You are on page 1of 44

 

 
Understanding the role of Propionibacterium acnes in acne vulgaris: the
critical importance of skin sampling methodologies

Hélène Omer PhD, Andrew McDowell PhD, Oleg A. Alexeyev MD,


PhD

PII: S0738-081X(16)30264-4
DOI: doi: 10.1016/j.clindermatol.2016.10.003
Reference: CID 7101

To appear in: Clinics in Dermatology

Please cite this article as: Omer Hélène, McDowell Andrew, Alexeyev Oleg A.,
Understanding the role of Propionibacterium acnes in acne vulgaris: the critical
importance of skin sampling methodologies, Clinics in Dermatology (2016), doi:
10.1016/j.clindermatol.2016.10.003

This is a PDF file of an unedited manuscript that has been accepted for publication.
As a service to our customers we are providing this early version of the manuscript.
The manuscript will undergo copyediting, typesetting, and review of the resulting proof
before it is published in its final form. Please note that during the production process
errors may be discovered which could affect the content, and all legal disclaimers that
apply to the journal pertain.
ACCEPTED MANUSCRIPT
O. Alexeyev

Title:

Understanding the role of Propionibacterium acnes in acne vulgaris: the critical importance of

skin sampling methodologies

P T
RI
Authors:

SC
Hélène Omer PhD1, Andrew McDowell PhD2, Oleg A. Alexeyev MD, PhD1

NU
Affiliations:
1
MA
Department of Medical Biosciences/Pathology, Umeå University, Umeå, Sweden.
2
Northern Ireland Centre for Stratified Medicine, School of Biomedical Sciences, University of
ED

Ulster, Londonderry, UK
PT

Correspondence
CE

Oleg A. Alexeyev Department of Pathology, Umeå University Hospital, S-90185 Umeå, Sweden,

Tel.: +46-90-7852841, Fax: +46-90-139853, e-mail: oleg.alexeyev@umu.se


AC

Conflicts of interest

None

1
ACCEPTED MANUSCRIPT
O. Alexeyev

Abstract

Acne vulgaris is a chronic inflammatory skin condition, classified by the Global Burden of

Disease Study as the eighth most prevalent disease worldwide. The pathophysiology of the

P T
condition has been extensively studied, with an increase in sebum production, abnormal

RI
keratinization of the pilosebaceous follicle, and an inflammatory immune response all implicated

SC
in its etiology. One of the most disputed points, however, is the role of the Gram-positive

anaerobic bacterium Propionibacterium acnes in the development of acne, particularly when this

NU
organism is also found in normal sebaceous follicles of healthy skin. Against this background, we
MA
now describe the different sampling strategies that have been adopted for qualitative and

quantitative study of P. acnes within intact hair follicles of the skin, and discuss the strengths and
ED

weaknesses of such methodologies for investigating the role of P. acnes in the development of

acne.
PT
CE

Key words: Skin, acne, Propionibacterium acnes, inflammation, gel biopsy, skin biopsy,

phylogroups
AC

2
ACCEPTED MANUSCRIPT
O. Alexeyev

Introduction

Acne vulgaris is a chronic, inflammatory skin disorder of the pilosebaceous follicles 1. It is

estimated that virtually all adolescents will be affected by acne at some point in their life, with

P T
15-20% suffering moderate-to-severe forms of the condition 2. The disease can also persist into

RI
adulthood, affecting 20-40% of all individuals 3. Strikingly, The Global Burden of Disease Study

SC
has estimated acne to affect approximately 10% of the global population, ranking it as the eighth

most prevalent disease worldwide, and the third most prevalent dermatological condition 4.

NU
MA
The earliest subclinical ‘lesion’ in acne, known as the microcomedone, can evolve into open and

closed comedones and into different inflammatory lesions, such as papules, pustules, nodules,
ED

and cysts 5. Historically, comedogenesis has been linked to a combination of increased sebum

production and hyperproliferation of keratinocyte epithelium prior to the development of


PT

inflammation; however, more recent studies are now providing evidence that inflammation may
CE

actually play a role in all stages of acne lesion progression, including subclinically before the

formation of the comedo 6.


AC

The Gram-positive, anaerobic skin commensal Propionibacterium acnes has long been suspected

to play a key role in the pathophysiology of acne because antimicrobials that reduce skin surface

P. acnes result in clinical improvement, and acne, which is recalcitrant to antibiotic treatment, is

associated with the presence of antibiotic resistant strains 7. Despite these observations, the exact

role played by P. acnes in the development of acne has been consistently questioned based on a

number of important findings. These include the anti-inflammatory properties of antimicrobials,

which may explain, at least in part, their effectiveness in acne treatment, combined with the
3
ACCEPTED MANUSCRIPT
O. Alexeyev

observations that counts of P. acnes on acneic and healthy skin are very similar, levels of P.

acnes within lesions do not correlate with the degree of inflammation, and some inflamed lesions

show no evidence of bacterial colonization 8.

P T
RI
Kligman 5 estimated the number of pilosebaceous follicles on the human face to total

SC
approximately 5,000. This number may actually be significantly under-estimated, because the

number of hair follicles on the face could total at least 292 follicles/cm2 7, 9, which for a skin

NU
surface estimated to be 675 cm2 in area 10 would result in a total of 200,000 follicles. In general,
MA
acne is considered to be severe when the patient has more than 50 comedones and 30

inflammatory lesions on the face 11. This would mean that < 0.04 % of the follicles on the face
ED

are actually involved in the disease. If hyperkeratinization and increased sebum production were

indeed factors triggering acne then one would expect many more follicles to be affected.
PT
CE

In this review, we critically appraise the different methodologies which have been used to study

the association of P. acnes with acne vulgaris, and describe the current ‘state of play’ based on
AC

the results obtained with some of these various sampling approaches. We also consider the

importance and relevance of biofilm formation and inflammatory processes in the context of

acne.

