Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

ORIGINAL ARTICLE

Identification of a Human Skin Commensal


Bacterium that Selectively Kills
Cutibacterium acnes
Alan M. O’Neill1, Teruaki Nakatsuji1, Asumi Hayachi2, Michael R. Williams1, Robert H. Mills3,
David J. Gonzalez3 and Richard L. Gallo1

The microbiome represents a vast resource for drug discovery, as its members engage in constant conflict to
outcompete one another by deploying diverse strategies for survival. Cutibacterium acnes is one of the most
common bacterial species on human skin and can promote the common disease acne vulgaris. By employing a
combined strategy of functional screening, genetics, and proteomics we discovered a strain of Staphylococcus
capitis (S. capitis E12) that selectively inhibited growth of C. acnes with potency greater than antibiotics
commonly used in the treatment of acne. Antimicrobial peptides secreted from S. capitis E12 were identified as
four distinct phenol-soluble modulins acting synergistically. These peptides were not toxic to human kerati-
nocytes and the S. capitis extract did not kill other commensal skin bacteria but was effective against C. acnes
on pig skin and on mice. Overall, these data show how a member of the human skin microbiome can be useful
as a biotherapy for acne vulgaris.
Journal of Investigative Dermatology (2020) 140, 1619e1628; doi:10.1016/j.jid.2019.12.026

INTRODUCTION undiscovered lantibiotics from a human skin S. hominus


Human skin is colonized by hundreds of diverse bacterial strain that targeted S. aureus (Nakatsuji et al., 2017). Another
species. The complex and dynamic chemical interactions interesting recent discovery was a novel antieS. aureus cyclic
between bacteria are important for host health and protection peptide produced by Staphylococcus lugdunensis (Zipperer
from pathogen colonization and infection (Grice and Segre, et al., 2016). Other examples include the production of
2011). The skin microbial community is regulated in part phenol-soluble modulin-gamma (PSMg, also known as
through production of antimicrobials. In particular, the delta-toxin) by S. epidermidis, which inhibits growth of
discovery of peptides from the human microbiome that S. aureus and group A Streptococcus (Cogen et al., 2010).
selectively kill Staphylococcus aureus, including methicillin- Human diseases are often associated with an imbalance in
resistant strains, represents a potential vanguard of new the microbial species that colonize the surface, a state known
antimicrobials (Chu et al., 2016; Cogen et al., 2010; as dysbiosis. In the case of atopic dermatitis, patients are
Nakatsuji et al., 2017; Saising et al., 2012; Zipperer et al., often colonized by S. aureus, which then further drives
2016). The major source for some of these new antimicro- inflammation and disease (Williams and Gallo, 2015). Like-
bials originates from coagulase-negative staphylococci wise, the common skin disease acne vulgaris is an inflam-
(CoNS), such as Staphylococcus epidermidis and Staphylo- matory disorder of the pilosebaceous unit that is linked to
coccus hominus. CoNS are a diverse bacterial species that colonization by Cutibacterium acnes. C. acnes is a lipophilic
frequently colonize human skin in great abundance and species that dominates sebaceous skin sites and produces a
therefore have evolved the most effective strategies for sur- multitude of toxins, adhesions, and immunomodulatory me-
vival in this environment. One mechanism responsible for tabolites that activate innate immune responses (O’Neill and
their competitive success is that specific strains of CoNS Gallo, 2018). Like many other bacterial species in the skin
harbor biosynthetic gene clusters that produce lantibiotics microbiome, C. acnes has several defense mechanisms that
(Donia et al., 2014). Our group recently identified several help it to establish a dominant niche in the hair follicle. For
example, the action of secreted C. acnes lipases on sebum
can result in generation of free fatty acids and short-chain
1
Department of Dermatology, University of California, San Diego, CA, fatty acids that can suppress the growth of methicillin-
USA; 2School of Medicine, Tohoku University, Sendi, Japan; and
3
Department of Pharmacology, University of California, San Diego, CA,
resistant S. aureus and group A Streptococcus (Shu et al.,
USA 2013). In the disease acne, distinct strains of C. acnes are
Correspondence: Richard L. Gallo, Department of Dermatology, University of enriched significantly compared with healthy individuals,
California San Diego, MC 0869, 9500 Gilman Drive, San Diego, California and these organisms appear to exacerbate disease because
92093, USA. E-mail: rgallo@ucsd.edu both systemic and topical antibiotic therapy that inhibits
Abbreviations: CFU, colony-forming unit; CoNS, coagulase-negative staph- C. acnes is therapeutically effective (Fitz-Gibbon et al.,
ylococci; PSM, phenol-soluble modulin; SLST, single locus sequence typing;
TSB, tryptic soy broth
2013). However, long-term use of pharmacologic antibi-
otics is undesirable because of the capacity to promote
Received 1 November 2019; revised 18 December 2019; accepted 26
December 2019; accepted manuscript published online 23 January 2020; antibiotic resistance and their indiscriminate killing of
corrected proof published online 22 February 2020 beneficial members of the healthy microbiome (Chien et al.,

ª 2020 The Authors. Published by Elsevier, Inc. on behalf of the Society for Investigative Dermatology. www.jidonline.org 1619
AM O’Neill et al.
Killing C. acnes with the Microbiome

