligosaccharides effectively promoted the growth of S . epidermidis CCSM0287 cells, increased the production of SCFAs, with FOS demonstrating the most significant effect. Analysis of the SCFAs indicated that S . epidermidis CCSM0287 predominantly employs oligosaccharides to produce acetic acid and isovaleric acid, differing from the SCFAs produced by gut microbiota. Among the four oligosaccharides, the addition of 2% FOS fermentation supernatant significantly inhibited S . aureus CCSM0424 biofilm formation. Furthermore, RNA sequencing revealed 162 differentially expressed genes (84 upregulated and 78 downregulated) of S . epidermidis CCSM0287 upon FOS treatment compared with glucose treatment. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis highlighted differences in the amino acid synthesis pathway, particularly in terms of arginine biosynthesis. Conclusion FOS promotes cell proliferation, increases the SCFA production of S . epidermidis CCSM0287 and enhance the inhibition of S . aureus biofilm formation, suggesting that FOS serves as a potential prebiotic for strain S . epidermidis CCSM0287.
Article
Full-text available
The skin microbiome undergoes constant exposure to solar radiation (SR), with its effects on health well‐documented. However, understanding SR's influence on host‐associated skin commensals remains nascent. This review surveys existing knowledge on SR's impact on the skin microbiome and proposes innovative sun protection methods that safeguard both skin integrity and microbiome balance. A team of skin photodamage specialists conducted a comprehensive review of 122 articles sourced from PubMed and Research Gateway. Key terms included skin microbiome, photoprotection, photodamage, skin cancer, ultraviolet radiation, solar radiation, skin commensals, skin protection, and pre/probiotics. Experts offered insights into novel sun protection products designed not only to shield the skin but also to mitigate SR's effects on the skin microbiome. Existing literature on SR's influence on the skin microbiome is limited. SR exposure can alter microbiome composition, potentially leading to dysbiosis, compromised skin barrier function, and immune system activation. Current sun protection methods generally overlook microbiome considerations. Tailored sun protection products that prioritize both skin and microbiome health may offer enhanced defense against SR‐induced skin conditions. By safeguarding both skin and microbiota, these specialized products could mitigate dysbiosis risks associated with SR exposure, bolstering skin defense mechanisms and reducing the likelihood of SR‐mediated skin issues.
Chapter
Skin, the body’s largest organ, harbors a unique microbial community crucial for maintaining skin health. The cutaneous immune system and skin microbiome keep the pathogens at bay, and any anomaly generated in this tightly linked network culminates in skin abnormalities. Dysbiotic microbiome conditions are often observed in skin disorders suggesting their prominent role in protecting skin health. This chapter will discuss the components of the normal skin microbiome and its interactions with the immune system and the modern environment. In addition, the implications of cosmetics on skin microbiome, the association of microbiota with skin disorders, and therapeutic interventions have been discussed in detail.
Preprint
Full-text available
The cytokine storm leads to acute respiratory distress syndrome (ARDS) and multi-organ dysfunction in COVID-19 cases. One of the major cytokines is IL-6, which is positively associated with the severity of the condition. In the present study, nasal inoculation of SARS-CoV-2 nucleocapsid phosphoprotein (NPP) elevated IL-6 abundancy in bronchoalveolar lavage fluid (BALF) in mice. Staphylococcus epidermidis K1 ( S. epidermidis K1)-colonized mice nasally administered with glycerol or FMN resulted in a reduction of NPP-induced IL-6. S. epidermidis fermented glycerol to generate a large amount of electrons which further supported bacterial colonization. Inhibition of riboflavin kinase in S. epidermidis considerably suppressed the effect of nasal S. epidermidis plus glycerol on the decrement of NPP-induced IL-6. Taken together, our results show that nasal colonization of S. epidermidis mitigates a cytokine storm caused by SARS-CoV-2 infection.
Article
Full-text available
Cosméticos estão entre os produtos mais inovadores do mercado da beleza, sendo os cuidados com a microbiota cutânea uma tendência. Neste trabalho, realizou-se uma prospecção na base patentária Questel Orbit® e nas bases científicas PubMed e ScienceDirect, a fim de identificar os mercados e as tecnologias envolvendo o uso cosmético de probióticos. Foram encontrados 115 depósitos de patentes entre 2010-2020, enquanto o número de artigos científicos foi de 48. A China é o país com mais patentes, mas a multinacional europeia L’oréal, do ramo cosmético, é a principal detentora dessas tecnologias, inclusive com parcerias com a centenária do setor alimentício, Nestlé. O Brasil destaca-se como segundo maior mercado de cosméticos e ocupa a terceira posição no ranking de produtos com probióticos. Diante do panorama de crescimento em inovações de cosméticos probióticos, compreender a dinâmica do setor para garantir a segurança e a qualidade dos produtos é de fundamental importância.