P. acnes population structure and key genomic and biochemical properties potentially

related to acne vulgaris development

Before we describe the current methodologies used for sampling and detection of P. acnes in

healthy and acneic skin, we will first provide a brief overview of the phylogenetics and
4
ACCEPTED MANUSCRIPT
O. Alexeyev

population structure of the bacterium, along with relevant genomic and biochemical properties

that will be important for our discussion of later sections.

T
P
P. acnes is an opportunistic pathogen and a member of the ‘cutaneous’ group of human

RI
propionibacteria along with P. avidum, P. granulosum and P. humerusii, although it can also be

SC
isolated from the oral cavity and the genitourinary and gastrointestinal tracts. The original

seminal work on the population structure of P. acnes by Johnson and Cummins 12 revealed two

NU
distinct serotypes of P. acnes, designated types I and II, which differed in the sugar composition
MA
of their cell walls, as well as bacteriophage susceptibility 13. More recently, our understanding of

the genetic population structure of this bacterium has increased tremendously based on single,
ED

multilocus and whole genome sequence (WGS) analyses 14-19. These studies have demonstrated

that types I and II represent phylogenetically distinct divisions or clades, and have further
PT

identified a new and distinct genetic group, known as type III, which has the capacity to adopt a
CE

long filamentous cell morphology which is not observed with types I and II 14, 15.
AC

Strains within this type III clade have recently been proposed as Propionibacterium acnes

subspecies elongatum subsp. nov., with strains of type I and II classified as Propionibacterium

acnes subspecies acnes subsp. nov 20. Further subdivisions within the type I clade, designated

types IA1, IA2, IB, IC, have now also been described based on the higher phylogenetic resolution

obtained with multiple gene analysis 17, 21-23. These latter groups also display differences in

biochemical phenotype, their association with disease, production of putative virulence

determinants and resistance to anti-acne antibiotics 21, 22, 24.

5
ACCEPTED MANUSCRIPT
O. Alexeyev

Overall, P. acnes is in linkage disequilibrium and has a clonal, epidemic population structure

characterized by a number of highly successful and globally disseminated lineages 16, 17, 22.

Multilocus sequence typing (MLST) and ribotyping schemes developed for the bacterium have

P T
shown that within the large type I clade, strains from the type IA1 phylogroup cluster into three

RI
major clonal complexes (CC) (CC1, CC3, CC4; MLST8 scheme), while type IA2, IB and IC

SC
phylogroups are tighter phylogenetic clusters currently comprised of only one CC each 17, 22. The

large type II clade also clusters into three distinct CCs (CC6, CC71, CC72), while strains from

NU
the type III division comprise a single CC (CC77) 17, 22 (Figure 1). In addition to these CCs, a
MA
number of singleton sequence types (STs) have also been described (Figure 1). The availability of

over 100 complete and draft whole genome sequences for P. acnes has also facilitated detailed
ED

comparative genomic studies, which have been well described in the literature 18, 25-27; these

analyses have found an average genome size and gene content of 2.5 Mb and 2,500, respectively,
PT

for the bacterium.


CE

While overall genome plasticity is low, differences between the phylogroups due to the presence
AC

of island-like genomic regions with aberrant G+C content and flanking insertion sequences have

been discovered 18, 25, 26, 28. This ‘flexible gene pool’ encodes putative virulence factors and traits

which may be associated with fitness and niche adaptation, including antimicrobial defense,

adhesion and metabolic functions 28. One of the key observations from these studies has been the

discovery in type II strains of clustered regularly interspaced short palindromic repeats

(CRISPR)/Cas loci that confer immunity to bacteriophage and foreign mobile genetic elements 18,
26, 29
.

6
ACCEPTED MANUSCRIPT
O. Alexeyev

In contrast, type I and also type III strains contain CRISPR/cas gene remnants indicating deletion

of the locus during their evolutionary history. As a consequence, these latter clades may be much

more susceptible to horizontal gene transfer and the acquisition of fitness or virulence traits that

P T
are important in the context of human disease, especially acne. For example, strains from type

RI
IA1 CC3 (MLST8), contain a novel plasmid with a tight adhesion (Tad) locus and two unique

SC
genomic islands, designated loci 1 and 2, that contain genes which are proposed to enhance

virulence via increased bacterial adhesion and host immune response 18, 26, 30. This may be

NU
important in the association of these strains with acne as discussed later.
MA
Transcriptomic, proteomic, and biochemical studies have also revealed important host-interacting
ED

properties common to different strains of P. acnes which may be important in the context of

human disease, including the production of proteins with degrading activities, such as proteases,
PT

glycoside hydrolases and esterases 15, 16, 25, 31, 32. Differences between phylogroups in
CE

inflammatory potential and the expression of various putative virulence factors, some of which

may be important in the emerging association of type IA1 strains with acne, have, however, also
AC

been described. These include haemolysins, neuraminidase, lipase, polyunsaturated fatty acid

isomerase, the iron acquisition protein HtaA, Christie-Atkins-Munch-Peterson (CAMP) factors,

and immunogenic phase and antigenically variable dermatan sulphate-binding proteins (DsA1,

PPA2127; DsA2, PPA2210) 15-17, 21, 23, 25, 31-33. These latter proteins have a variable number of C

residues in a CnTCn motif which is upstream of a putative signal peptide, along with variable

numbers of repeats towards the mid-region and the carboxy-terminus that alter molecular mass

and protein expression 23, 33. This variation is most likely generated by slipped strand mispairing,

leading to phenotypic variation and possible evasion of the immune system as well as
7
ACCEPTED MANUSCRIPT
O. Alexeyev

dissemination via alteration of adhesin specificity. Recently, type IA1 strains isolated from

patients with acne (CC3; MLST8) were also found to produce significantly higher levels of

porphyrins, proinflammatory metabolites important in acne development, which were further

P T
enhanced by vitamin B12 supplementation 24. In contrast, strains believed to be associated with

RI
skin health produced low levels and did not respond to vitamin B12.

