You are on page 1of 8

Review Article

Dermatology 2019;235:287–294 Received: December 13, 2018


Accepted after revision: March 25, 2019
DOI: 10.1159/000499858 Published online: May 21, 2019

Staphylococcus epidermidis:
A Potential New Player in the
Physiopathology of Acne?
Jean-Paul Claudel a Nicole Auffret b Marie-Thérèse Leccia c Florence Poli b
       

Stéphane Corvec d Brigitte Dréno e
   

a Private
Practice, Tours, France; b Private Practice, Paris, France; c Department of Dermatology, Allergology and
   

Photobiology, CHU A. Michallon, Grenoble, France; d CHU Nantes, Service de Bactériologie-Hygiène Hospitalière,
 

CRCINA, INSERM, U1232, Université de Nantes, Nantes, France; e UF dermato-cancérologie, CHU Nantes-Hôtel-Dieu,
 

Inserm U1232, CIC Biothérapie Inserm 05031, Nantes, France

Keywords Introduction
Acne vulgaris · Acne · Cutibacterium acnes · Dysbiosis ·
Microbiota · Pilosebaceous unit · Sebum · Staphylococcus Two different types of microorganisms are present on
epidermidis the skin and appendages. One of them consists of resident
microorganisms or commensals, which, most of the time,
are harmless and non-pathogenic, and sometimes may
Abstract even have beneficial effects for the host. The second group
Background: Cutibacterium acnes has been identified as one consists of transient microorganisms, which may be
of the main triggers of acne. However, increasing knowledge harmful and pathogenic and colonise the skin after injury
of the human skin microbiome raises questions about the or trauma, and lead to infections and inflammation [1–3].
role of other skin commensals, such as Staphylococcus epi- With the availability of bioinformatic tools leading to
dermidis, in the physiopathology of this skin disease. Sum- new phylogenetic approaches, important advances have
mary: This review provides an overview of current knowl- been made over the last years either to investigate the di-
edge of the potential role of S. epidermidis in the physiopa- versity and the topology of skin microbiota or to identify
thology of acne. Recent research indicates that acne might the different commensal microorganisms present on the
be the result of an unbalanced equilibrium between C. acnes skin, to evaluate the relative abundance of each popula-
and S. epidermidis, according to dedicated interactions. Cur- tion, and to understand their beneficial role or contribu-
rent treatments act on C. acnes only. Other treatment op- tion to dermatological conditions such as acne [4–6].
tions may be considered, such as probiotics derived from S. Metagenomic analysis and sequencing of the 16S ribo-
epidermidis to restore the naturally balanced microbiota or somal RNA gene are the predominant bacteriological
through targeting the regulation of the host’s AMP media- sampling methods employed for analysing the bacterial
tors. Key Messages: Research seems to confirm the benefi- composition of microbial communities, and choosing the
cial role of S. epidermidis in acne by limiting C. acnes over- most effective study design is crucial for obtaining mean-
colonisation and inflammation. © 2019 S. Karger AG, Basel ingful analysis results [7]. Thus, physiological character-

© 2019 S. Karger AG, Basel Brigitte Dréno


UF dermato-cancérologie, CHU Nantes-Hôtel-Dieu
Inserm U1232, CIC Biothérapie Inserm 05031
E-Mail karger@karger.com
Place Alexis Ricordeau, FR–44093 Nantes Cedex 01 (France)
www.karger.com/drm E-Mail brigitte.dreno @ atlanmed.fr
AMPs TLRs PAR
AMPs
TLRs
C. acnes

S. epidermidis

Corynebacterium

Other commensal
bacteria

S. aureus

M. furfur
a b

Fig. 1. Healthy skin and skin with acne lesion in formation. a nisation of C. acnes during puberty leads to dysbiosis and acne.
Healthy skin. In healthy skin S. epidermidis controls the prolifera- AMP, antimicrobial peptides; TLR, Toll-like receptors; PAR, pro-
tion of C. acnes. b Formation of a microcomedone after over-col- tease-activated receptors.
onisation of the skin by C. acnes leading to dysbiosis. Over-colo-

istics of different skin sites sampled have been associated Results


with different levels of bacterial diversity including Acti-
nobacteria such as Propionibacteria, very recently re- The Interplay of Host Skin and the Cutaneous
named Cutibacteria (comprising Cutibacterium acnes Microbiota
and also Cutibacterium granulosum and Cutibacterium The interplay of host skin and the cutaneous microbi-
avidum), Proteobacteria, Bacteroides and Firmicutes; ota in the skin’s immune system is able to differentiate
Staphylococcus epidermidis belongs to the latter group [5, between harmless commensal microorganisms including
6, 8, 9]. Of these, C. acnes and S. epidermidis are two of Corynebacterium sp., Staphylococcus sp. excluding S. au-
the main commensal skin bacteria [3, 10–12]. Until very reus, Cutibacterium sp., Malassezia furfur, and transient,
recently, research in acne has mainly focused on the role harmful pathogenic microorganisms such as S. aureus,
of C. acnes while the role of S. epidermidis in acne has Streptococcus pyogenes, and Enterobacteriaceae. Even
been discussed for several years but still remains to be though it seems as if this recognition involves the host’s
elucidated [10, 13]. immune response, this mechanism is not fully under-
The aim of the present article is to review published stood. Toll-like receptors (TLRs) have been reported to be
data concerning the role of S. epidermidis in the physio- desensitised through prolonged exposure to commensal
pathology of acne and to present future treatment per- microorganisms, either through a decreased expression of
spectives based on available data and evidence. TLR on the cell surface or by activation of the TLR path-
way inhibitors interleukin-1 receptor-associated kinase 3
(IRAK3) and the suppressor of cytokine signalling 1
Methodology (SOCS1) [14, 15]. Moreover, specificity may be achieved
by a combined recognition of pathogen-associated mo-
A review of the literature published between the beginning of lecular patterns through pattern recognition receptors [6].
2000 and 2018 and available from PubMed was carried out using Thus, both such recognised commensals C. acnes and
the following key words: acne, acne vulgaris + microbiota, skin +
microbiota, acne + microbiota + Propionibacterium acnes + Staph- S. epidermidis seem to interact with the host, helping to
ylococcus epidermidis, bacterial resistance, skin commensal + Pro- protect the healthy skin from colonisation by pathogens
pionibacterium acnes + Staphylococcus epidermidis. [9, 16]. Figure 1 provides visual support.