Article
The electrogenicity of environmental bacteria has been thoroughly explored and has been known to have the unique capability of decomposing hazardous chemicals for environmental remediation. However, electrogenic bacteria in human skin in regards to their electrical properties and locations have not yet been determined. Here, electrodermal activities and metabolite compositions at different locations of arm skin were assessed. Compared to the uppermost part of arm, we found that the forearm elicited high electrodermal activity and carried abundant lactate and alpha-ketoglutarate, two components commonly present in sweat. Upon culturing bacteria from the forearm, an iron-resistant strain of Staphylococcus warneri (S. warneri) was identified through 16S ribosomal RNA sequencing. Voltage changes induced by S. warneri in the presence of glucose were detected by two voltmeters of different electrode materials, demonstrating the electrogenicity of skin bacteria. Furthermore, we discovered that S. warneri has the ability to metabolize lactate to generate electricity. The results of this study reveal changes in skin conductance caused by bacterial electricity that are mediated by skin endogenous molecules and may provide a novel method of monitoring environmental skin insults.
Article
Full-text available
Objective: The infection caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spreads worldwide and is considered a pandemic. The most common manifestation of SARS-CoV-2 infection (coronavirus disease 2019 - COVID-19) is viral pneumonia with varying degrees of respiratory compromise and up to 40% of hospitalized patients might develop acute respiratory distress syndrome. Several clinical trials evaluated the role of corticosteroids in non-COVID-19 acute respiratory distress syndrome with conflicting results. We designed a trial to evaluate the effectiveness of early intravenous dexamethasone administration on the number of days alive and free of mechanical ventilation within 28 days after randomization in adult patients with moderate or severe acute respiratory distress syndrome due to confirmed or probable COVID-19. Methods: This is a pragmatic, prospective, randomized, stratified, multicenter, open-label, controlled trial including 350 patients with early-onset (less than 48 hours before randomization) moderate or severe acute respiratory distress syndrome, defined by the Berlin criteria, due to COVID-19. Eligible patients will be randomly allocated to either standard treatment plus dexamethasone (Intervention Group) or standard treatment without dexamethasone (Control Group). Patients in the intervention group will receive dexamethasone 20mg intravenous once daily for 5 days, followed by dexamethasone 10mg IV once daily for additional 5 days or until intensive care unit discharge, whichever occurs first. The primary outcome is ventilator-free days within 28 days after randomization, defined as days alive and free from invasive mechanical ventilation. Secondary outcomes are all-cause mortality rates at day 28, evaluation of the clinical status at day 15 assessed with a 6-level ordinal scale, mechanical ventilation duration from randomization to day 28, Sequential Organ Failure Assessment Score evaluation at 48 hours, 72 hours and 7 days and intensive care unit -free days within 28.
Article
Full-text available
Bacteria that use electron transport proteins in the membrane to produce electricity in the gut microbiome have been identified recently. However, the identification of electrogenic bacteria in the skin microbiome is almost completely unexplored. Using a ferric iron-based ferrozine assay, we have identified the skin Staphylococcus epidermidis (S. epidermidis) as an electrogenic bacterial strain. Glycerol fermentation was essential for the electricity production of S. epidermidis since the inhibition of fermentation by 5-methyl furfural (5-MF) significantly diminished the bacterial electricity measured by voltage changes in a microbial fuel cell (MFC). A small-scale chamber with both anode and cathode was fabricated in order to study the effect of ultraviolet-B (UV-B) on electricity production and bacterial resistance to UV-B. Although UV-B lowered bacterial electricity, a prolonged incubation of S. epidermidis in the presence of glycerol promoted fermentation and elicited higher electricity to suppress the effect of UV-B. Furthermore, the addition of glycerol into S. epidermidis enhanced bacterial resistance to UV-B. Electricity produced by human skin commensal bacteria may be used as a dynamic biomarker to reflect the UV radiation in real-time.