SC
Establishing the role of P. acnes in acne vulgaris

NU
Despite over 100 years of history, the role of P. acnes in acne vulgaris is still controversial.
MA
Numerous reviews have been published discussing arguments for and against the role of the

bacterium in the condition 6, 34-39. In order to critically evaluate a plethora of studies using
ED

different sampling and detection techniques, we need to define the extent to which Koch

postulates can be applied to support or to refute an etiological link between P. acnes and acne
PT

vulgaris. The very first postulate stating that the offending organism must be found in diseased
CE

but not in healthy individuals would formally reject a role for P. acnes in the condition since it is

present in persons without acne and in unaffected hair follicles from acne patients 40-42. As the
AC

organism is an opportunistic pathogen that forms part of the indigenous skin microbiota, its

presence in non-diseased skin would, however, be expected; a causative microbial component

that completely satisfies Koch’s postulates has rarely been identified in skin diseases 43. As a

result, other quantitative or qualitative differences must be demonstrated between acne-affected

and intact hair follicles if we are to establish a clear link to the disease. We therefore suggest a

modification of Koch’s postulates, and propose that a key finding should be significantly higher

bacterial counts of viable P. acnes within individual acne lesions compared to unaffected hair

follicles. To meet this requirement, the content of an entire hair follicle must be sampled and
8
ACCEPTED MANUSCRIPT
O. Alexeyev

contamination from nearby anatomical structures (skin surface, stratum corneum) avoided. The

choice of a control hair follicle is vital as age, gender, diet, usage of cosmetics, localization of

skin biopsy, hair follicle functional status and a magnitude of other known and unknown factors

P T
can influence the abundance of P. acnes in the skin at any given time. An anatomically intact hair

RI
follicle derived from the same patient and the same area as the acne lesion is required in order to

SC
avoid sampling bias. Furthermore, both the acne lesion and control skin must be sampled at the

same point in time. In addition, our improved understanding of P. acnes phylogeny and the

NU
identification of distinct genetic divisions and lineages with apparent heightened capacity to
MA
cause infection also raises the possibility that differences in the nature of the phylogroups and

STs isolated from individual acneic lesions versus normal follicles may also be an important,
ED

although not conclusive, observation in the association of P. acnes with the etiology of acne

development 16, 17, 22.


PT
CE

Contamination from other non-follicular skin structures is also a major concern in studies

investigating the role of P. acnes in acne, especially when applying sensitive nucleic acid assays.
AC

A recent study showed that P. acnes DNA can be detected in all next generation sequencing

datasets generated from multiple samples of different origin, including non-template controls 44.

This highlights the importance of careful interpretation of data generated by molecular methods,

and further experiments to validate any association identified.

Overview of methodologies to sample P. acnes in the skin

The skin is a diverse environment, containing different habitats such as invaginations,

appendages, glands and follicles 43. As bacteria may inhabit many different skin appendices,
9
ACCEPTED MANUSCRIPT
O. Alexeyev

different methodologies have been developed to sample these various areas. Within a clinical

setting, swab, scrub ,and gel biopsy are used to sample the upper skin levels, while a skin biopsy

is required in order to sample hair follicles and subcutaneous fat tissue (Table 1) 45. Because acne

P T
vulgaris is a disease of the hair follicle, it is imperative that sampling strategies target the whole

RI
follicular P. acnes community. Some studies have utilized multiple sampling methodologies

SC
with their cohorts, making meaningful comparison of the data across healthy skin and lesion

types very difficult 46. In the following section, we will discuss the advantages and disadvantages

NU
of current sampling methods in their capacity to target the follicular population.
MA
Swab, scrub and tape stripping techniques
ED

The swab technique refers to surface sampling using a sterile tissue to brush the skin. The method

is non-invasive, cheap and can be applied to the sampling of many patients in a short period of
PT

time. Its limitation, however, is the difficulty of standardization since variation in the user-
CE

dependent force applied during sampling may affect microbe retrieval 47. As a result, it is difficult

to compare quantitative data between and within studies, which is further compounded by
AC

variation in the structure, chemical composition and toxicity of swab tissues which may also

affect bacterial retrieval 48. The method only samples skin surface bacteria, which is appropriate

when screening for antibiotic resistant strains, but tells us little regarding the microbiology of

individual follicles.

Williamson and Kligman 40 developed a technique known as the “surface scrub technique” or

“detergent scrub technique” that also allows isolation of skin surface microbiota. This technique

uses a blade to gently scrape the skin surface but, similar to swab sampling, considerable
10
ACCEPTED MANUSCRIPT
O. Alexeyev

variations in quantification are observed. To minimize this variation, triplicate samples at a given

site on a given day are recommended 40. While comparing the swab and scrub method, Keyworth
49
concluded that they gave similar concentrations of the bacterial populations sampled.

P T
RI
Another way to collect skin surface bacteria is the direct application of a contact plate, velvet pad

SC
or stripping tape to the skin. The plate and pad only target surface microbiota and, therefore, are

generally used for aerobic bacteria. The tape application was developed to study the different

NU
layers of human epidermis 50 and was later adapted by Updegraff 51 to quantify the bacterial
MA
population of up to 14 stratum corneum layers of the skin. The bacteriostatic activity of different

tapes against some bacteria is, however, an issue 51. Tape stripping can also remove different
ED

quantities of material again making it difficult for the method to be standardized.


PT

Overall, these techniques should be considered unsuitable for quantitative studies examining the
CE

relationship between P. acnes and acne vulgaris, because they target surface and stratum corneum

bacterial populations, provide very little material from inside the hair follicle, and are difficult to
AC

standardize.

Gel biopsy

In order to provide insight into follicular microbial populations, Holland and Roberts in 1974 52

developed a sampling technique based on cyanoacrylate gel (Permabond). In this approach, a

drop of cyanoacrylate gel is deposited on the skin, enabling varying amounts of stratum corneum

and follicular infundibulum content to be sampled 53. Cyanoacrylate gel polymerizes quickly, and

will immediately form a very strong adhesive whenever exposed to water and amino acid content
11
ACCEPTED MANUSCRIPT
O. Alexeyev

present in the skin 54; the kinetics of gel polymerization upon contact with sebum/lipids is not

known. The extent of penetration into the hair follicle will likely be confined to the superficial

region, and again this method is highly variable due to skin humidity, pH and other unaccounted

P T
environmental factors.