288 Dermatology 2019;235:287–294 Claudel/Auffret/Leccia/Poli/Corvec/


DOI: 10.1159/000499858 Dréno
However, the composition of the cutaneous microbi- comedones, papules, and pustules [31–34, 38]. Figure 1
ota evolves constantly and changes over time. As an ex- shows the difference between healthy microbiome and
ample, a disturbed balance or dysbiosis of previously microbiome suffering from dysbiosis.
healthy skin caused by exogenous factors such as injury,
stress, or pollution, or endogenous factors (hormonal The Role of S. epidermidis
changes, changes of pH) may induce inflammatory skin S. epidermidis is the most frequently isolated commen-
disorders such as acne, atopic dermatitis, rosacea, and sal species from human epithelia [25, 39]. It colonises pre-
psoriasis [13, 17–21]. Specifically, during puberty, the in- dominantly axillae, the head, and nares, and has gener-
crease and qualitative modification of sebum due to hor- ally a non-pathogenic relationship with its host [11, 39].
monal modifications may lead to an unbalanced skin mi- S. epidermidis belongs to the group of coagulase-neg-
crobiota via an increased colonisation of the piloseba- ative staphylococci, which differs from coagulase-posi-
ceous unit and of the skin surface by C. acnes [6]. This tive staphylococci such as S. aureus by lacking the enzyme
over-representation may be due to a selective advantage coagulase. According to multi-locus sequence typing, the
linked to the nutritive substance availability [22]. species showed a high degree of diversity with more than
400 identified sequence types (STs) compared to 155 for
The Role of C. acnes C. acnes [40, 41]. Most clinical isolates belong to CC2,
In healthy skin, C. acnes plays a beneficial role in the which comprises the most frequently isolated ST2. Pos-
cutaneous microbiota of the pilosebaceous unit. It limits sibly, the successful spread of ST2 may be due to the fact
the growth of S. aureus, such as community-acquired that all ST2 isolates contain IS256 insertion sequences
methicillin-resistant S. aureus, as well as that of S. pyo- and ica genes, two factors found to be correlated with S.
genes in maintaining an acidic pH of the pilosebaceous epidermidis invasiveness [42, 43]. Nevertheless, contro-
follicle by hydrolysing sebum triglycerides and by secret- versy still exists about the fact that icaA is a useful marker
ing propionic acid [6, 23, 24]. for S. epidermidis biofilm [44].
However, during puberty, over-colonisation of the pi- In the past, S. epidermidis has been considered an in-
losebaceous unit by C. acnes may lead to a loss of diversi- nocuous commensal microorganism on the human skin.
fication and dysbiosis, potentially causing acne [6, 25– Nowadays it is seen as an important opportunistic patho-
27]. A recent clinical study using the single-locus se- gen. It is the most frequent cause of nosocomial infec-
quence typing method investigated C. acnes subgroups tions, at a rate about as high as that due to its more viru-
on the face and back in patients with severe acne and in lent cousin S. aureus. In particular, S. epidermidis repre-
healthy subjects [28]. In almost 75% of the acne patients, sents the most common source of infection on indwelling
C. acnes phylotypes were identical on the face and back, medical devices [45].
whereas this was only the case in about 45% of the healthy Treatment is complicated by specific antibiotic-resis-
subjects. In the healthy group, phylotypes IA1 (39%) and tant genes and the formation of biofilms, multicellular
II (43%) were the main phylotypes, whereas in the acne agglomerations that have intrinsic resistance and toler-
group IA1 (84%), especially on the back (96%), was the ance to antibiotics and mechanisms of host defence [46].
main phylotype. This may confirm the hypothesis that Furthermore, investigations have identified specific mo-
acne severity may be associated with a loss of diversity of lecular determinants allowing S. epidermidis immune in-
C. acnes phylotypes, following a selection of phylotype vasion and its ability to cause chronic diseases [47]. These
IA1/clonal complex (CC) 18 present in all acne patients causes are believed to have original functions in the non-
[28–30]. infectious lifestyle of this microorganism, emphasising
Therefore, different inflammatory profiles, depending the accidental nature of S. epidermidis infections allowing
on the phylotype (i.e., phylotype IA1, which has been differentiating between portage, contamination, and in-
mainly observed on the face and back of acne patients and fection. A better understanding of the physiology of S.
cluster of C. acnes activating the innate immunity via the epidermidis is important to evaluate therapeutic strate-
expression of protease-activated receptors (PARs), tu- gies against S. epidermidis infections [39].
mour necrosis factor-α, and the production of interferon-γ Certain strains of S. epidermidis may modulate the
and interleukins [IL-8], have been observed [28, 31–37]. host innate immune response, especially of TLR 2, and
Moreover, C. acnes activates the release of lipases, matrix thus allow the host to fight against pathogens [6, 28]. Phe-
metalloproteinases, and hyaluronidases, leading to hy- nol-soluble modulins produced by S. epidermidis were
perkeratinisation of the pilosebaceous unit and finally to shown to selectively inhibit skin pathogens, such as S. au-