2019; Ng et al., 2013; Shaw et al., 2019). It is therefore antimicrobial activity to outcompete C. acnes C2 (Figure 1e).
logical to explore use of the human skin microbiome as a To investigate if the antimicrobial supernatant is bactericidal,
source of antagonistic strains and/or undiscovered antimi- the number of surviving C. acnes colonies was enumerated
crobials that could selectively target C. acnes. The selection after 24 hours of treatment with increasing concentrations of
of microbes that have evolved to adapt and be tolerated by S. capitis E12. Consistent with results from the functional
humans optimizes the probability that they will promote screen, S. capitis supernatant was generally not bactericidal
establishment of normal homeostasis. against other CoNS species, with the exception of the
Here, we report discovery of a human skin commensal S. hominus strain 27844 only at the highest concentration
bacterial strain that produces antimicrobial molecules tar- (10). However, the S. capitis E12 supernatant was bacteri-
geted against C. acnes. This strain of Staphylococcus capitis cidal against C. acnes, showing a 6-log decrease of C. acnes
exhibited potent activity against multiple isolates of C. acnes, C2 during exposure to 0.6 (60%) supernatant and complete
including acne-associated strains, but did not kill other sterilization of the culture at highest concentrations
commensal skin bacteria. Identification of this unique (Figure 1f).
member of the human skin microbial community represents
an attractive alternative therapeutic approach to acne Purification and identification of PSMb antimicrobial
vulgaris. peptides
Initial examination into the nature of the antimicrobial factor
RESULTS revealed it to be resistant to heat treatment but sensitive to
Screening for antimicrobial activity from human skin proteolysis, suggestive of a proteinaceous molecule
bacteria identifies a S. capitis strain with potent and (Supplementary Figure S1a and b). Furthermore, the antimi-
selective activity against C. acnes crobial factor was precipitated from supernatant during
To discover antimicrobials from commensal bacteria on hu- incubation with 60e80% ammonium sulphate and concen-
man skin that inhibit the growth of C. acnes, a library of 288 trated by centrifugation (Supplementary Figure S1c). Next,
CoNS isolates were collected from skin swabs of the forearm sample preparation was carried out on the concentrated
and face of healthy volunteers. An unbiased analysis of ammonium sulphate precipitate using solid-phase extraction.
antieC. acnes activity was performed by live coculture with Samples were first loaded onto a hydrophilic-lipophilic
the CoNS isolate growing on agar with C. acnes C2 and balanced column and the antieC. acnes molecule was
measuring the growth of C. acnes following the addition of found to be eluted at 80% acetonitrile, as determined 72
50% v/v conditioned CoNS supernatant that was sterile hours after inoculation onto C. acnes C2 agar (Figure 2a) or
filtered (Figure 1a). Results of the functional screen revealed a with C. acnes C2 in liquid culture (Figure 2b). Next, the 80%
total of 11 isolates that were active against the growth of active elution was subsequently loaded onto a hydrophobic
C. acnes C2 in both the liquid assays and agar coculture C8 cartridge and the antieC. acnes molecule was eluted at
(Figure 1b and c). DNA extracted from the CoNS isolates 50% acetonitrile (Figure 2a and b). Total protein staining of
with antimicrobial activity was subjected to 16S sequencing the fractions revealed a highly enriched band(s) of roughly
and several different species were identified, including 4e5 kDa in size, with greater purity visible during subse-
S. epidermidis and S. hominus, which are considered quent elution steps (Figure 2c). Reverse-phase HPLC revealed
frequent colonizers of human skin. The most potent isolate several peaks, of which a single fraction (fraction 21) was
was identified as S. capitis strain E12 and was used for further found to suppress the growth of C. acnes C2 in liquid culture
characterization. Of the 11 antimicrobial strains identified, (Figure 2d). To ensure the highest purity, the active fractions
all were isolated from the forearm. None of the facial isolates were then pooled together from four separate HPLC runs and
were found to inhibit the growth of C. acnes C2. The a second step of HPLC purification was performed. This time,
lantibiotic-producing S. hominus A9 strain that is active several small peaks were visualized (Figure 2f), and two
against S. aureus (Nakatsuji et al., 2017) was ineffective fractions (fractions 15 and 16) were found to suppress the
against C. acnes C2, illustrating the selective nature of anti- growth of C. acnes C2 in liquid culture (Figure 2g). Mass
microbials derived from the mixed bacterial community of spectrometry analysis of active fractions (15 and 16) and
the skin. control nonactive fractions (13, 14, 17, and 18) revealed four
Next, the selectivity of S. capitis E12 to inhibit the growth candidate peptides referred to as antibacterial protein, each 5
of several different CoNS commensal species and common kDa in size (Figure 2h). A Basic Local Alignment Search Tool
skin pathogens was tested by agar assay. Although S. hominus search of the peptide sequences identified these as belonging
A9 potently inhibited the growth of S. aureus and group A to the beta class family of phenol-soluble modulins (PSMb)
Streptococcus, S. capitis E12 exhibited only weak activity present within multiple Staphylococcal species.
against CoNS and was ineffective against other skin patho- Although four distinct PSMb peptides were detected by
gens (Figure 1d). S. capitis E12 exhibited potent activity mass spectrometry, analysis of the S. capitis E12 genome
against a wide range of C. acnes strains, including strains that revealed up to six PSMb-encoding genes, contained within
were isolated from either lesional or nonlesional skin of pa- an operon-like structure (Figure 3a). All six PSMb-encoding
tients with acne. A 50% dilution of unconcentrated media genes are closely related, with amino acid sequence identity
sterile supernatant from S. capitis E12 was sufficient to inhibit ranging from 56% to 91% and predicted charge ranging
C. acnes growth, thus validating the findings from the func- from 1 to þ1 (Figure 3b). Further analyses found these
tional screen and demonstrating that, under normal growth genes to be absent from genomes of American Type Culture
conditions, this strain of S. capitis can produce sufficient Collection strains of S. capitis 35661 and 27844 (data not

1620 Journal of Investigative Dermatology (2020), Volume 140


AM O’Neill et al.
Killing C. acnes with the Microbiome

a
Isolation Antimicrobial Antimicrobial
agar assay liquid assay
Culture A
1 2 3 4 5 6 7 8 9 10 11 12

Selection B

1. Facial swab 2. Arm swab

b d
Skin pathogens and S. capitis S. hominus C. acnes isolates & site S. capitis S. hominus
Strain commensal species N030_E12 A9 SLS type of origin N030_E12 A9
1.5
Newman S. aureus + +++ K1 C. acnes Health +++
C. acnes growth OD600

USA300 S. aureus + +++ K2 C. acnes Health +++


** c NZ131 Group A Streptococcus +++ K1 C. acnes Health ++ +
DK23 Group B Streptococcus +++ A2 C. acnes Health +++
1.0 27844 S. hominus + H1 C. acnes Health +++
S. hominus A9 UCSD1 S. lugdunensis + H1 C. acnes NonLesional +++
UCSD1 S. warneri C2 C. acnes NonLesional ++
12228 S. epidermidis + E3 C. acnes NonLesional +++
0.5 RS218 E. coli E3 C. acnes NonLesional +++
PA01 P. aeruginosa F4 C. acnes Lesional +++
S. capitis E12 PA4 P. aeruginosa C2 C. acnes Lesional +++
14028 S. Typhimurium A1 C. acnes Lesional +++
AB5075 A. baumannii C2 C. acnes Lesional +++
0
Face Arm

e f
1×1012
C. acnes growth (OD600 nm)

3.0 1×1010
Log CFU/mL

2.5 1×108
2.0
1×106
1.5
1×104
1.0
0.5 1×102
limit of detection
0 1×100
10 20 50 100
0
1X

3X

6X

5X

5X

X
25

10
0.

0.

0.

2.

Sterile CoNS supernatant (%)


1.

Concentrated S. capitis E12 supernatant


S. capitis E12
S. hominus A9 S. lugdunensis UCSD1 S. epidermidis 12228
Media S. hominus 27844 C. acnes K1 (health)
S. aureus 113 C. acnes C2 (acne)

Figure 1. Functional screen to identify antimicrobial CoNS species that target Cutibacterium acnes. (a) Schematic for the high-throughput antimicrobial
functional screen, outlining the collection and selection of CoNS strains from two distinct healthy skin sites and assays to detect antimicrobial activity against
C. acnes via coculture on agar or growth in sterile conditioned supernatant of CoNS. (b) Growth of C. acnes C2 after 24-hour incubation in 50% sterile filtered
supernatant of the CoNS strain library. (c) Growth of C. acnes C2 with CoNS coculture in agar. Each dot represents data from an individual CoNS clone. The
most potent antimicrobial CoNS isolate was selected and identified as Staphylococcus capitis E12 (red), whereas Staphylococcus hominus A9 (blue) did
not exhibit activity against C. acnes C2. (d) Table showing the inhibitory activity of S. capitis E12 and S. hominus A9 against several skin commensal and
pathogen strains, including several strains of C. acnes isolated from healthy and acne skin, as measured by size of zone inhibition by antimicrobial agar assay
(þ small, þþ medium, þþþ large). (e) Growth of C. acnes C2 after 24-hour incubation in media alone or increasing concentrations of sterile supernatant of
S. capitis E12 or S. hominus A9. (f) Survival of several species of staphylococci and C. acnes 24 hours post-treatment with increasing concentrations of sterile
S. capitis E12 supernatant, as measured by the number of surviving CFUs plated onto the appropriate selective agar. CFU, colony-forming unit; CoNS,
coagulase-negative staphylococci.