RI
SC
Gel sampling will yield numerous follicular casts, which makes it extremely difficult to link an

individual cast to an acne-affected or intact but dilated hair follicle; acneic follicles are

NU
intermixed with healthy follicles which are numerically predominant, even in severe cases of
MA
disease. Another limitation is that varying amounts of intrafollicular content will be extracted

from individual hair follicles, while unknown and unpredictable amounts of material will be left
ED

in the skin. While the gel biopsy method remains an attractive approach to make an approximate

quantitative analysis of hair follicle infundibulum, it is unsuitable for analysis of the entire hair
PT

follicle.
CE

Microcomedone / Comedone extraction methods


AC

The contents of an individual inflamed follicle (or lesion) can be extracted by using a special

device such as stylet, lancet or comedone extractor. In an elegant experiment, Plewig 55 showed

that by taking a biopsy directly after extraction, the hair follicles still retained abundant horny

materials. While the technique can be attractive for larger skin lesions it may be problematic for

smaller ones and is not suitable for retrieving the contents of healthy hair follicles needed as

controls.

The same applies for the vacuum extractor technique largely advertised by dermatologic

companies. For example, the presence of P. acnes in different parts of the skin with and without
12
ACCEPTED MANUSCRIPT
O. Alexeyev

acne has been investigated using a constant negative pressure to aspirate the contents of

sebaceous follicles, followed by DNA extraction and quantitative PCR (qPCR) for numeration of

P. acnes populations.56 Another major limitation of the vacuum extraction method is that the

P T
number of sebaceous follicles sampled is unknown, as well as the quantity of material extracted.

RI
SC
A more recent approach to sample the contents within pilosebaceous units, including the

anaerobic portion, is the utilization of commercially available pore strips (eg., Bioré®) 18.

NU
Individual follicular contents can then be plucked of the resulting strip using microforceps,
MA
pooled and processed for further analyses. With this approach, minimal sampling of the skin

surface or human cells within the follicular lining appears to occur as judged by metagenomic
ED

shotgun sequencing and determination of human sequence reads 57. While the method can be

used for small (microcomedones) and healthy follicles, it is limited in its ability to separate
PT

lesional from non-lesional comedones, but it has the advantage of reaching deeper into the
CE

sebaceous follicle.
AC

Biopsy

Skin biopsy represents the gold standard approach, enabling the whole hair follicle to be

retrieved. This approach is, however, invasive and therefore requires approval from ethical

regulatory bodies. Isolation of an intact hair follicle from the surrounding tissue is also

problematic. A 1 M CaCl2 solution has been used to isolate pilosebaceous units from skin

biopsies, a technique first described by Kellum 58. 59. Briefly, a punch biopsy is excised from

the skin and soaked in cold calcium chloride to facilitate separation of the epidermis and follicles

from the dermis; each follicle is then excised using surgical microscissors. The cumbersome

13
ACCEPTED MANUSCRIPT
O. Alexeyev

nature of the procedure, exposure of biological material to chemicals, and unavoidable risks of

cross-contamination of skin structures during manipulation makes it a poor method for

quantitative analysis.

P T
RI
As an alternative, the biopsy can be subjected to direct visualization by various staining assays

SC
such as immuno-fluorescence microscopy (IFM) 42, 60 and fluorescence in-situ hybridization

(FISH) 61-63. With this approach, however, a conventional tissue section which is between 4-10

NU
µm thick provides only a limited insight or glimpse into the hair follicle content. As we have
MA
shown that P. acnes biofilms can be as large as 2 mm, as many as 400 skin sections may need to

be evaluated in order to visualize the whole hair follicle 64.


ED

Overview of methodologies to detect P. acnes sampled from the skin


PT

Three main methods used to detect microorganisms in skin samples are culture, analysis of
CE

nucleic acid content, and direct visualization by IFM / FISH. These methods detect the bacteria in

different physiologic states and this information needs to be considered since such bacteria may
AC

have different pathogenic potentials (Table 2) 65. The physiologic states of bacteria in natural

environments include (i) live bacteria that are viable and metabolically active, (ii) dormant

bacteria that are alive but metabolically inactive and (iii) dead bacteria that are non-viable,

usually due to membrane defects.

The direct culture of a sample will only reveal viable bacteria at a snapshot in time, and will be

biased towards those organisms that flourish under the nutritional milieu presented to them 43.

Restriction of typing schemes to only a small number of colonies may also underrepresent
14
ACCEPTED MANUSCRIPT
O. Alexeyev

specific lineages that may be present in a mixed type sample in low numbers; this may be

especially relevant to P. acnes 19. Nucleic acid detection is usually done on samples by PCR or

qPCR using highly conserved regions of the 16S rDNA gene ubiquitously present in bacteria.

P T
The availability of low cost methods for WGS analysis also makes this an attractive alternative,

RI
although both approaches will amplify viable and non-viable bacterial DNA, as well as

SC
extracellular DNA which is abundant in many natural environments 66.

NU
Microscopic visualization also enables the direct quantitative analysis of bacteria in the skin, and
MA
is an extremely attractive approach for the analysis of P. acnes and visualization of probable

biofilm formation. Furthermore, as bacteria in the skin can appear in biofilms, pretreatment of
ED

samples by sonication may also be important, especially to maximize cell numbers for

quantitative analyses 67.


PT
CE

P. acnes analysis based on swab and scrub sampling

The swab and scrub techniques have been used to define the pattern of Propionibacterium
AC

colonization on normal human skin and acneic skin with similar results. Propionibacteria are

ubiquitous on the human skin, and P. acnes is dominant in oily regions with high sebum

concentration, including the face and the upper trunk. Definitive quantitative differences linking

P. acnes levels on the skin to acne have not, however, been found based on the culture or

sequencing of organisms sampled using these techniques 68-70, although age-dependent

differences in P. acnes levels have been described 71. P. acnes appears more prevalent in acne

patients during the time when the disease begins (age 11-20), whereas after the age of 20 years,

the levels of P. acnes in healthy subjects is comparable to patients with acne.