Staphylococcus epidermidis and Acne Dermatology 2019;235:287–294 289


DOI: 10.1159/000499858
reus and Group A Streptococcus, and, together with host lated from acne lesions and healthy skin, respectively, were
antimicrobial peptides (AMPs), enhanced their elimina- not significantly different in their susceptibility patterns
tion [48–50]. Moreover, S. epidermidis activates the AMP towards S. epidermidis. Conversely, the study showed a
expression of keratinocytes through a TLR2-dependent higher frequency of antimicrobial activity among certain
mechanism [51]. This confirms that this skin commensal C. acnes strains, such as I-2 phylogroup, against S. epider-
interacts closely with the host innate immune mechanism midis. The authors hypothesised a likely possible presence
[48, 49]. This was also shown through the microdissec- and secretion of a bacteriocin or bacteriocin-like substance
tion of epidermal compartments of the dermis and of the specific to C. acnes phylogroup I-2 [13]. C. acnes strains
adipose tissue followed by 16S ribosomal RNA sequenc- and their antimicrobial activities against S. epidermidis
ing demonstrating that colonisation with S. epidermidis have also been investigated. However, regarding the skin
tunes T-cell homing and function in an IL-1-dependent status origin (healthy, moderate, or severe acne), the anti-
manner [52, 53]. microbial activity ranged from 29 to 39%. Consequently,
Other very recent research work reported that S. epi- type IA1 CC18 C. acnes strains largely involved in acne did
dermidis may also protect against skin neoplasia through not present higher activity compared to healthy strains
the production of 6-N-hydroxyaminopurine, a molecule [13].
that inhibits DNA polymerase activity [54]. Moreover, Different antagonism investigations demonstrated
butyric acid, released by S. epidermidis, allows adipose- that, in vivo, S. epidermidis controls the proliferation of
derived stem cells to differentiate into adipocytes and lip- C. acnes via the release of succinic acid, a fatty acid fer-
id accumulation in the cytoplasm, resulting in an in- mentation product, which inhibits surface TLRs of kera-
creased dermal layer [55]. tinocytes and tumour necrosis factor and suppresses C.
acnes-induced IL-6 [27, 56, 58].
The Interplay between S. epidermidis and C. acnes in With both C. acnes and S. epidermidis being present on
Acne the skin, the inhibition of C. acnes-induced inflammation
S. epidermidis and C. acnes use glycerol as a shared car- by S. epidermidis may potentially be dependent on staph-
bon source to produce different short-chain fatty acids ylococcal LTA-induced miR-143 on keratinocytes,
(SCFAs) used as antimicrobial agents to compete against known for limiting inflammation. Research suggested
each other. They are both present in acne lesions. This that the mechanism for LTA-miR-143-mediated sup-
concomitant presence has raised questions about their re- pression of TLR2 signalling is accomplished by miR-143
spective role in the pathogenesis of acne [56, 57]. While targeting the 3′UTR of TLR2. It thereby decreases the
there is no evidence that S. epidermidis plays an active role TLR2 protein production, which plays a major role in the
in the onset of acne, C. acnes is currently associated with inhibition of C. acnes-induced cutaneous inflammation
acne [9]. [58–60]. Thus, it helps to regulate skin homeostasis and
Some S. epidermidis isolates have been shown through to suppress the pathogenic inflammation that is induced
an antagonism assay to possess antimicrobial activity by C. acnes [58, 61].
against C. acnes [27, 56]. Among the S. epidermidis strains Accordingly, an unbalanced equilibrium between C.
with an elevated antimicrobial activity, differences in inhi- acnes and S. epidermidis in pilosebaceous units of acne
bition zone diameter and appearance were observed, indi- patients in favour of phylotype IA1 CC18 C. acnes strains
cating that the antimicrobial substances were of a different (75–80 of cases) may not allow S. epidermidis to fully play
nature. Most of the S. epidermidis strains displayed small its role as a regulator of the natural skin homeostasis in
zones of inhibition (2–4 mm) against C. acnes and some limiting the growth of C. acnes. Figure 2 shows visually
strains produced opaque zones of inhibition. One strain, the interplay between C. acnes and S. epidermidis.
FS1, produced very large inhibitory zones (>10 mm), but
was not active against all C. acnes strains tested. Another S. epidermidis and its Implication in the Choice of
strain, 14.1.R1, inhibited all C. acnes strains, but produced Current and Future Acne Treatments
only small zones of inhibition (2–5 mm). No difference in Current topical acne treatments principally include
prevalence and intensity was noted in the antimicrobial different retinoids, such as adapalene and tretinoin,
activity of S. epidermidis strains isolated from normal and which reduce inflammation by modulating the innate im-
acne-affected skin, respectively. Likewise, the origin of C. munity activated by C. acnes [62–65]. Apart from ben-
acnes strains did not determine their susceptibility to the zoyl peroxide, the use of topical antibacterials such as
antimicrobial activity of S. epidermidis, since strains iso- erythromycin and clindamycin in monotherapy resulted