Preprint
Full-text available
OBJECTIVES: The infection caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) spreads worldwide and is considered a pandemic. The most common manifestation of SARS-CoV2 infection (Coronavirus disease 2019 - COVID-19) is viral pneumonia with varying degrees of respiratory compromise and up to 40% of hospitalized patients might develop Acute Respiratory Distress Syndrome (ARDS). Several clinical trials evaluated the role of corticosteroids in non-COVID-19 ARDS with conflicting results. We designed a trial to evaluate the effectiveness of early intravenous dexamethasone administration on the number of days alive and free of mechanical ventilation within 28 days after randomization in adult patients with moderate or severe ARDS due to confirmed or probable COVID-19. METHODS: This is a pragmatic, prospective, randomized, stratified, multicenter, open-label, controlled trial including 350 patients with early-onset (less than 48h before randomization) moderate or severe ARDS, defined by the Berlin criteria, due to COVID-19. Eligible patients will be randomly allocated to either standard treatment plus dexamethasone (intervention group) or standard treatment without dexamethasone (control group). Patients in the intervention group will receive dexamethasone 20mg IV once daily for 5 days, followed by dexamethasone 10mg IV once daily for additional 5 days or until Intensive Care Unit (ICU) discharge, whichever occurs first. The primary outcome is ventilator-free days within 28 days after randomization, defined as days alive and free from invasive mechanical ventilation. Secondary outcomes are all-cause mortality rates at day 28, evaluation of the clinical status at day 15 assessed with a 6-level ordinal scale, mechanical ventilation duration from randomization to day 28, Sequential Organ Failure Assessment (SOFA) Score evaluation at 48h, 72h and 7 days and ICU-free days within 28. ETHICS AND DISSEMINATION: This trial was approved by the Brazilian National Committee of Ethics in Research (Comissão Nacional de Ética em Pesquisa - CONEP) and National Health Surveillance Agency (ANVISA). An independent data monitoring committee will perform interim analyses and evaluate adverse events throughout the trial. Results will be submitted for publication after enrolment and follow-up are complete.
Article
Full-text available
Background Skin aging is characterized by slacking and loss of density, especially under ultraviolet (UV) radiation exposure. Objective To investigate the beneficial effects of a combination containing bakuchiol (BK) and vanilla tahitensis extract (VTE) to prevent skin photoaging in vitro and to improve clinical outcomes for naturally aged skin. Materials and Methods Human dermal fibroblasts were treated with active compounds, exposed to an acute dose of UVA and analyzed by confocal microscopy: actin network for morphology, interleukin-8 (IL-8) for inflammation and p16 for senescence. Human skin was used to evaluate chronic UVA-induced glycosaminoglycan (GAG) loss and to assess the benefit of topical application of a BK+VTE serum (Alcian blue staining). An open-label clinical trial was conducted in women applying the serum twice daily for 56 days (n=43). Skin remodeling was assessed by FaceScan®. Firmness was evaluated through Dynaskin® and clinical scoring. Skin radiance was also rated on standardized full-face photographs. Results UVA induced a significant increase in IL-8 and p16 expression and marked morphological changes in fibroblasts. Treatment with BK or VTE alone prevented both actin network alteration and IL-8 upregulation. Interestingly, BK+VTE demonstrated synergistic protection against IL-8 and p16 overexpression. Serum application prevented GAG loss at the dermo-epidermal junction and increased dermal GAG in UVA-exposed skin explants. In the clinical trial, face ptosis was reduced by 11% on average for 26 responsive subjects and up to 23%. Depth of skin deformation was also reduced by 24% on average for 30 responsive subjects and up to 30%. This firming effect was confirmed by clinical scoring. Radiance was significantly improved by 29% on average for 33 responsive subjects. The serum demonstrated good tolerance/safety. Conclusion BK+VTE combination demonstrated anti-aging efficacy and might provide a substantial benefit in the daily care of naturally aged skin in women, through their synergistic effect on inflammaging and senescence.
Article
Full-text available
Shewanella oneidensis MR-1 is quickly becoming a synthetic biology workhorse for bioelectrochemical technologies due to a high level of understanding of its interaction with electrodes. Transmembrane electron transfer via the Mtr pathway has been well characterized, however, the role of NADH dehydrogenases in feeding electrons to Mtr has been only minimally studied in S. oneidensis MR-1. Four NADH dehydrogenases are encoded in the genome, suggesting significant metabolic flexibility in oxidizing NADH under a variety of conditions. A strain lacking the two dehydrogenases essential for aerobic growth exhibited a severe growth defect with an anode (+0.4 VSHE) or Fe(III)-NTA as the terminal electron acceptor. Our study reveals that the same NADH dehydrogenase complexes are utilized under oxic conditions or with a high potential anode. Our study also supports the previously indicated importance of pyruvate dehydrogenase activity in producing NADH during anerobic lactate metabolism. Understanding the role of NADH in extracellular electron transfer may help improve biosensors and give insight into other applications for bioelectrochemical systems.