15
ACCEPTED MANUSCRIPT
O. Alexeyev

A limited number of early studies utilizing low resolution biotyping of P. acnes isolated from

healthy and acneic skin via the swab sampling technique have been described; 46, 72, 73.however,

P T
much more recent high resolution phylogenetic and population genetic analysis of P. acnes

RI
isolated from healthy and acneic skin by swab and scrub sampling have now been reported,

SC
although in some cases this was also combined with gel biopsy methods (which we discuss later)

making interpretation of the results slightly more complicated 16, 17, 19, 20, 22, 23, 74. These latter

NU
independent studies, which have been based on subjects in geographically widespread regions,
MA
have consistently demonstrated that acneic skin of patients with moderate-to-severe acne appears

associated with, or enriched for, particular strains from the type IA1 phylogroup, while strains
ED

from the type IA2, IB, IC and II divisions are infrequently recovered, and those from the type III

phylogroup never recovered. Other strains have also been identified that appear associated with
PT

skin health 16.


CE

In particular, patients with acne appear to be enriched for epidemic clones and other STs from the
AC

type IA1 clonal complexes CC1, CC3 and CC4 (MLST8 scheme). Nevertheless, while these

recent studies clearly indicate significant differences in the phylogroup composition of the skin

from acne sufferers and healthy individuals, the swab and scrub sampling methods do not directly

sample follicular contents, but measure the content or output of all follicles, both healthy and

acneic, on the skin surface. It therefore remains to be determined exactly what role P. acnes

strains from these different CCs may play in the pathophysiology of acne.

P. acnes analysis based on gel biopsy


16
ACCEPTED MANUSCRIPT
O. Alexeyev

P. acnes distribution has also been studied using the gel biopsy method 52, 75 followed by bacterial

culture. Holland and Roberts 52 described their Permabond-based method as more effective than

the scrub technique of Williamson and Kligman, 40 because the procedure can be applied several

P T
times to the same region of the skin, enabling part of the hair follicle microbiota and varying

RI
amounts of the stratum corneum populations to be sampled. In order to investigate the early

SC
follicular changes in acne, the structural organization and colonization of follicular casts of

prepuberal children has been studied 76. This work demonstrated by microscopy that even though

NU
the follicular casts were present in all samples and had the same anatomy as those present in acne
MA
patients, they were not as abundant and were also sterile as assessed by microscopy and culture.

This led to the conclusion that there was no evidence for any bacterial role in the initiation of
ED

acne lesions. Additional studies 77 focused on the collection of cutaneous microbiota from various

areas of facial skin using cyanoacrylate gel, followed by the measurement of bacterial numbers
PT

after culture. There were significantly higher numbers of P. acnes in follicular casts from patients
CE

compared with their control volunteers for female facial skin and male back skin, and that

approximately 90% of normal follicles had no detectable viable microorganisms versus only 10%
AC

in acne follicles. No differences in the skin microbiology of adolescent acne patients, persistent

acne patients or late-onset acne patients were, however, observed.

A later study 78 used a molecular biology-based approach to investigate the microbial diversity of

follicles from acne patients and healthy individuals sampled by cyanoacrylate gel biopsies. In this

study, pooled follicular casts were microdissected before DNA sequence analysis of cloned 16s

rRNA genes. Interestingly, and in contrast to the data previously generated 77, the researchers

observed that follicles from healthy skin were exclusively colonized by P. acnes, and that the
17
ACCEPTED MANUSCRIPT
O. Alexeyev

follicular microbiota of acne patients included, in addition to P. acnes, Staphylococcus

epidermidis and minor proportions of other species. As this was a molecular based project, no

information on the physiological state of the bacteria was available, which is a major limitation of

P T
the study.

RI
SC
A more recent proteomic investigation examined the protein content of follicular casts extracted

from healthy and acne-affected individuals using cyanoacrylate-gel biopsies processed for mass

NU
spectrometry analysis 79. Bacterial proteins were identified in both normal and acne affected skin,
MA
and were exclusively from P. acnes; the most prevalent P. acnes proteins identified were surface-

exposed dermatan sulphate adhesins (DsA1; DsA2), CAMP factors, and an uncharacterized
ED

lipase. Since DsA1 and DsA2 were abundantly found in the follicular cast proteome, this

provides clear evidence that healthy as well as diseased individuals are colonized with P. acnes
PT

strains from the type IA phylogroup, although other types may also be present. In addition to
CE

bacterial proteins, normal follicular casts were enriched in proteins (prohibitins and

peroxiredoxins) which are involved in protection from various stresses, including reactive oxygen
AC

species. By contrast, follicular casts extracted from acne-affected skin contained proteins

involved in inflammation, wound healing and tissue remodeling, including myeloperoxidase,

lactotransferrin, and neutrophil elastase inhibitor. This indicates that the biological processes

taking place within acneic lesions reflect a ‘host response to a bacterium’. In addition to the

problems previously described utilizing a gel-based sampling methodology, this study did

highlight other potential limitations including variation due to differences in sampling sites,

which was the nose for healthy controls and facial and back skin areas for patients with acne.

18
ACCEPTED MANUSCRIPT
O. Alexeyev

While some of these studies provide tantalizing hints of a role for P. acnes in the pathological

processes leading to acne, the variability in results, especially in relation to P. acnes colonization

of follicles in healthy subjects, restrict our ability to make solid conclusions; such differences

P T
may reflect the nature of cohorts studied along with other inherent study variabilities arising from

RI
gel biopsy sampling.

SC
P. acnes analysis based on microcomedone/ comedone extraction methods

NU
To determine the microbiota specific to acne, methods based on follicle extraction have been
MA
used as described earlier. Some of these techniques, however, make it difficult to extract control

follicles, but they do give insight into the follicular microbiota of acneic lesions. Using such
ED

approaches, a number of studies have found that the organisms present in inflamed and non-

inflamed acne lesions are similar to that of normal microbiota 58, 70, 80-84. In most of these studies,
PT

a varying number of sterile lesions have been found supporting the hypothesis that the presence
CE

of P. acnes in a follicle is not necessary to trigger comedogenesis 8, 85-87, but the lack of a

matched control population makes these comparisons between “lesional” and “normal” bacterial
AC

populations difficult to interpret.