290 Dermatology 2019;235:287–294 Claudel/Auffret/Leccia/Poli/Corvec/


DOI: 10.1159/000499858 Dréno
Dysbiosis Balanced microbiome

C. acnes releases
coproporhyrin III S. epidermidis inhibits
promoting S. aureus biofilm
S. aureus
X X C. acnes inhibits proliferation of
biofilm X
X X X S. epidermidis through hydrolyzing
S. aureus sebum triglycerides and release
biofilm of propionic acid

S. epidermidis inhibits proliferation


of C. acnes through glycerol and
succinic acid

S. epidermidis induces keratinocytes


to produce AMPs to kill S. aureus

CoproporphyrinIII
Antimicrobial peptides
C. acnes
S. epidermidis
S. aureus

Fig. 2. Bacterial interactions on the skin. AMP, antimicrobial peptides.

in the development of antibiotic-resistant strains of not spectrum of Gram-positive and Gram-negative bacteria,
only C. acnes but also of S. epidermidis in 4–6 weeks [30, as well as some fungi, parasites, and enveloped viruses
57, 66]. Systemic antibiotics have been reported to cause [70]. They are produced as a response by keratinocytes
only little resistance but can lead to collateral damage in and sebocytes. However, they also promote additional
gut microbiota [30, 33, 67]. Oral isotretinoin normalises inflammatory responses. The production of AMPs com-
the response of the innate immune system to C. acnes by prises ß-defensins, RNase 7, the S100-protein psoriasin,
inhibiting its proliferation [68]. and cathelicidins, and is mediated by the MyD88 pathway
Thus, eliminating only C. acnes may favour the prolif- and IL-1 signalling. In addition, it has been demonstrated
eration of S. aureus, triggering inflammatory acne flare- that more cells express TLR-2 as acne severity increases
up and proliferation of S. epidermidis, leading to another [33]. This may be one explanation why agents that target
unbalanced skin homeostasis and a risk of nosocomial TLR-2, such as topical retinoids, have a greater efficacy in
infections. patients with more severe acne [71]. Cytokines are also
AMPs are effector molecules of the innate immune produced as a result of the interaction between C. acnes
system of the skin. They are amphipathic and disrupt the and TLR-2, defensins, and matrix metalloproteinase via
lipid membrane of the bacterium, leading to cell lysis and PAR-2R activation [72].
death by interacting preferentially with negatively charged Another treatment approach could be the regular oral
bacterial membranes rather than with neutrally charged or topical supplementation of the skin microbiota with a
mammalian cell membranes. The importance of the beneficial microorganism (probiotics) for patients with
AMPs’ contribution to host immunity is undeniable as acne by re-equilibrating the cutaneous microbiota [73,
alterations in AMP expression have been associated with 74]. In 2010, Arck et al. [75] suggested that not only a
various pathologic processes. Nevertheless, data regard- brain-gut or brain-skin axis, but also a gut-skin axis ex-
ing the role of AMPs in acne vulgaris are limited. A re- ists. The authors showed that oral supplementation of a
cently published study reports that the AMPs hBD-1 and Lactobacillus strain in mice dampens stress-induced neu-
cathelicidins play an important role in the pathogenesis rogenic skin inflammation and hair growth inhibition.
of acne [69]. AMPs have shown activity against a broad Their concept suggested that modulation of the micro-

Staphylococcus epidermidis and Acne Dermatology 2019;235:287–294 291


DOI: 10.1159/000499858
biota by deployment of probiotics reduces stress-induced tion [9, 33]. However, disequilibrium in favour of S. epi-
neurogenic skin inflammation. These observations raise dermidis may also result in other health consequences
hope that the right kind of bacteria may have beneficial such as nosocomial infections. Therefore, a balanced skin
effects on the skin homoeostasis, skin inflammation, and homeostasis should be the final aim in any acne treatment.
peripheral tissue responses to perceived stress. Other In this perspective, other treatment options may be
studies conducted in humans confirmed this hypothesis considered, such as probiotics derived from S. epidermi-
[76–78]. Thus, probiotics may be efficient in acne and dis, allowing for a restoration of the naturally balanced
other inflammatory skin diseases such as atopic dermati- microbiota, and through the targeting of the regulation of
tis and potentially psoriasis [79–83]. the host’s AMP mediators, without increasing the local S.
In this perspective, studies indicate that S. epidermidis epidermidis population.
may control dysbiosis caused by C. acnes, thus reducing
acne severity [27, 56]. However, it still remains unclear
which SCFA in the products of S. epidermidis glycerol fer- Key Message
mentation primarily contributes to the anti-C. acnes ef-
S. epidermidis limits C. acnes-induced colonisation of the skin,
fect. It is also undetermined whether SCFAs act together thus potentially playing a role in the physiopathology of acne.
with other antimicrobial molecules in fermentation prod-
ucts to display their anti-C. acnes activities. The anti-C.
acnes activity of the fermented media persisted after boil- Acknowledgements
ing the said media, thereby suggesting that the antimicro-
bial proteins/peptides may not be the major contributors The authors acknowledge the writing support of Patrick Göritz,
to the anti-C. acnes activity of fermented media [56]. SMWS-Scientific, and Medical Writing Services.
Pro-drugs such as the SCFAs pivaloylomethyl butyrate
(AN-9) have been developed to achieve pharmacologic
concentrations of such SCFAs in vivo [84]. Moreover, live Statement of Ethics
S. epidermidis may potentially be used as an active com- The authors have no ethical conflicts to disclose
ponent in acne probiotics [27, 56].