shown), and indeed no antimicrobial activity was detected (Joo et al., 2011). This resulted in a relatively pure extract
against C. acnes from these laboratory strains (Figure 3c). enriched for all PSMb peptides and retained activity against
Alpha helical wheel plots (HeliQuest) also predicted these C. acnes C2 (Supplementary Figure S2). To confirm that PSMb
peptides to form an a-helix, amphipathic-like structure for peptides were active as the antieC. acnes molecule and
each PSMb, with separate hydrophobic and hydrophilic faces identify which peptide had optimal activity, we individually
on the opposite sides of the long axis of the peptide. This synthesized PSMb1 and 6, the most abundant molecules
amphipathic structure is common among many well- from the purified active fractions (15 and 16, Figure 2h), as
characterized antimicrobial peptides (Figure 3d) (Tossi well as PSMb4, which was less abundant. All three PSMs
et al., 2000). were shown to possess antimicrobial activity in vitro
(Figure 4a). PSMb1 and 6 both effectively inhibited growth of
PSMb inhibits skin colonization by C. acnes C. acnes C2 at 62 mg/ml, whereas PSMb4 was less potent,
S. capitis E12econditioned supernatant was extracted with n- inhibiting at 125 mg/ml. The combination of all three peptides
butanol, a procedure that enriches for PSM peptides exhibited optimal activity against C. acnes, inhibiting growth

www.jidonline.org 1621
AM O’Neill et al.
Killing C. acnes with the Microbiome

a b c
HLB column C8 column

Protein marker
9

Flow through

Flow through
C. acnes growth (OD600 nm)

80% elution

20% elution

50% elution
Live S. capitis E12 8

60% AS
7
6 Size
60% AS supernatant (kDa)
5
4 250
Flow through 3 150
HLB column 100
2 75
* ** ** 50
20% elution 1 ** ** ** 37
0 25
20

gh

gh
ia

% on

n
t
n

io

io

io

io

0% tio

io
ed

ta
ou

ou

i
50 luti
80% elution 15

ut

ut

ut

ut

ut

ut
10 elu
na
M

el

el

el

el

el

el
r

e
th

th
er

%
0%

%
ow

ow
p

20

80

20

30

40
Su
10

10
Fl

Fl
Flow through
Untreated HLB column C8 column 5
C8 column

2
20% elution

50% elution

d mAU e
3500 65
7
Absorbance 215 nm

3000 55
C. acnes (OD600 nm)

2500 45 6
MeCN (%)

Fraction 21
5
2000 35
4
1500 25
3
1000
15 2
500
1
0 0
0
0 5 10 15 20 25 30 35 40 ml 19 20 21 22 23
Fraction number

f mAU g
3000 7
50
Absorbance 215 nm

C. acnes (OD600 nm)

2500 6
40
2000 5
MeCN (%)

30
1500 Fractions 15 & 16 4
20
1000 3
500 10 2
0 0 1
-250 0
0 5 10 15 20 25 30 35 40 ml 13 14 15 16 17 18
Fraction number
h
Unique Size Fraction Fraction Fraction Fraction Fraction Fraction
Accession Protein description Coverage Peptides peptides PSMs (kDa) 13 14 15 16 17 18

A0A0S4MAE4 Antibacterial protein 1 homolog (PSMβ4) 79.5 9 8 25 4.5 2.07E+07 1.21E+06 6.80E+06 4.86E+05
A0A0S4MC59 Antibacterial protein 3 homolog (PSMβ6) 100.0 9 6 18 4.5 3.82E+05 1.06E+06 7.00E+06 1.91E+06 6.90E+04
A0A0U1EEI2 Uncharacterized protein 86.9 6 6 13 2.6 9.68E+06 4.82E+06 3.89E+05
A0A0S4M5T9 Antibacterial protein 1 homolog (PSMβ3) 81.8 6 4 12 4.6 4.42E+06 3.24E+05 3.35E+06 3.67E+04
A0A0S4MEM2 Immunodominant antigen B 10.5 2 2 8 20.1 8.12E+06 3.13E+06 6.04E+05 3.37E+06 7.00E+04
A0A0S4MFS8 Antibacterial protein 2 (PSMβ1) 50.0 3 3 3 4.8 3.62E+06 3.25E+06
A0A0U1E7C6 Elongation factor G 2.1 1 1 2 76.8 3.86E+06 3.49E+05
A0A0U1E2B3 Uncharacterised protein 34.3 2 2 4 7.1 3.14E+06 5.57E+05

Figure 2. HPLC purification and identification of Staphylococcus capitis E12 antimicrobial peptides. (a, b) Supernatant of S. capitis E12 precipitated by 60%
ammonium sulphate was loaded onto an HLB column and the antimicrobial factor eluted by 80% acetonitrile. The HLB 80% eluant was loaded onto a
C8 cartridge (indicated by arrow) and the antimicrobial factor was eluted by 50% acetonitrile. Fractions were measured for activity against Cutibacterium acnes
C2 by (a) agar assay and (b) liquid culture. Error bars depict mean  SEM. *P < 0.05; **P < 0.01 by t-tests comparing values against media control. (c) Silver
stain of the total protein content of S. capitis E12 treated or untreated supernatant and the SPE flow-through and eluted fractions. (d) HPLC purification of
S. capitis supernatant identified several peptide peaks, of which a single fraction (21) was identified as having antieC. acnes C2 activity by (e) liquid culture
assay. (f) Fraction 21 was pooled from five separate HPLC runs and purified by second step HPLC resulting in two fractions (15 and 16) with antieC. acnes C2
activity by (g) by liquid culture assay. (h) Results of the top eight peptide hits from MS detection of HPLC-purified active fractions (15 and 16) and control

1622 Journal of Investigative Dermatology (2020), Volume 140


AM O’Neill et al.
Killing C. acnes with the Microbiome

a PSMβ1-6 Figure 3. Whole genome sequencing


of Staphylococcus capitis E12 reveals
sequence and predicted properties of
5’Nucleotidase tRNA Phosphoesterase
the PSMb peptides. (a) Schematic
highlighting the S. capitis E12 genetic
cluster containing six gene-encoding
b 10 20 30 40
PSMb peptides (PSMb1e6). (b)
PSMβ1: M T K L AE A I A N T VK A GQD H DWAK L G T S I VG I AE N G I G L L GK V F G F +1 Multiple sequence alignment
PSMβ2: M T K L AE A I A N T VK A GQD H DWAK L G T S I VG I AE N G I GA L S K I F GG +1 (ClustalW) of all six PSMb peptides,
PSMβ3: MQK L AE A I A N T VK A GQD H DWAK L G T S I VG I AE N G I N A I T K I F GG +1
including the predicted charge for
PSMβ4: M T K L AE A I A N A VK A GQD QDWAK L G T S I VG I AE N G I S L L GK V F G F 0
each peptide. (c) Absence of
PSMβ5: MQK L AE A I A N T VK A GQD H DW T K L G T S I VD I VE N G V S A L T K V F GG 0
antimicrobial activity against
PSMβ6: ME K L F D A I RN T VD A G I N QDW T K L G T S I VD I VD N G V K V I S K F I G A -1
Cutibacterium acnes C2 during live
* ** ** * * ** * * * * * ** * * * ** * *
coculture or sterile supernatant
c d exposure with American Type Cell
0

PSMβ1 (aa 1-44)


84

66

2 Culture S. capitis strains 35661 and


E1
27

35

27840 that lack PSMb genes. White


s

Hydrophilic
iti

iti

iti

G
p

G
ca

ca

ca

A E T G arrow indicates zone of inhibition


S.

S.

S.