The contents of sebaceous follicles have also been sampled by aspiration using a constant

negative pressure, with subsequent quantification of the P. acnes population by qPCR56. This

sampling approach demonstrated that numbers of P. acnes genome-equivalents were extremely

low in control subjects less than 10 years of age, but higher in older subjects. This investigation

also observed that P. acnes counts on the forehead and nose were higher in acne patients aged 10-

14 years than in the age-matched controls in both sexes, and that the proportion of P. acnes
19
ACCEPTED MANUSCRIPT
O. Alexeyev

present on one subject is highly individual and variable between subjects, even in the same age

range. Overall, their results suggested that hair follicles containing many P. acnes cells are not

particularly prone to acne, except in younger teenagers. While this method is sensitive it doesn’t

P T
discriminate between acne and non-acne follicles and takes into account all DNA present,

RI
including non-viable bacteria.

SC
A more recent study.examined the P. acnes strain populations on the skin of patients with acne

NU
and healthy subjects18. The contents of pilosebaceous follicles on the nose of both cohorts,
MA
irrespective of the presence or absence of acneic lesions, were extracted using commercially

available pore strips, followed by metagenomic analysis. While the relative abundances of P.
ED

acnes were found to be similar between the two cohorts, the strain population structures were

significantly different. In particular, using a novel ribotyping scheme, a sub-population of type


PT

IA1 strains representing ribotypes (RT) 4 and 5 appeared highly enriched on the skin of patients
CE

with acne, while other lineages (RT7, RT8, RT9, RT10) were also associated with the disease and

a subpopulation of type II strains (RT6) was also strongly associated with the skin of healthy
AC

subjects.

Interestingly, although this study utilised a metagenomics approach, thereby identifying all strain

populations, the results were in part agreement with data from previous culture-based MLST

studies, which similarly found that strains corresponding to RT4/5 (CC3; MLST8) and RT8

(CC4; MLST8) dominated isolates from acneic skin 16, 17, 22. However, strains from CC1

(MLST8), also found to be strongly associated with moderate-to-severe acne based on previous

MLST studies 16, 17, 22, were not found to be enriched within the skin of acne patients versus
20
ACCEPTED MANUSCRIPT
O. Alexeyev

controls using this metagenomic approach 18. This study not only differed from previous

investigations in the nature of the detection methodology utilised, which would measure non-

viable DNA, but also in the fact that it attempted to look for differences in the nature of the

P T
underlying follicular microbiome of pre-lesional samples at the same anatomical site of subjects,

RI
rather than focus on regions where diseased, lesional skin was present.

SC
NU
P. acnes analysis based on biopsies

As the sebaceous follicle population is heterogeneous, methods isolating each single follicle by
MA
microdissection of skin biopsies are a particularly potent means to investigate the correlation

between the presence of bacteria and inflammation. Such studies have emphasized the high
ED

variability of the follicular content between and within different individuals, as well as the large
PT

proportion of sterile lesions, which has led to the conclusion that the presence of microorganisms

is not essential for the initiation of comedogenesis and inflammation in acne 58, 85, 88. Using an
CE

IFM-based detection method on skin biopsies, Imamura et al. 60 found P. acnes in the sebaceous
AC

follicles of the face in normal subjects and in the uninvolved skin of acne subjects, but not the

normal skin on the back of healthy subjects and the uninvolved skin on the back of acne patients.

Evidence for P. acnes in 7/11 comedones and in all 30 inflammatory papules, pustules, and

nodules investigated was also found. Most of the fluorescence in the inflammatory lesion was

present within the confines of the follicle, with small amounts of bacterial antigen present in the

dermal infiltrate and within macrophages.

21
ACCEPTED MANUSCRIPT
O. Alexeyev

Using a similar approach, a more recent IFM study by Jahns et al. 42 found a statistically

significant higher prevalence of P. acnes in follicles from acne patients compared to controls

(47% vs 21%). Furthermore, these studies suggest that P. acnes may be present in the follicles in

P T
the form of macrocolonies / biofilms (Figure 2).

RI
SC
P. acnes biofilm in the follicle

Most bacteria exist as biofilms in their natural habitat. A biofilm is defined as a bacterial

NU
community embedded in an extracellular matrix attached to a surface; both mono- and poly-

species biofilms have been described


MA
89, 90
. Biofilms are usually more resistant to antibiotics than
91-93 94
planktonic cells , at least in part due to the presence of persister cells . They are associated
ED

with a variety of infections and diseases, including chronic wounds, miliaria, atopic dermatitis,

onychomycosis, impetigo and furuncles, and are often challenging to treat 95-97.
PT

98
Biofilm formation by P. acnes was first described in relation to prosthetic hip infections and
CE

various orthopedic biomaterials 99. The sequence of the P. acnes genome 100 has revealed clusters
AC

of genes involved in the polysaccharide biosynthesis of a glycocalyx polymer, which can be used
101 102-
for biofilm matrix formation . Several studies have investigated P. acnes biofilms in vitro
104
and, as expected, P. acnes growing within a biofilm is more resistant to antibiotics than its

planktonic counterparts; the biofilms can even develop in the presence of some antibiotics 105 and

biofilm dispersal and removal by antibiotics does not necessarily correlate with the death of the
106
bacteria . Furthermore an increase in the lipase activity and auto-inducer 2 (AI-2) production

was detected in the cells grown as a biofilm 106.