Article
BACKGROUND: Although several therapeutic agents have been evaluated for the treatment of coronavirus disease 2019 (Covid-19), none have yet been shown to be efficacious. METHODS: We conducted a double-blind, randomized, placebo-controlled trial of intravenous remdesivir in adults hospitalized with Covid-19 with evidence of lower respiratory tract involvement. Patients were randomly assigned to receive either remdesivir (200 mg loading dose on day 1, followed by 100 mg daily for up to 9 additional days) or placebo for up to 10 days. The primary outcome was the time to recovery, defined by either discharge from the hospital or hospitalization for infection-control purposes only. RESULTS: A total of 1063 patients underwent randomization. The data and safety monitoring board recommended early unblinding of the results on the basis of findings from an analysis that showed shortened time to recovery in the remdesivir group. Preliminary results from the 1059 patients (538 assigned to remdesivir and 521 to placebo) with data available after randomization indicated that those who received remdesivir had a median recovery time of 11 days (95% confidence interval [CI], 9 to 12), as compared with 15 days (95% CI, 13 to 19) in those who received placebo (rate ratio for recovery, 1.32; 95% CI, 1.12 to 1.55; P < 0.001). The Kaplan-Meier estimates of mortality by 14 days were 7.1% with remdesivir and 11.9% with placebo (hazard ratio for death, 0.70; 95% CI, 0.47 to 1.04). Serious adverse events were reported for 114 of the 541 patients in the remdesivir group who underwent randomization (21.1%) and 141 of the 522 patients in the placebo group who underwent randomization (27.0%). CONCLUSIONS: Remdesivir was superior to placebo in shortening the time to recovery in adults hospitalized with Covid-19 and evidence of lower respiratory tract infection. (Funded by the National Institute of Allergy and Infectious Diseases and others; ACCT-1 ClinicalTrials.gov number, NCT04280705.).
Article
Background Although several therapeutic agents have been evaluated for the treatment of coronavirus disease 2019 (Covid-19), none have yet been shown to be efficacious. Methods We conducted a double-blind, randomized, placebo-controlled trial of intravenous remdesivir in adults hospitalized with Covid-19 with evidence of lower respiratory tract involvement. Patients were randomly assigned to receive either remdesivir (200 mg loading dose on day 1, followed by 100 mg daily for up to 9 additional days) or placebo for up to 10 days. The primary outcome was the time to recovery, defined by either discharge from the hospital or hospitalization for infection-control purposes only. Results A total of 1063 patients underwent randomization. The data and safety monitoring board recommended early unblinding of the results on the basis of findings from an analysis that showed shortened time to recovery in the remdesivir group. Preliminary results from the 1059 patients (538 assigned to remdesivir and 521 to placebo) with data available after randomization indicated that those who received remdesivir had a median recovery time of 11 days (95% confidence interval [CI], 9 to 12), as compared with 15 days (95% CI, 13 to 19) in those who received placebo (rate ratio for recovery, 1.32; 95% CI, 1.12 to 1.55; P<0.001). The Kaplan-Meier estimates of mortality by 14 days were 7.1% with remdesivir and 11.9% with placebo (hazard ratio for death, 0.70; 95% CI, 0.47 to 1.04). Serious adverse events were reported for 114 of the 541 patients in the remdesivir group who underwent randomization (21.1%) and 141 of the 522 patients in the placebo group who underwent randomization (27.0%). Conclusions Remdesivir was superior to placebo in shortening the time to recovery in adults hospitalized with Covid-19 and evidence of lower respiratory tract infection. (Funded by the National Institute of Allergy and Infectious Diseases and others; ACCT-1 ClinicalTrials.gov number, NCT04280705.)