V K A A H G
N T produced by live S. capitis E12. (d)
I D K
L K Representative helical wheel plot
F G E
Live bacteria GN (HeliQuest) of PSMb1 indicating a
A K I S
T
W A M Fc predicted amphipathic structure. PSM,
L V D
I Q A I L phenol-soluble modulin; tRNA;
G L
Supernatant V transfer RNA.
Hydrophobic

at 31 mg/ml. However, this was not as inhibitory as the same primary human keratinocytes to extracts did not produce a
concentration of the butanol extract, which inhibited at 8 significant cytotoxic response from these epidermal cells as
mg/ml. This discrepancy could be because of the presence of measured by lactate dehydrogenase release (Figure 4b).
the additional fourth PSMb peptide (PSMb3) found within Likewise, mouse back skin exposed to different strains of
the extract. Several bacterial antimicrobials have been CoNS for 24 hours showed that S. capitis E12 did not induce
shown to synergize with human antimicrobials, such as observable erythema or injury (Figure 4c). Finally, having
defensins and LL-37 (Nakatsuji et al., 2017). However, confirmed the expected result that this commensal skin
exposure of C. acnes C2 to S. capitis extract with or without bacterium was well tolerated by the epidermis, C. acnes was
LL-37 peptide did not result in enhanced killing activity epicutaneously applied to colonize pig skin ex vivo or back
(Supplementary Figure S3). Given the greater potency and skin of live SKH1 mouse, and then extract from S. capitis E12
relative purity of the PSMb-containing extract compared was applied. A single treatment resulted in a significant
with the synthetically produced PSMb peptides, the extract reduction in C. acnes colony-forming units (CFUs) in both
was used in subsequent experiments to determine the effi- models (Figure 4d and e). Overall, these results demonstrate
cacy and potency of S. capitis E12 in in vitro and in vivo that PSMb from S. capitis E12 has the capacity to kill C. acnes
experiments. and illustrate the potential of exploiting naturally occurring
S. capitis E12 extract was next compared with several an- microbial events in the human skin for development of
tibiotics commonly used for acne treatment. Among four innovative therapeutics.
distinct acne-associated strains tested, sensitivity to each
antibiotic varied extensively, with three out of four strains DISCUSSION
insensitive to erythromycin at the maximum tested concen- In acne vulgaris, C. acnes is the dominant bacteria that plays
tration of 100 mg/ml (Supplementary Figure S4). In contrast, an important role in the disease (Barnard et al., 2016; Hall
all four strains were found to be equally sensitive to the et al., 2018). Treatment approaches for moderate to severe
extract (3e6 mg/ml). In three out of four C. acnes strains acne typically consist of an extended regimen of topical and/
tested, the extract exhibited greater potency than erythro- or oral application of antibiotics (Zaenglein et al., 2016).
mycin, tetracycline, and clindamycin (Supplementary However, such approaches can be ineffective for several
Figure S4aec), with one strain (C. acnes K2) found have reasons, including poor activity and penetration on skin.
greater sensitivity to antibiotics than the extract Furthermore, there are negative consequences of long-term
(Supplementary Figure S4d). Moreover, no increase in the antibiotic treatment, such as the ability to negatively disrupt
minimum inhibitory concentration above 16 mg/ml was the microbiome, thereby eliminating commensal bacteria
recorded for C. acnes during treatment with the extract for up important for skin health, and the increased risk for devel-
to 20 generations (data not shown), indicating relative opment of antibiotic resistance (Chien et al., 2019; Kelhälä
inability to promote resistance. Furthermore, exposure of et al., 2018). Here we report the discovery of several
=
nonactive fractions (13, 14, 17, and 18) revealing peptides corresponding to antibacterial protein and later identified as PSMb peptides by BLAST analyses.
BLAST, Basic Local Alignment Search Tool; HLB, hydrophilic-lipophilic balanced; MS, mass spectrometry; PSM, phenol-soluble modulin; SEM, standard error of
the mean; SPE, solid-phase extraction.

www.jidonline.org 1623
AM O’Neill et al.
Killing C. acnes with the Microbiome

a b
10
9

C. acnes growth (OD600 nm)


50
8
7 40

Cytotoxicity (%)
6
5 30
4
20
3
2 10
1
0 0
0 1 2 4 8 16 31 62 125 250 500 0 1.5 3.1 6.2 12.5 25 50 100
Concentration (μg/mL) Concentration (μg/mL)
PSMβ1 PSMβ6 S. capitis extract S. capitis E12 S. epidermidis 1457
PSMβ4 PSMβ1+4+6 S. epidermidis C5 S. capitis 35561

c d e
1×105 * 1×108 *
1×107
1×104
1×106
C. acnes CFU/mL

C. acnes CFU/mL
1×103 1×105
1×104
1×102 1×103
1×102
1×101
1×101
0
1×10 1×100
PBS Extract PBS Extract

Figure 4. Efficacy of PSMb peptides and extract as an alternative therapeutic for acne vulgaris. (a) Inhibition of Cutibacterium acnes C2 after 72-hour culture
in the presence of Staphylococcus capitis E12 extract or with synthetic peptides PSMb1, PSMb4, and PSMb6 individually or in combination of all three. (b)
Cytotoxicity of NHEKs treated with S. capitis E12, Staphylococcus epidermidis C5, S. epidermidis 1457, and S. capitis 35561 supernatant for 24 hours,
presented as the percentage of maximal LDH release. (c) Visual representation of erythema on the back skin of SKH1 mouse after inoculation with 1  107 CFU
of S. capitis E12 (1), S. epidermidis C5 (2), S. epidermidis 1457 (3), and S. capitis 35661 (4) for 24 hours. (d) Total number of surviving CFUs of C. acnes C2 on
ex vivo pig skin explants 24 hours post-treatment with S. capitis E12 extract (10 mg/ml) or PBS control. (e) Total number of surviving CFUs of C. acnes C2 on
SKH1 mouse back skin 24 hours post-treatment with S. capitis E12 extract (10 mg/ml) or PBS control. Data represents  SEM from five pig skin sheets or five
independent mice. *P < 0.05 by two tailed independent t-test. CFU, colony-forming unit; NHEK, normal human epidermal keratinocyte; LDH, lactate
dehydrogenase; PSM, phenol-soluble modulin; SEM, standard error of the mean.

bacterial species from healthy human skin that exhibit potent Discovery of therapeutically useful organisms must be
and selective antimicrobial activity against C. acnes. These designed with the specific target in mind.
results demonstrate an alternative approach for discovery of To discover potentially undiscovered antimicrobials effec-
effective antimicrobials that avoid the negative consequences tive against C. acnes, we conducted an expanded screen of
of current nonspecific antimicrobial therapies. CoNS from different healthy skin sites. Swabs were collected
There have been several recent discoveries of novel anti- from the face and forearm; the objective was not only to
microbials from skin bacteria. One example is lugdunin, a capture a diverse collection of CoNS species but also to
cyclic peptide antibiotic produced by S. lugdunensis that is sample an area of skin where C. acnes is less dominant,
active against S. aureus (Zipperer et al., 2016). It was reported potentially showing the success of CoNS against C. acnes.
that lugdunin was active only during agar coculture and not Results from our screen showed that a significantly greater
in liquid assay. Therefore, our functional screen and several number of antieC. acnes strains were identified from the
antimicrobial assays were conducted using both independent forearm than the face. One explanation for this could be a
approaches to ensure validity and reproducibility. Using such greater diversity of CoNS species on the forearm. Further-
approaches, our group have also identified several unique more, C. acnes can produce antimicrobial molecules, such as
antimicrobial peptides from CoNS including Sh-lantibiotics short-chain fatty acids, from the breakdown of sebum, which
a/b from S. hominus (Nakatsuji et al., 2017). This strain was have been shown to inhibit staphylococci (Shu et al., 2013);
found to kill S. aureus in vitro and in vivo. The selectivity of therefore, the increase in CoNS on the forearm may be aided
bacteriocins made by the skin microbiome was evident with by the lesser abundance of C. acnes. Together, these obser-
our observations that these strains of CoNS with vations suggest that the regional differences described in
antieS. aureus activity were not inhibitory against C. acnes. microbiomes at different body sites (Bouslimani et al., 2015;