22
ACCEPTED MANUSCRIPT
O. Alexeyev

P. acnes biofilms have been directly observed in acne lesions more frequently than in control

sebaceous follicles (37% vs 13%) 42. The biofilms are present in the stratum corneum as well as

matrix-encased macro-colonies in the hair follicles (Figure 2). Most interestingly, these large

P T
macro-colonies appear to consist of different P. acnes phylogroups existing within the same

RI
follicle 42, which is intriguing given the association of type IA1 strains with acne. To further

SC
characterize P. acnes biofilms in the skin, a 3-D model has been reconstructed by sequential

cutting of the entire skin biopsy 63, 64. The biofilms are found attached to the follicle wall, the hair

NU
shaft, spreading over the lumen, and/or in the centre of the hair follicles without any evident
MA
attachment. They may start from the stratum corneum and go as deep as 2 mm under the skin

surface. Such observations therefore highlight the need for correct sampling methods when
ED

studying the presence or absence of P. acnes in skin samples.


PT

P. acnes biofilms have also been observed in other skin diseases such as folliculitis 107, folliculitis
CE

decalvans 108 and hidradenitis suppurativa (acne inversa) 109. Interestingly, in the above

conditions the biofilms were seen in terminal hair follicles.


AC

Acne vulgaris and inflammation

P. acnes can activate a variety of immune pathways and induce the production of interleukin

(IL)-1α, tumor necrosis factor (TNF)-α and granulocyte-macrophage colony-stimulating factor

(GM-CSF) 110 in keratinocytes 111, 112. Pro-inflammatory cytokines have also been directly

detected in acne lesions including IL-1α in open comedones and IL-1 expression in early
23
ACCEPTED MANUSCRIPT
O. Alexeyev

inflamed lesions 111, 113. Other inflammatory components, such as CD4+ T-cells, macrophages,

cytokines and integrins, have also been observed in the perifollicular area of uninvolved skin

from acne patients. Recent in vitro studies have revealed that P. acnes can induce IL-17

P T
production by PBMC 114 and T-cells (Th1/Th17) 115, and that IL-17-expressing cells are present

RI
in acne biopsies but not detectable in healthy controls 114. P. acnes can activate monocytes via

SC
TLR2 or TLR4, and these receptors are co-localized with CD14 in acne lesions but not in

controls 116.

NU
MA
The activation of TLR2 induces the production of pro-inflammatory cytokines including IL-1,

TNF-α, prostaglandins, leukotrienes and chemokines (IL-8). An increased expression of TLR2


ED

and TLR4 was detected in the epidermis of inflammatory acne lesions, and this was more intense

with TLR2 than with TLR4, as observed in normal skin; the expression level increased with the
PT

degree of differentiation of the keratinocytes 117. P. acnes can also trigger the activation of the
CE

NLRP3-inflammasome pathway in antigen-presenting cells 118 and myeloid cells via IL-1β 119.

Expression of the G-coupled protein receptor PAR-2 was also shown to be increased in
AC

keratinocytes from acne 120, and this was correlated with a production of IL-1α, β-defensin 2,

antimicrobial peptide LL-37 and different matrix metalloproteinases.

Studies of the evolution of acne lesions in vivo have shown that the initial infiltrate consisted of

mononuclear cells, predominantly CD4+ T cells 121. Neutrophils could be seen appearing in some

of the later lesions (72h), and were associated with disruption of the duct. The study of gene

expression by array profiling on skin biopsies from unaffected control, normal skin of acne

patients, and from inflammatory papules of the same patients showed 211 genes upregulated in
122
acne lesions when compared to controls . Most of these genes were involved in inflammation
24
ACCEPTED MANUSCRIPT
O. Alexeyev

and matrix remodeling, including matrix metalloproteinases, IL-8, β-defensin 4, and granulysin.
113
Jeremy et al. showed the presence of elevated numbers of macrophages and T-cells in

perifollicular and papillary dermis of uninvolved acne skin compared to control skin in the

P T
absence of keratinocytes hyperproliferation.

RI
All these findings suggest that inflammation and not keratinocyte hyperproliferation is the prime

SC
event in acne pathogenesis, placing the role of P. acnes in the later stages of inflammation 6. The

NU
major limitation of these studies is, however, a lack of information on the presence or absence of

P. acnes in acne lesions. We previously demonstrated clusters of CD3+ cells in the vicinity of the
MA
P. acnes-positive comedones, but those cells were virtually absent from the surroundings of P.
63
acnes-positive inflamed lesions . On this basis, the co-localization of P. acnes and CD3+
ED

lymphocytes in early acne stage may actually suggest a role of P. acnes in the initiation of the
PT

inflammation.
CE

Conclusio

Over the years, a large number of studies have examined the relationship between P. acnes and
AC

acne vulgaris. Despite this, the role played by the bacterium in the condition still remains unclear,

especially as various investigations have found no difference in the counts of P. acnes on the skin

of patients with acne versus healthy subjects, alongside the detection of inflammation in lesions

apparently devoid of bacteria. While such studies have appeared to question the importance of P.

acnes in acne development, we would argue that in many cases they may not have been based on

the most appropriate sampling methods we described earlier, making the interpretation and

relevance of the results unclear. In the future, consideration of such methods when designing

25
ACCEPTED MANUSCRIPT
O. Alexeyev

experiments aimed at understanding the role of P. acnes in acne development should be a critical

component of any study.

P T
RI
SC
NU
MA
ED
PT
CE
AC

26
ACCEPTED MANUSCRIPT
O. Alexeyev

Figure legends

Figure 1.

P T
eBURST population snapshot of P. acnes based on data from the P. acnes MLST8 database

RI
(http://pubmlst.org/pacnes/). A total of nine clonal complexes, where the isolates share 7/8 loci

SC
with at least one other ST in the group, and 21 singletons are illustrated. The frequency of each

ST within the isolate database is indicated by the size of each circle. Founding genotypes are

NU
highlighted in blue and sub-founders in yellow. Note, the spacing between the singletons and
MA
clonal complexes is not related to the genetic distance between them. Adapted from McDowell et

al. 22.
ED

Figure 2.
PT

P. acnes biofilm within a hair follicle (stained by IFM using specific monoclonal anti-P. acnes
CE

antibodies).
AC

Table 1.