Disclosure Statement
Discussion and Conclusion
The authors have no financial interest to disclose.
Acne is a chronic and multifactorial inflammatory dis-
ease of the skin and the pilosebaceous unit. Dysbiosis in
acne patients is associated with a decreased number of S. Funding Sources
epidermidis and an over-colonisation by selected C. acnes
phylotypes in the sebaceous unit, leading to different lev- This Expert Group was organised by Galderma International,
France.
els of activation of the innate immunity, thus resulting in
different severity levels of inflammatory acne. Recent re-
search seems to confirm the beneficial role of S. epidermi- Author Contributions
dis in the physiopathology of acne through limiting C.
acnes-induced colonisation of the skin and of inflamma- All cited authors contributed equally to this paper.

References
  1 Kong HH, Segre JA. Skin microbiome: look- gram. Temporal Stability of the Human   5 Grice EA, Kong HH, Conlan S, Deming CB,
ing back to move forward. J Invest Dermatol. Skin  Microbiome. Cell. 2016 May; 165(4): Davis J, Young AC, et al.; NISC Comparative
2012 Mar;132(3 Pt 2):933–9. 854–66. Sequencing Program. Topographical and
  2 Cogen AL, Nizet V, Gallo RL. Skin microbio-   4 Peterson J, Garges S, Giovanni M, McInnes P, temporal diversity of the human skin micro-
ta: a source of disease or defence? Br J Derma- Wang L, Schloss JA, et al.; NIH HMP Work- biome. Science. 2009 May; 324(5931): 1190–
tol. 2008 Mar;158(3):442–55. ing Group. The NIH Human Microbiome 2.
 3 Oh J, Byrd AL, Park M, Kong HH, Segre Project. Genome Res. 2009 Dec;19(12):2317–   6 Grice EA, Segre JA. The skin microbiome. Nat
JA;  NISC Comparative Sequencing Pro- 23. Rev Microbiol. 2011 Apr;9(4):244–53.