1624 Journal of Investigative Dermatology (2020), Volume 140


AM O’Neill et al.
Killing C. acnes with the Microbiome

Oh et al., 2014) are not only owing to the host moisture or (Cheung et al., 2014). S. aureus contains two PSMbs
lipid levels but also reflect the outcome of microbial strate- (PSMb1/2), but neither are reported to be antimicrobial.
gies to outcompete their community members. Other studies have shown other synthetic PSMb peptides
In several acne microbiome studies, S. epidermidis is re- to be inhibitory against S. aureus in vitro but are not
ported to be most frequent CoNS species in health and dis- known to be active against C. acnes (Kumar et al., 2017).
ease (Barnard et al., 2016; Fitz-Gibbon et al., 2013), but only Therefore, adopting the present screening approach iden-
S. epidermidis strains located on the forearm were antimi- tified activity that would not have been detectable by
crobial in our study. Nevertheless, an important finding using a genetic screening approach alone. The optimum
originating from the acne microbiome studies showed that killing efficacy and specificity of the combination, rather
distinct strain-types of C. acnes are associated with disease. A than individual peptides, suggests that a therapeutic
valid hypothesis is that these strains are more pathogenic and approach combining multiple PSMs or the live organism
may drive inflammation and disease. Crucially, we have itself may be most effective. In conclusion, we have
shown that S. capitis E12 was capable of inhibiting all shown that the skin microbiome is a valuable resource for
C. acnes strains tested, including lesional strains isolated discovering antimicrobials that can be remarkably selec-
from patients with acne, but not other commensal bacteria. tive against specific pathogens and valuable in combating
This selective antimicrobial activity would be an attractive skin disease such as acne vulgaris.
therapeutic approach for acne, targeting disease-causing
bacteria while leaving other essential commensal bacteria MATERIALS AND METHODS
unperturbed. Human subjects and sample swab collection
S. capitis E12 constitutively produces antimicrobial pep- All experiments involving human subjects were carried out ac-
tides in sufficient quantities to kill C. acnes. Strikingly, in- cording to protocols approved by the University of California, San
cubation of a 0.6 concentration of S. capitis E12 Diego institutional review board (Project#140144). Written
supernatant with a strain of C. acnes that is associated with informed consent was obtained from all subjects. Swab sample
acne resulted in a greater than 4-log reduction in survival of collection was performed as previously described (Nakatsuji et al.,
the disease-associated bacterium and complete sterilization 2017) on 12 healthy adults, with swabs taken from the upper arm
of the culture after 24 hours. Moreover, for many acne- and the face. Healthy adults included both males and females aged
associated C. acnes strains, the S. capitis E12 extract 22e34 years with normal healthy skin and were instructed to avoid
showed greater potency than other antibiotics commonly washing and cosmetic application to the skin sites 24 hours before
prescribed for acne. This could be particularly important for sampling. Swabs were incubated in 3% tryptic soy broth (TSB),
patients who are colonized with strains that are insensitive vortexed briefly, and plated onto a mannitol salt agar containing egg
and/or have developed genetic resistance. Another desirable yolk plate for selective growth of Staphylococcal species (Parisi and
feature of the extract was that C. acnes C2 did not show Hamory, 1986). S. aureus was distinguished from CoNS according to
acquisition of resistance up to 20 generations. The potency mannitol metabolism and the egg yolk reaction (Carter, 1960).
and poor capacity to induce resistance from the PSMs pro- C. acnes clinical isolates were obtained by swabbing lesional and
duced by S. capitis E12 is consistent with the mode of action nonlesional facial skin sites of five patients with acne, as well as
of the a-helix amphipathic structure of these peptides. Such a facial skin from four healthy volunteers. Subjects included both
structure is common among many antimicrobial peptides and males and females aged 18e24 years, with patients with acne
enables insertion into and destabilization of bacterial mem- diagnosed with mild-to-moderate facial acne. Patients with acne at
branes (Tossi et al., 2000). Such a mechanism of direct the time of sample collection were not using any known topical or
membrane disruption is ideal for an antimicrobial that works systemic antibiotic or retinoid and were instructed to avoid any
on an epithelial surface. washing and cosmetic application to the skin sites in the preceding
Although the S. capitis genome contains a cluster of up 24 hours. For C. acnes strain collection, facial swabs were incubated
to six PSM-encoding genes, only PSMb1, 3, 4, and 6 were in reinforced clostridial media, vortexed, and plated onto brucella
detected by mass spectrometry analysis of the purified blood agar plates supplemented with Vitamin K, hemin, and 5%
fractions. Antimicrobial assays with synthetic peptides sheep’s blood and incubated for 5 days at 37  C in an anaeroPack
PSMb1, 4, and 6 revealed greater activity against C. acnes (Thermo Fisher Scientific, Waltham, MA).
when the bacteria were treated with a combination of all
three. Likewise, the expected overall charge of each PSMb Screening for antimicrobial activity from skin-derived CoNS
is subtly different, thus likely impacting binding capacity Individual isolated colonies of CoNS that were selected from each
to a hydrophobic column as well as interaction with the skin site (n ¼ 24) were randomly picked from a mannitol salt agar
bacterial membrane. Individually, the synthetic PSM pep- containing egg yolk plate and transferred to TSB (1 ml) in a deep
tides were far less potent than the extract. It remains to be 96-well plate. Each plate contained a previously characterized
determined if PSMb2 and PSMb5 were also secreted by CoNS strain, which included S. epidermidis ATCC1457 that failed
S. capitis E12 but eluted in different fractions that were to demonstrate antimicrobial activity against other staphylococci
not biologically active by themselves but would increase (negative control), as well as S. hominus A9, which was previously
the activity of PSMb1, 3, 4, and 6. The b class PSMs have demonstrated to produce lantibiotics that kill S. aureus (positive
been found in other staphylococci and typically number control) (Nakatsuji et al., 2017). The CoNS plate was sealed with
between two and four (Wang et al., 2011). However, their sterile Aeraseal film (Sigma, St. Louis, MO) and cultured at 37  C
exact role has remained elusive and, unlike the smaller overnight, shaking at 250 r.p.m. Bacterial growth was evaluated by
PSMa, are not thought to be involved in virulence measuring OD600 and only CoNS grown to a high density (OD600