Article
This study aimed to develop a microemulsion using PEG-6 Caprylic/Capric Glycerides as a surfactant to enhance the dermal delivery of celecoxib. Confocal laser scanning microscopy (CLSM) using the colocalization technique was also used to investigate the skin penetration pathway of the microemulsion. The prepared microemulsion formulations were characterized in terms of size, surface charge, size distribution and type. The celecoxib-loaded microemulsion had particle sizes ranging from 48-214 nm with neutral charge and significantly increased the skin penetration of celecoxib. According to the CLSM study, the microemulsion might attach to any part of the skin before releasing the entrapped drug to penetrate the tissue. The transfollicular pathway might be the major skin penetration pathway for the microemulsion, whereas the intercellular and transcellular pathways are minor ones.
Article
The idea of increasing the performance of sunscreens without adding more UV-filters is very attractive. Early studies reported an influence of solvents on the absorbing properties of UV-absorbers which was shown to be connected to the solvent polarity. However, the polarity differed a lot between tested solvents and most were unsuitable UV-filter solubilizers. The aim of the present study was to focus exclusively on emollients pertinent for sunscreens and investigate their impact on the performance of UV-filter combinations. The UV absorbance of Bis-ethylhexyloxyphenol Methoxyphenyl Triazine, Ethylhexyl Triazone, Diethylamino Hydroxybenzoyl Hexyl Benzoate, and Ethylhexyl Methoxycinnamate was measured in suncare relevant emollients comprising C12-15 Alkyl Benzoate, Dibutyl Adipate, Caprylic/Capric Triglyceride, Coco-caprylate, Isopropyl Myristate, Dicaprylyl Carbonate. The wavelength of maximum absorbance (λmax) and specific extinction at λmax (E1,1 (λmax)) were assessed for each UV-filter - emollient system. The performance of market relevant UV-filter combinations based on the studied UV-filters was simulated for each emollient with a computational method using the absorbance values measured for each UV-filter - emollient system. The difference in polarity of emollients led to a 2-3 nm bathochromic shift and a variation of the E1,1 (λmax) ranging from 4 to 20% for tested UV-filters. The emollient type showed nearly no influence on the sun protection factor (SPF) of market relevant UV-filter combinations probably due to a different influence an emollient shows on the UVB filters resulting in cancelling of the corresponding effect. Conversely, for all UV-filter combinations the UVA protection decreased with a decrease in the emollient polarity. Whilst the SPF was not impacted by standardly used cosmetic oils, the results advocate to use polar emollients to optimize the UVA protection. This is of advantage since polar emollients better dissolve crystalline UV-filters. From tested emollients, Dibutyl Adipate performed the best for both SPF and PPD factors.
Article
Role of lipid peroxidation products, particularly 4-hydroxynonenal (4-HNE) in cell cycle signaling is becoming increasingly clear. In this article, recent studies suggesting an important role of 4-HNE in stress mediated signaling for apoptosis are critically evaluated. Evidence demonstrating the modulation of UV, oxidative stress, and chemical stress mediated apoptosis by blocking lipid peroxidation by the alpha-class glutathione S-transferases (GSTs) is presented which suggest an important role of these enzymes in protection against oxidative stress and a role of lipid peroxidation products in stress mediated signaling. Overexpression of 4-HNE metabolizing GSTs (mGSTA4-4, hGSTA4-4, or hGST5.8) protects cells against 4-HNE, oxidative stress (H(2)O(2) or xanthine/xanthine oxidase), and UV-A mediated apoptosis by blocking JNK and caspase activation suggesting a role of 4-HNE in the mechanisms of apoptosis caused by these stress factors. The intracellular concentration of 4-HNE appears to be crucial for the nature of cell cycle signaling and may be a determinant for the signaling for differentiation, proliferation, transformation, or apoptosis. The intracellular concentrations of 4-HNE are regulated through a coordinated action of GSTs (GSTA4-4 and hGST5.8) which conjugate 4-HNE to GSH to form the conjugate (GS-HNE) and the transporter 76 kDa Ral-binding GTPase activating protein (RLIP76), which catalyze ATP-dependent transport of GS-HNE. A mild stress caused by heat, UV-A, or H(2)O(2)with no apparent effect on the cells in culture causes a rapid, transient induction of hGST5.8 and RLIP76. These stress preconditioned cells acquire ability to metabolize and exclude 4-HNE at an accelerated pace and acquire relative resistance to apoptosis by UV and oxidative stress as compared to unconditioned control cells. This resistance of stress preconditioned cells can be abrogated by coating the cells with anti-RLIP76 antibodies which block the transport of GS-HNE. These studies and previous reports discussed in this article strongly suggest a key role of 4-HNE in stress mediated signaling.