292 Dermatology 2019;235:287–294 Claudel/Auffret/Leccia/Poli/Corvec/


DOI: 10.1159/000499858 Dréno
  7 Meisel JS, Hannigan GD, Tyldsley AS, San- acnes, a commensal bacterium in the human fensin-2 and interleukin-8 expression in hu-
Miguel AJ, Hodkinson BP, Zheng Q, et al. skin microbiome, as skin probiotics against man keratinocytes through toll-like recep-
Skin Microbiome Surveys Are Strongly Influ- methicillin-resistant Staphylococcus aureus. tors. J Invest Dermatol. 2005 May; 124(5):
enced by Experimental Design. J Invest Der- PLoS One. 2013;8(2):e55380. 931–8.
matol. 2016 May;136(5):947–56. 24 Tax G, Urbán E, Palotás Z, Puskás R, Kónya 37 Jasson F, Nagy I, Knol AC, Zuliani T, Kham-
  8 Scholz CF, Kilian M. The natural history of Z, Bíró T, et al. Propionic Acid Produced by mari A, Dréno B. Different strains of Propi-
cutaneous propionibacteria, and reclassifica- Propionibacterium acnes Strains Contri- onibacterium acnes modulate differently the
tion of selected species within the genus Pro- butes to Their Pathogenicity. Acta Derm Ve- cutaneous innate immunity. Exp Dermatol.
pionibacterium to the proposed novel genera nereol. 2016 Jan;96(1):43–9. 2013 Sep;22(9):587–92.
Acidipropionibacterium gen. nov., Cutibac- 25 Bek-Thomsen M, Lomholt HB, Kilian M. 38 Dessinioti C, Antoniou C, Katsambas A. Ac-
terium gen. nov. and Pseudopropionibacte- Acne is not associated with yet-uncultured neiform eruptions. Clin Dermatol. 2014 Jan-
rium gen. nov. Int J Syst Evol Microbiol. 2016 bacteria. J Clin Microbiol. 2008 Oct; 46(10): Feb;32(1):24–34.
Nov;66(11):4422–32. 3355–60. 39 Otto M. Staphylococcus epidermidis—the
  9 Byrd AL, Belkaid Y, Segre JA. The human skin 26 Di Domizio J, Pagnoni A, Huber M, Hohl D, ‘accidental’ pathogen. Nat Rev Microbiol.
microbiome. Nat Rev Microbiol. 2018 Mar; Gilliet M. [The skin microbiota: a colossus 2009 Aug;7(8):555–67.
16(3):143–55. steps into the spotlight]. Rev Med Suisse. 2016 40 Multi Locus Sequence Typing. Staphylococ-
10 Coates R, Moran J, Horsburgh MJ. Staphylo- Mar;12(512):660–4. cus epidermidis. 2018. Available from: http://
cocci: colonizers and pathogens of human 27 Wang Y, Kao MS, Yu J, Huang S, Marito S, sepidermidis.mlst.net/.
skin. Future Microbiol. 2014;9(1):75–91. Gallo RL, et al. A Precision Microbiome Ap- 41 Méric G, Mageiros L, Pensar J, Laabei M, Ya-
11 Christensen GJ, Brüggemann H. Bacterial proach Using Sucrose for Selective Augmen- hara K, Pascoe B, et al. Disease-associated
skin commensals and their role as host guard- tation of Staphylococcus epidermidis Fer- genotypes of the commensal skin bacterium
ians. Benef Microbes. 2014 Jun;5(2):201–15. mentation against Propionibacterium acnes. Staphylococcus epidermidis. Nat Commun.
12 Fischbach MA, Segre JA. Signaling in Host- Int J Mol Sci. 2016 Nov;17(11):E1870. 2018 Nov;9(1):5034.
Associated Microbial Communities. Cell. 28 Dagnelie MA, Corvec S, Saint-Jean M, 42 Li M, Wang X, Gao Q, Lu Y. Molecular char-
2016 Mar;164(6):1288–300. Bourdès V, Nguyen JM, Khammari A, et al. acterization of Staphylococcus epidermidis
13 Christensen GJ, Scholz CF, Enghild J, Rohde Decrease in Diversity of Propionibacterium strains isolated from a teaching hospital in
H, Kilian M, Thürmer A, et al. Antagonism acnes Phylotypes in Patients with Severe Acne Shanghai, China. J Med Microbiol. 2009 Apr;
between Staphylococcus epidermidis and on the Back. Acta Derm Venereol. 2018 Feb; 58(Pt 4):456–61.
Propionibacterium acnes and its genomic ba- 98(2):262–7. 43 Yao Y, Sturdevant DE, Otto M. Genomewide
sis. BMC Genomics. 2016 Feb;17(1):152. 29 Aubin GG, Portillo ME, Trampuz A, Corvec analysis of gene expression in Staphylococcus
14 Fukao T, Koyasu S. PI3K and negative regula- S. Propionibacterium acnes, an emerging epidermidis biofilms: insights into the patho-
tion of TLR signaling. Trends Immunol. 2003 pathogen: from acne to implant-infections, physiology of S. epidermidis biofilms and the
Jul;24(7):358–63. from phylotype to resistance. Med Mal Infect. role of phenol-soluble modulins in formation