www.jidonline.org 1625
AM O’Neill et al.
Killing C. acnes with the Microbiome

> 6.0) were used for subsequent analysis. To measure antimicro- acetonitrile in water and eluted in 80% acetonitrile in water. The
bial activity in the secreted supernatant, the CoNS supernatant eluted fractions were lyophilized and reconstituted in water. The
from overnight culture was harvested and sterile filtered. To 80% fraction was then loaded onto a C8 Sep-Pak cartridge
measure antimicrobial activity from live growing CoNS, bacteria (Waters), washed in 20% acetonitrile, and eluted in 50% aceto-
from overnight culture were pelleted by centrifugation then the nitrile. The fractions were lyophilized, reconstituted in water, and
supernatant was discarded, and the bacteria were resuspended in then assessed for antimicrobial activity and visualized by silver
fresh TSB. stain (Pierce, Thermo Fisher Scientific) according to manufac-
turer’s instructions. First step HPLC purification was carried out
In vitro antimicrobial assays with 1 mg of S. capitis E12 supernatant loaded into a CapCel
For the initial CoNS antimicrobial screen, the radial diffusion agar Pak C8 (5 mm, 300 Å, 4.6  250 mm) (Shiseido, Tokyo, Japan)
assays were performed using S. aureus 113 or either C. acnes with a linear gradient of acetonitrile from 10% to 60% in
HL110PA3 (health-associated, SLST K1) or C. acnes HL096PA1 0.1% (v/v) trifluoroacetic acid at 0.8 ml/min. Fractions were
(acne-associated, SLST C2). For the radial diffusion agar assays, lyophilized then resuspended in water and antimicrobial activity
melted TSB or reinforced clostridial media agar (12 ml) was mixed assessed by liquid culture assay. Up to five sequential purifica-
with S. aureus or C. acnes (1  106 CFU) and poured into a 10-cm2 tions were carried out with each single antimicrobial fraction
petri square. When the agar was solidified, a 10-ml aliquot of pooled together for second step HPLC. For the second purifica-
bacteria was inoculated onto a single grid. The plates were incu- tion, a linear gradient of acetonitrile from 25% to 50% was
bated at 37  C overnight without shaking (for S. aureus) or incu- adopted. Peptides were synthesized with N-terminal formulation
bated at 37  C for 3 days in an anaeroPack (for C. acnes) to allow to at least 95% purity by a commercial vendor (LifeTein LLC,
visible growth of bacteria. Antibacterial activity was indicated by a Somerset, NJ).
clear zone of inhibition within the agar that surrounds the colony.
The size of the inhibitory zone was recorded as a measure of Purification of antimicrobials produced by CoNS strains
antimicrobial activity (þ slight, þþ moderate, or þþþ potent). For Conditioned media from overnight cultures of the selected anti-
the liquid culture assay, conditioned supernatant from overnight bacterial S. capitis E12 strain, including positive control antimicro-
cultures of CoNS were harvested and sterile filtered (0.22 mm). bial S. hominus A9 strain, were first sterilized by filtration through a
Conditioned supernatant (50%) was mixed with 50% fresh TSB or 0.22-mm Millipore filter. Initial characterization involved treating the
reinforced clostridial media containing 1  105 CFU/ml of sterile bacterial supernatant to boiling at 100  C for 15 minutes or
S. aureus or C. acnes in a 96-well round bottom plate. S. aureus incubation with proteolytic enzymes papain and proteinase K at 200
plates were incubated at 37  C with shaking overnight and C. acnes mg/ml at 37  C for 60 minutes. Antimicrobial activity was measured
plates were incubated for 48 hours standing at 37  C in an anae- by radial diffusion assay. For size exclusion of the antimicrobial
roPack. Bacterial growth was measured by OD600, and positive molecule, 20 ml of conditioned media was loaded onto a 3, 10, and
antimicrobial strains were identified as those that suppressed bac- 30 kDa molecular weight cutoff columns (Pierce) and centrifuged at
terial growth to less than 50% of growth measured for negative 4,000g for 15 minutes. The flow-through fraction was set aside, and
control strains. Bacterial survival was measured by counting the the retained fraction was washed twice in PBS and resuspended with
number of CFU on TSB plates (S. aureus) or brucella blood agar 20 ml TSB. Antimicrobial activity was assessed for both fractions. To
plates (C. acnes). At selected times post-treatment with bacterial precipitate the active molecule, ammonium sulphate was added to
supernatant or extracts, the number of CFUs was determined by the sterile supernatant (30e80% saturation) for 1 hour under con-
serial dilution in PBS and plating onto appropriate agar media. stant rotation at room temperature, followed by centrifugation at
Bacterial survival was measured as the total number of CFUs per 4,000g for 45 minutes. The resulting pellet was washed three times
milliliter. with water and reconstituted in water. Antimicrobial activity was
determined by S. aureus and C. acnes growth in agar and liquid
Bacterial classification culture.
Genomic DNA was isolated from CoNS and C. acnes isolates by
MasterPure kit (Epicentre, Madison, WI) according to manufacturer’s Mass spectrometry
instructions. For species identification for CoNS, full-length 16S ri- A portion of the fractions of interest (< 1 mg) were dried under
bosomal RNA genes were amplified from representative isolates vacuum and resuspended in 5 ml of 5% acetonitrile with 5% formic
with universal 16S primers, 27-F and 1525-R. Amplicons were acid. Next, individual liquid chromatography- mass spectrometry
sequenced from both ends by Sanger method. Obtained 16S ribo- (Elias and Gygi, 2007) experiments were conducted on one fifth of
somal RNA gene sequence was searched against Ribosomal Data- each sample through 85 minutes of data acquisition on an Orbitrap
base Project (http://rdp.cme.msu.edu/) (Cole et al., 2014). For Fusion (Thermo Fisher Scientific) mass spectrometer with an in-line
C. acnes strain typing, single locus sequence typing (SLST) was used Easy-nLC 1000 (Thermo Fisher Scientific). A home-pulled and
with primers SLSTFor and SLSTRev (Scholz et al., 2014). Amplicons packed 30-cm column was triple-packed with 0.5 cm, 0.5 cm, and
were sequenced from both ends by Sanger method. C. acnes strain 30 cm of 5-mm C4, 3-mm C18, and 1.8-mm C18, respectively, and
types were assigned to amplicons using the SLST database (http:// heated to 60  C for use as the analytical column. Peptides were first
medbac.dk/slst/pacnes). loaded at 500 bar, which was followed by a chromatography
gradient ranging from 6% to 25% acetonitrile over 70 minutes fol-
Solid-phase extraction and HPLC purification of supernatant lowed by a 5-minute gradient to 100% acetonitrile, which was held
S. capitis E12 sterile supernatant that had been retained on a 10 for 10 minutes. Electrospray ionization was performed by applying
kDa molecular weight cutoff column and precipitated in 60% 2,000 V through a stainless-steel T-junction connecting the analyt-
ammonium sulphate was applied on an Oasis HLB cartridge ical column and Easy-nLC system. Each sample was followed by two
(Waters, Milford, MA). The cartridge was washed in 20% washes starting with a gradient from 3% to 100% acetonitrile over

1626 Journal of Investigative Dermatology (2020), Volume 140


AM O’Neill et al.
Killing C. acnes with the Microbiome

15 minutes with an additional 10 minutes at 100% acetonitrile. An RHM; Visualization: AMO; Writing - Original Draft Preparation: AMO, RLG;
m/z range of 375e1,500 was scanned for peptides with charge states Writing - Review and Editing: AMO, TN, MRW, AH, RHM, DJG, RLG.
between 2 and 6. Centroided data was used for quantitation of
peaks. Acquisition was run in a data-dependent positive ion mode. SUPPLEMENTARY MATERIAL
Raw spectra was searched in Proteome Discoverer Version 2.1 Supplementary material is linked to the online version of the paper at www.
jidonline.org, and at https://doi.org/10.1016/j.jid.2019.12.026.
against a uniprot reference database for Staphylococcus capitis
(Uniprot proteome UP000042965, accessed 10/01/2018) using the
Sequest algorithm (Eng et al., 1994) alongside a reverse database REFERENCES
approach used to control peptide and protein false discoveries to 1% Barnard E, Shi B, Kang D, Craft N, Li H. The balance of metagenomic ele-
ments shapes the skin microbiome in acne and health. Sci Rep 2016;6:
(Elias and Gygi, 2007). No enzyme was specified in the search and a 39491.
minimum peptide length was set to six amino acids. Search pa-
Bouslimani A, Porto C, Rath CM, Wang M, Guo Y, Gonzalez A, et al. Mo-
rameters included a precursor mass tolerance of 50 p.p.m and lecular cartography of the human skin surface in 3D. Proc Natl Acad Sci
fragment mass tolerance of 0.6 Da and variable oxidation for USA 2015;112:E2120e9.
modifications. Carter CH. Egg yolk agar for isolation of coagulase-positive staphylococci.
J Bacteriol 1960;79:753e4.