15 Strober W. Epithelial cells pay a Toll for pro- 2014 Jun;44(6):241–50. of biofilms. J Infect Dis. 2005 Jan;191(2):289–
tection. Nat Med. 2004 Sep;10:898–900. 30 Nakase K, Nakaminami H, Takenaka Y, 98.
16 Burian M, Bitschar K, Dylus B, Peschel A, Hayashi N, Kawashima M, Noguchi N. Rela- 44 Frank KL, Hanssen AD, Patel R. icaA is not a
Schittek B. The Protective Effect of Microbio- tionship between the severity of acne vulgaris useful diagnostic marker for prosthetic joint
ta on S. aureus Skin Colonization Depends on and antimicrobial resistance of bacteria iso- infection. J Clin Microbiol. 2004 Oct; 42(10):
the Integrity of the Epithelial Barrier. J Invest lated from acne lesions in a hospital in Japan. 4846–9.
Dermatol. 2017 Apr;137(4):976–9. J Med Microbiol. 2014 May;63(Pt 5):721–8. 45 National Nosocomial Infections Surveillance
17 van Rensburg JJ, Lin H, Gao X, Toh E, Fortney 31 Zouboulis CC, Jourdan E, Picardo M. Acne is System. National Nosocomial Infections Sur-
KR, Ellinger S, et al. The Human Skin Micro- an inflammatory disease and alterations of se- veillance (NNIS) System Report, data sum-
biome Associates with the Outcome of and Is bum composition initiate acne lesions. J Eur mary from January 1992 through June 2004,
Influenced by Bacterial Infection. MBio. 2015 Acad Dermatol Venereol. 2014 May; 28(5): issued October 2004. Am J Infect Control.
Sep;6(5):e01315–15. 527–32. 2004 Dec;32(8):470–85.
18 Scanlan PD, Buckling A, Kong W, Wild Y, 32 Picardo M, Ottaviani M, Camera E, Mastro- 46 Costerton JW, Stewart PS, Greenberg EP.
Lynch SV, Harrison F. Gut dysbiosis in cystic francesco A. Sebaceous gland lipids. Derma- Bacterial biofilms: a common cause of persis-
fibrosis. J Cyst Fibros. 2012 Sep;11(5):454–5. toendocrinol. 2009 Mar;1(2):68–71. tent infections. Science. 1999 May;284(5418):
19 Zeeuwen PL, Kleerebezem M, Timmerman 33 Dreno B, Gollnick HP, Kang S, Thiboutot D, 1318–22.
HM, Schalkwijk J. Microbiome and skin dis- Bettoli V, Torres V, et al.; Global Alliance to 47 Nakamizo S, Egawa G, Honda T, Nakajima S,
eases. Curr Opin Allergy Clin Immunol. 2013 Improve Outcomes in Acne. Understanding Belkaid Y, Kabashima K. Commensal bacteria
Oct;13(5):514–20. innate immunity and inflammation in acne: and cutaneous immunity. Semin Immuno-
20 Picardo M, Ottaviani M. Skin microbiome implications for management. J Eur Acad pathol. 2015 Jan;37(1):73–80.
and skin disease: the example of rosacea. J Dermatol Venereol. 2015 Jun; 29 Suppl 4: 3– 48 Cogen AL, Yamasaki K, Sanchez KM,
Clin Gastroenterol. 2014 Nov-Dec; 48 Suppl 11. Dorschner RA, Lai Y, MacLeod DT, et al. Se-
1:S85–6. 34 Dréno B. What is new in the pathophysiology lective antimicrobial action is provided by
21 Prescott SL, Larcombe DL, Logan AC, West of acne, an overview. J Eur Acad Dermatol phenol-soluble modulins derived from Staph-
C, Burks W, Caraballo L, et al. The skin mi- Venereol. 2017 Sep;31 Suppl 5:8–12. ylococcus epidermidis, a normal resident of
crobiome: impact of modern environments 35 Yu Y, Champer J, Agak GW, Kao S, Modlin the skin. J Invest Dermatol. 2010 Jan; 130(1):
on skin ecology, barrier integrity, and system- RL, Kim J. Different Propionibacterium acnes 192–200.
ic immune programming. World Allergy Or- Phylotypes Induce Distinct Immune Re- 49 Cogen AL, Yamasaki K, Muto J, Sanchez KM,
gan J. 2017 Aug;10(1):29. sponses and Express Unique Surface and Se- Crotty Alexander L, Tanios J, et al. Staphylo-
22 McGinley KJ, Webster GF, Leyden JJ. Region- creted Proteomes. J Invest Dermatol. 2016 coccus epidermidis antimicrobial delta-toxin
al variations of cutaneous propionibacteria. Nov;136(11):2221–8. (phenol-soluble modulin-gamma) cooper-
Appl Environ Microbiol. 1978 Jan;35(1):62–6. 36 Nagy I, Pivarcsi A, Koreck A, Széll M, Urbán ates with host antimicrobial peptides to kill
23 Shu M, Wang Y, Yu J, Kuo S, Coda A, Jiang Y, E, Kemény L. Distinct strains of Propionibac- group A Streptococcus. PLoS One. 2010 Jan;
et al. Fermentation of Propionibacterium terium acnes induce selective human beta-de- 5(1):e8557.