Transplantation of antimicrobial CoNS on pig skin and mice Cheung GYC, Joo HS, Chatterjee SS, Otto M. Phenol-soluble modulins e
critical determinants of staphylococcal virulence. FEMS Microbiol Rev
All experiments involving live animal work were in accordance with 2014;38:698e719.
the approval of the Institutional Animal Care and Use Guidelines of
Chien AL, Tsai J, Leung S, Mongodin EF, Nelson AM, Kang S, et al. Associ-
the University of California, San Diego (protocol no. S09074). Fresh- ation of systemic antibiotic treatment of acne with skin microbiota char-
frozen pig skin sheets were obtained from Loretta Tomlin Animal acteristics. JAMA Dermatol 2019;155:425e34.
Technologies (Livermore, CA) and sanitized by surgical brush with Chu J, Vila-Farres X, Inoyama D, Ternei M, Cohen LJ, Gordon EA, et al.
3% chloroxylenol. The skin sheet was cut into 2.5 cm  2.5 cm and Discovery of MRSA active antibiotics using primary sequence from the
human microbiome. Nat Chem Biol 2016;12:1004e6.
rinsed 20 times with sterile PBS. C. acnes (1  107 CFU, ATCC6919)
was epicutaneously challenged on pig skin for 1 hour and 100 ml of Cogen AL, Yamasaki K, Muto J, Sanchez KM, Crotty Alexander L, Tanios J,
et al. Staphylococcus epidermidis antimicrobial delta-toxin (phenol-
S. capitis E12 extract (10 mg/m) or PBS control was applied for 24 soluble modulin-gamma) cooperates with host antimicrobial peptides to
hours. Live bacteria were harvested by swabbing to measure kill group A Streptococcus. PLOS ONE 2010;5:e8557.
C. acnes survival. For mouse experiments, the backs of hairless Cole JR, Wang Q, Fish JA, Chai B, McGarrell DM, Sun Y, et al. Ribosomal
SKH1 mice were scrubbed with alcohol wipes and 1  107 CFU Database Project: data and tools for high throughput rRNA analysis.
Nucleic Acids Res 2014;42:D633e42.
C. acnes was inoculated onto sterile gauze pads, which were placed
onto the dorsal skin and secured with tegaderm film for 24 hours. Donia MS, Cimermancic P, Schulze CJ, Wieland Brown LC, Martin J,
Mitreva M, et al. A systematic analysis of biosynthetic gene clusters in the
The C. acnesecontaining gauze pad was removed and 100 ml of human microbiome reveals a common family of antibiotics. Cell
S. capitis E12 extract (10 mg/ml) or PBS control was inoculated onto 2014;158:1402e14.
a fresh gauze pad and placed back onto the same dorsal skin site and Elias JE, Gygi SP. Target-decoy search strategy for increased confidence in
secured with tegaderm film for an additional 24 hours. Skin was large-scale protein identifications by mass spectrometry. Nat Methods
swabbed to measure surviving CFUs of C. acnes 48 hours 2007;4:207e14.
postinoculation. Eng JK, McCormack AL, Yates JR. An approach to correlate tandem mass
spectral data of peptides with amino acid sequences in a protein database.
J Am Soc Mass Spectrom 1994;5:976e89.
Statistical analysis Fitz-Gibbon S, Tomida S, Chiu BH, Nguyen L, Du C, Liu M, et al. Propioni-
Statistical analysis was performed by using Graphpad Prism bacterium acnes strain populations in the human skin microbiome asso-
(GraphPad Software, Inc., La Jolla, CA; version 7.03). Statistically ciated with acne. J Invest Dermatol 2013;133:2152e60.
significant differences were calculated by appropriate statistical Grice EA, Segre JA. The skin microbiome [published correction appears
methods as outlined. P values of  0.05 were considered significant. in Nat Rev Microbiol. 2011;9(8):626]. Nat Rev Microbiol 2011;9:
244e53.
ORCIDs Hall JB, Cong Z, Imamura-Kawasawa Y, Kidd BA, Dudley JT, Thiboutot DM,
Alan M. O’Neill: http://orcid.org/0000-0002-5892-6477 et al. Isolation and identification of the follicular microbiome: implications
Teruaki Nakatsuji: http://orcid.org/0000-0002-6187-2991 for acne research. J Invest Dermatol 2018;138:2033e40.
Asumi Hayachi: http://orcid.org/0000-0002-4504-9583 Joo HS, Cheung GYC, Otto M. Antimicrobial activity of community-
Michael R. Williams: http://orcid.org/0000-0002-1523-6353 associated methicillin-resistant Staphylococcus aureus is caused by
Robert H. Mills: http://orcid.org/0000-0002-0749-9524 phenol-soluble modulin derivatives. J Biol Chem 2011;286:8933e40.
David J. Gonzalez: http://orcid.org/0000-0003-1423-5970
Richard L. Gallo: http://orcid.org/0000-0002-1401-7861 Kelhälä HL, Aho VTE, Fyhrquist N, Pereira PAB, Kubin ME, Paulin L, et al.
Isotretinoin and lymecycline treatments modify the skin microbiota in
CONFLICT OF INTEREST acne. Exp Dermatol 2018;27:30e6.
AMO, TN, and RLG are coinventors of University of California San Diego Kumar R, Jangir PK, Das J, Taneja B, Sharma R. Genome analysis of
technology related to the bacterial AMPs discussed herein, and RLG is a Staphylococcus capitis TE8 reveals repertoire of antimicrobial peptides
cofounder and consultant of MatriSys Bioscience. Funding for this project was and adaptation strategies for growth on human Skin. Sci Rep 2017;7:
provided by Almirall pharmaceutical company. AMO and RLG were funded 10447.
by National Institutes of Health grants R01AR074302, R01AR076082, and
R37AI052453. Nakatsuji T, Chen TH, Narala S, Chun KA, Two AM, Yun T, et al. Antimi-
crobials from human skin commensal bacteria protect against Staphylo-
AUTHOR CONTRIBUTIONS coccus aureus and are deficient in atopic dermatitis. Sci Transl Med
Conceptualization: AMO, RLG; Formal Analysis: AMO, TN, RHM; Funding 2017;9.
Acquisition: RLG; Investigation; AMO, TN, MRW, RHM; Methodology: Ng KM, Ferreyra JA, Higginbottom SK, Lynch JB, Kashyap PC, Gopinath S,
AMO, RLG, DJG, RHM; Project Administration: AMO, RLG; Resources: et al. Microbiota-liberated host sugars facilitate post-antibiotic expansion
AMO, RLG, DJG, AH, TN; Supervision: RLG; Validation: AMO, TN, MRW, of enteric pathogens. Nature 2013;502:96e9.

www.jidonline.org 1627
AM O’Neill et al.
Killing C. acnes with the Microbiome

O’Neill AM, Gallo RL. Host-microbiome interactions and recent progress into Shu M, Wang Y, Yu J, Kuo S, Coda A, Jiang Y, et al. Fermentation of Propio-
understanding the biology of acne vulgaris. Microbiome 2018;6:177. nibacterium acnes, a commensal bacterium in the human skin micro-
Oh J, Byrd AL, Deming C, Conlan S, , NISC Comparative Sequencing Pro- biome, as skin probiotics against methicillin-resistant Staphylococcus
gram, Kong HH, et al. Biogeography and individuality shape function in the aureus. PLOS ONE 2013;8:e55380.
human skin metagenome. Nature 2014;514:59e64. Tossi A, Sandri L, Giangaspero A. Amphipathic, alpha-helical antimicrobial
Parisi JT, Hamory BH. Simplified method for the isolation, identification, and peptides. Biopolymers 2000;55:4e30.
characterization of Staphylococcus epidermidis in epidemiologic studies. Wang R, Khan BA, Cheung GYC, Bach TH, Jameson-Lee M, Kong KF, et al.
Diagn Microbiol Infect Dis 1986;4:29e35. Staphylococcus epidermidis surfactant peptides promote biofilm matura-
Saising J, Dube L, Ziebandt AK, Voravuthikunchai SP, Nega M, Götz F. tion and dissemination of biofilm-associated infection in mice. J Clin Invest
Activity of gallidermin on Staphylococcus aureus and Staphylococcus 2011;121:238e48.
epidermidis biofilms. Antimicrob Agents Chemother 2012;56: Williams MR, Gallo RL. The role of the skin microbiome in atopic dermatitis.
5804e10. Curr Allergy Asthma Rep 2015;15:65.
Scholz CFP, Jensen A, Lomholt HB, Brüggemann H, Kilian M. A novel high- Zaenglein AL, Pathy AL, Schlosser BJ, Alikhan A, Baldwin HE, Berson DS,
resolution single locus sequence typing scheme for mixed populations of et al. Guidelines of care for the management of acne vulgaris. J Am Acad
Propionibacterium acnes in vivo. PLOS ONE 2014;9:e104199. Dermatol 2016;74:945e973.e33.
Shaw LP, Bassam H, Barnes CP, Walker AS, Klein N, Balloux F. Modelling Zipperer A, Konnerth MC, Laux C, Berscheid A, Janek D, Weidenmaier C,
microbiome recovery after antibiotics using a stability landscape frame- et al. Human commensals producing a novel antibiotic impair pathogen
work. ISME J 2019;13:1845e56. colonization. Nature 2016;535:511e6.

1628 Journal of Investigative Dermatology (2020), Volume 140


AM O’Neill et al.
Killing C. acnes with the Microbiome

SUPPLEMENTARY MATERIAL

a b Supplementary Figure S1.


Characterizing the biological nature

K
of the Staphylococcus capitis active

se
ed
4

a
at

in

n
molecule(s). (a) Growth of

ai
re

te
C. acnes growth (OD600 nm)

p
nt

o
3.5

Pa
Pr
Cutibacterium acnes C2 was

U
3 measured in liquid culture with or
2.5 S. hominus A9 without S. capitis E12 supernatant
untreated, boiled at 100  C for 15
2
minutes, or separated through 10 kDa
S. capitis E12
1.5 or 3 kDa MWCO columns into
1 retained or flow-through fractions for
S. capitis 10kDa Retain 24 hours. (b) S. capitis E12 and control
0.5
Staphylococcus hominus A9
0 supernatant was subjected to papain
or proteinase K proteolytic digestion
w

w
ia

n
tn

ile

i
flo

flo
ed

ta

ta
ta

(200 mg/ml) for 60 minutes at 37  C


bo

re

re
na
M

a
kD

D
a

a
er

SN

3k
kD

D
p

10

and inoculated onto C. acnes


3k
Su

10

econtaining agar to measure


c antimicrobial activity. (c) CoNS
supernatant was subjected to
%

precipitation in different saturation


30

40

50

60

70

80

amounts of ammonium sulphate, and


antimicrobial activity of each
S. capitis E12 precipitate was measured by
visualizing growth of C. acnes C2 on
agar after 72 hours. AS, ammonium
S. hominus A9 sulphate precipitate; CoNS,
coagulase-negative staphylococci;
MWCO, molecular weight cutoff.

Supplementary Figure S2. PSMb


peptides are extracted by n-butanol
treatment of Staphylococcus capitis
E12 supernatant. (a) Antimicrobial
activity against Cutibacterium acnes
C2 is retained and enhanced after
butanol extraction of PSMb from
sterile supernatant of S. capitis E12 but
not with the lantibiotic-producing
Staphylococcus hominus A9. (b) Silver
stain of the total protein content of
S. capitis E12 supernatant before and
after butanol extraction showing
enrichment of small PSMb peptides.
AS, ammonium sulphate; PSM,
phenol-soluble modulin.

www.jidonline.org 1628.e1
AM O’Neill et al.
Killing C. acnes with the Microbiome

18
C. acnes growth (OD600 nm) 16
14
12
10
8
6
4
2
0
05

7
01

03

06

1
00

00

00

00

0.

0.

0.
00

0.

0.

0.
0.

0.

0.

0.
0.

Dilution factor

Extract & LL37 Extract (1 mg/mL)


LL37 (64 µM) Media

Supplementary Figure S3. PSMb does not synergize with host LL-37
antimicrobial peptide. Assessment of synergistic antimicrobial activity against
Cutibacterium acnes C2 from Staphylococcus capitis E12 extract and/or
human LL-37 antimicrobial peptide, at indicated concentrations for 72 hours.
PSM, phenol-soluble modulin.

a b
8 9
7 8
Growth of C. acnes C2

Growth of C. acnes K1

6 7
6 Tetracycline
5
5 Clindamycin
4
4 Erythromycin
3
3 S. capitis extract
2 2
1 1
0 0
0 0.1 0.2 0.4 0.8 1.6 3.2 6.25 12.5 25 50 100 0 0.1 0.2 0.4 0.8 1.6 3.2 6.25 12.5 25 50 100
Concentration (µg/mL) Concentration (µg/mL)

c d
1.8 7
1.6
Growth of C. acnes A1

Growth of C. acnes K2

6
1.4
5 Tetracycline
1.2
1 4 Clindamycin
0.8 3 Erythromycin
0.6 S. capitis extract
2
0.4
0.2 1
0 0
0 0.1 0.2 0.4 0.8 1.6 3.2 6.25 12.5 25 50 100 0 0.1 0.2 0.4 0.8 1.6 3.2 6.25 12.5 25 50 100
Concentration (µg/mL) Concentration (µg/mL)

Supplementary Figure S4. Staphylococcus capitis E12 extract can exhibit greater potency against Cutibacterium acnes compared with several antibiotics.
(aed) Several distinct strains of C. acnes, isolated from skin of patients with acne (A1, C2, K1, and K2), were cultured for 48 hours in the presence of increasing
concentrations of S. capitis extract, tetracycline, clindamycin, or erythromycin.

1628.e2 Journal of Investigative Dermatology (2020), Volume 140

You might also like