Staphylococcus epidermidis and Acne Dermatology 2019;235:287–294 293


DOI: 10.1159/000499858
50 Laborel-Préneron E, Bianchi P, Boralevi F, coccus epidermidis increases antimicrobial acnes. Br J Dermatol. 2005 Dec;153(6):1105–
Lehours P, Fraysse F, Morice-Picard F, et al. defense against bacterial skin infections. J In- 13.
Effects of the Staphylococcus aureus and vest Dermatol. 2010 Sep;130(9):2211–21. 73 Zaenglein AL, Pathy AL, Schlosser BJ, Alikhan
Staphylococcus epidermidis Secretomes Iso- 61 Białecka A, Mak M, Biedroń R, Bobek M, A, Baldwin HE, Berson DS, et al. Guidelines
lated from the Skin Microbiota of Atopic Kasprowicz A, Marcinkiewicz J. Different of care for the management of acne vulgaris. J
Children on CD4+ T Cell Activation. PLoS pro-inflammatory and immunogenic poten- Am Acad Dermatol. 2016 May;74(5):945–73.
One. 2015 Oct;10(10):e0141067. tials of Propionibacterium acnes and Staphy- e33.
51 Lai Y, Di Nardo A, Nakatsuji T, Leichtle A, lococcus epidermidis: implications for chron- 74 Bowe W, Patel NB, Logan AC. Acne vulgaris,
Yang Y, Cogen AL, et al. Commensal bacteria ic inflammatory acne. Arch Immunol Ther probiotics and the gut-brain-skin axis: from
regulate Toll-like receptor 3-dependent in- Exp (Warsz). 2005 Jan-Feb;53(1):79–85. anecdote to translational medicine. Benef Mi-
flammation after skin injury. Nat Med. 2009 62 Michel S, Jomard A, Démarchez M. Pharma- crobes. 2014 Jun;5(2):185–99.
Dec;15(12):1377–82. cology of adapalene. Br J Dermatol. 1998 Oct; 75 Arck P, Handjiski B, Hagen E, Pincus M, Bru-
52 Naik S, Bouladoux N, Wilhelm C, Molloy MJ, 139 Suppl 52:3–7. enahl C, Bienenstock J, et al. Is there a ‘gut-
Salcedo R, Kastenmuller W, et al. Compart- 63 Tenaud I, Khammari A, Dreno B. In vitro brain-skin axis’? Exp Dermatol. 2010 May;
mentalized control of skin immunity by resi- modulation of TLR-2, CD1d and IL-10 by 19(5):401–5.
dent commensals. Science. 2012 Aug; adapalene on normal human skin and acne 76 Gueniche A, Philippe D, Bastien P, Reuteler
337(6098):1115–9. inflammatory lesions. Exp Dermatol. 2007 G, Blum S, Castiel-Higounenc I, et al. Ran-
53 Naik S, Bouladoux N, Linehan JL, Han SJ, Jun;16(6):500–6. domised double-blind placebo-controlled
Harrison OJ, Wilhelm C, et al. Commensal- 64 Ozolins M, Eady EA, Avery AJ, Cunliffe WJ, study of the effect of Lactobacillus paracasei
dendritic-cell interaction specifies a unique Po AL, O’Neill C, et al. Comparison of five NCC 2461 on skin reactivity. Benef Microbes.
protective skin immune signature. Nature. antimicrobial regimens for treatment of mild 2014 Jun:5(2):137-45.
2015 Apr;520(7545):104–8. to moderate inflammatory facial acne vulgar- 77 Gueniche A, Benyacoub J, Philippe D, Bastien
54 Nakatsuji T, Chen TH, Butcher AM, Trzoss is in the community: randomised controlled P, Kusy N, Breton L, et al. Lactobacillus para-
LL, Nam SJ, Shirakawa KT, et al. A commen- trial. Lancet. 2004 Dec;364(9452):2188–95. casei CNCM I-2116 (ST11) inhibits substance
sal strain of Staphylococcus epidermidis pro- 65 Fulton JE Jr, Farzad-Bakshandeh A, Bradley P-induced skin inflammation and accelerates
tects against skin neoplasia. Sci Adv. 2018 S. Studies on the mechanism of action to top- skin barrier function recovery in vitro. Eur J
Feb;4(2):eaao4502. ical benzoyl peroxide and vitamin A acid in Dermatol. 2010 Nov-Dec;20(6):731–7.
55 Wang Y, Zhang L, Yu J, Huang S, Wang Z, acne vulgaris. J Cutan Pathol. 1974;1(5):191– 78 Fabbrocini G, Bertona M, Picazo Ó, Pareja-
Chun KA, et al. A Co-Drug of Butyric Acid 200. Galeano H, Monfrecola G, Emanuele E. Sup-
Derived from Fermentation Metabolites of 66 Moon SH, Roh HS, Kim YH, Kim JE, Ko JY, plementation with Lactobacillus rhamnosus
the Human Skin Microbiome Stimulates Ad- Ro YS. Antibiotic resistance of microbial SP1 normalises skin expression of genes im-
ipogenic Differentiation of Adipose-Derived strains isolated from Korean acne patients. J plicated in insulin signalling and improves
Stem Cells: Implications in Tissue Augmenta- Dermatol. 2012 Oct;39(10):833–7. adult acne. Benef Microbes. 2016 Nov; 7(5):
tion. J Invest Dermatol. 2017 Jan; 137(1): 46– 67 Simonart T, Dramaix M, De Maertelaer V. Ef- 625–30.
56. ficacy of tetracyclines in the treatment of acne 79 Fuchs-Tarlovsky V, Marquez-Barba MF, Sri-
56 Wang Y, Kuo S, Shu M, Yu J, Huang S, Dai A, vulgaris: a review. Br J Dermatol. 2008 Feb; ram K. Probiotics in dermatologic practice.
et al. Staphylococcus epidermidis in the hu- 158(2):208–16. Nutrition. 2016 Mar;32(3):289–95.
man skin microbiome mediates fermentation 68 Leyden JJ, Del Rosso JQ, Baum EW. The use 80 Baquerizo Nole KL, Yim E, Keri JE. Probiotics
to inhibit the growth of Propionibacterium of isotretinoin in the treatment of acne vul- and prebiotics in dermatology. J Am Acad
acnes: implications of probiotics in acne vul- garis: clinical considerations and future direc- Dermatol. 2014 Oct;71(4):814–21.
garis. Appl Microbiol Biotechnol. 2014 Jan; tions. J Clin Aesthet Dermatol. 2014 Feb; 7(2 81 Huang R, Ning H, Shen M, Li J, Zhang J, Chen
98(1):411–24. Suppl):S3–21. X. Probiotics for the Treatment of Atopic
57 Nishijima S, Kurokawa I, Katoh N, Watanabe 69 Ozlu E, Karadag AS, Ozkanli S, Oguztuzun S, Dermatitis in Children: A Systematic Review
K. The bacteriology of acne vulgaris and anti- Kilic M, Zemheri E, et al. Comparison of TLR- and Meta-Analysis of Randomized Con-
microbial susceptibility of Propionibacterium 2, TLR-4, and antimicrobial peptide levels in trolled Trials. Front Cell Infect Microbiol.
acnes and Staphylococcus epidermidis isolat- different lesions of acne vulgaris. Cutan Ocul 2017 Sep;7:392.
ed from acne lesions. J Dermatol. 2000 May; Toxicol. 2016 Dec;35(4):300–9. 82 Thio HB. The Microbiome in Psoriasis and
27(5):318–23. 70 Marcinkiewicz M, Majewski S. The role of an- Psoriatic Arthritis: The Skin Perspective. J
58 Skabytska Y, Biedermann T. Staphylococcus timicrobial peptides in chronic inflammatory Rheumatol Suppl. 2018 Jun;94:30–1.
epidermidis Sets Things Right Again. J Invest skin diseases. Postepy Dermatol Alergol. 2016 83 Chen YH, Wu CS, Chao YH, Lin CC, Tsai HY,
Dermatol. 2016 Mar;136(3):559–60. Feb;33(1):6–12. Li YR, et al. Lactobacillus pentosus GMNL-77
59 Xia X, Li Z, Liu K, Wu Y, Jiang D, Lai Y. Staph- 71 Feldman SR, Tan J, Poulin Y, Dirschka T, inhibits skin lesions in  imiquimod-induced
ylococcal LTA-Induced miR-143 Inhibits Kerrouche N, Manna V. The efficacy of ada- psoriasis-like mice. Yao Wu Shi Pin Fen Xi.
Propionibacterium acnes-Mediated Inflam- palene-benzoyl peroxide combination in- 2017 Jul;25(3):559–66.
matory Response in Skin. J Invest Dermatol. creases with number of acne lesions. J Am 84 Blank-Porat D, Gruss-Fischer T, Tarasenko
2016 Mar;136(3):621–30. Acad Dermatol. 2011 Jun;64(6):1085–91. N, Malik Z, Nudelman A, Rephaeli A. The an-
60 Lai Y, Cogen AL, Radek KA, Park HJ, Ma- 72 Jugeau S, Tenaud I, Knol AC, Jarrousse V, ticancer prodrugs of butyric acid AN-7 and
cleod DT, Leichtle A, et al. Activation of TLR2 Quereux G, Khammari A, et al. Induction of AN-9, possess antiangiogenic properties.
by a small molecule produced by Staphylo- toll-like receptors by Propionibacterium Cancer Lett. 2007 Oct;256(1):39–48.

294 Dermatology 2019;235:287–294 Claudel/Auffret/Leccia/Poli/Corvec/


DOI: 10.1159/000499858 Dréno

You might also like