Overview of various P. acnes skin populations targeted by different sampling methods

(adapted from Alexeyev et al. 45).

Table 2.

Physiological states of bacteria found on human skin (adapted from Alexeyev et al. 65).

27
ACCEPTED MANUSCRIPT
O. Alexeyev

References

1) Williams HC, Dellavalle RP, Garner S. Acne vulgaris. Lancet. 2012; 379:361-372.

2) Bhate K, Williams HC. Epidemiology of acne vulgaris. Br J Dermatol. 2013; 168:474-485.

P T
3) Dreno B, Thiboutot D, Layton AM, et al. Large-scale international study enhances understanding

RI
of an emerging acne population: adult females. J Eur Acad Dermatol Venereol. 2015; 29:1096-1106.

SC
4) Hay RJ, Johns NE, Williams HC, et al. The global burden of skin disease in 2010: an analysis of the

prevalence and impact of skin conditions. J Invest Dermatol. 2014; 134:1527-1534.

NU
5) Kligman AM. An overview of acne. J Invest Dermatol. 1974; 62:268-287.

6) Tanghetti EA. The role of inflammation in the pathology of acne. The Journal of clinical and
MA
aesthetic dermatology. 2013; 6:27-35.

7) Dessinioti C, Katsambas AD. The role of Propionibacterium acnes in acne pathogenesis: facts and
ED

controversies. Clin Dermatol. 2010; 28:2-7.


PT

8) Shaheen B, Gonzalez M. A microbial aetiology of acne: what is the evidence? Br J Dermatol.

2011; 165:474-485.
CE

9) Otberg N, Richter H, Schaefer H, et al. Variations of hair follicle size and distribution in different
AC

body sites. J Invest Dermatol. 2004; 122:14-19.

10) Gordon CR, Zor F, Siemionow M. Skin area quantification in preparation for concomitant upper

extremity and face transplantation: a cadaver study and literature review. Transplantation. 2011;

91:1050-1056.

11) Ramli R, Malik AS, Hani AF, et al. Acne analysis, grading and computational assessment methods:

an overview. Skin research and technology : official journal of International Society for Bioengineering

and the Skin. 2012; 18:1-14.

28
ACCEPTED MANUSCRIPT
O. Alexeyev

12) Johnson JL, Cummins CS. Cell wall composition and deoxyribonucleic acid similarities among the

anaerobic coryneforms, classical propionibacteria, and strains of Arachnia propionica. Journal of

bacteriology. 1972; 109:1047-1066.

P T
13) Webster GF, Cummins CS. Use of bacteriophage typing to distinguish Propionibacterium acne

RI
types I and II. J Clin Microbiol. 1978; 7:84-90.

SC
14) McDowell A, Valanne S, Ramage G, et al. Propionibacterium acnes types I and II represent

phylogenetically distinct groups. J Clin Microbiol. 2005; 43:326-334.

NU
15) McDowell A, Perry AL, Lambert PA, et al. A new phylogenetic group of Propionibacterium acnes. J

Med Microbiol. 2008; 57:218-224.


MA
16) Lomholt HB, Kilian M. Population genetic analysis of Propionibacterium acnes identifies a

subpopulation and epidemic clones associated with acne. PLoS One. 2010; 5:e12277.
ED

17) McDowell A, Barnard E, Nagy I, et al. An expanded multilocus sequence typing scheme for
PT

propionibacterium acnes: investigation of 'pathogenic', 'commensal' and antibiotic resistant strains. PLoS

One. 2012; 7:e41480.


CE

18) Fitz-Gibbon S, Tomida S, Chiu BH, et al. Propionibacterium acnes strain populations in the human
AC

skin microbiome associated with acne. J Invest Dermatol. 2013; 133:2152-2160.

19) Scholz CF, Jensen A, Lomholt HB, et al. A novel high-resolution single locus sequence typing

scheme for mixed populations of Propionibacterium acnes in vivo. PLoS One. 2014; 9:e104199.

20) Dekio I, Culak R, Misra R, et al. Dissecting the taxonomic heterogeneity within Propionibacterium

acnes: proposal for Propionibacterium acnes subsp. acnes subsp. nov. and Propionibacterium acnes

subsp. elongatum subsp. nov. International journal of systematic and evolutionary microbiology. 2015;

65:4776-4787.

21) Valanne S, McDowell A, Ramage G, et al. CAMP factor homologues in Propionibacterium acnes: a

new protein family differentially expressed by types I and II. Microbiology. 2005; 151:1369-1379.
29
ACCEPTED MANUSCRIPT
O. Alexeyev

22) McDowell A, Nagy I, Magyari M, et al. The opportunistic pathogen Propionibacterium acnes:

insights into typing, human disease, clonal diversification and CAMP factor evolution. PLoS One. 2013;

8:e70897.

P T
23) McDowell A, Gao A, Barnard E, et al. A novel multilocus sequence typing scheme for the

RI
opportunistic pathogen Propionibacterium acnes and characterization of type I cell surface-associated

SC
antigens. Microbiology. 2011; 157:1990-2003.

24) Johnson T, Kang D, Barnard E, et al. Strain-Level Differences in Porphyrin Production and

NU
Regulation inPropionibacterium acnesElucidate Disease Associations. mSphere. 2016; 1:e00023-00015.

25) Brzuszkiewicz E, Weiner J, Wollherr A, et al. Comparative genomics and transcriptomics of


MA
Propionibacterium acnes. PLoS One. 2011; 6:e21581.

26) Tomida S, Nguyen L, Chiu BH, et al. Pan-genome and comparative genome analyses of
ED

propionibacterium acnes reveal its genomic diversity in the healthy and diseased human skin
PT

microbiome. mBio. 2013; 4:e00003-00013.

27) Scholz CF, Bruggemann H, Lomholt HB, et al. Genome stability of Propionibacterium acnes: a
CE

comprehensive study of indels and homopolymeric tracts. Scientific reports. 2016; 6:20662.
AC

28) Bruggemann H, Lomholt HB, Kilian M. The flexible gene pool of Propionibacterium acnes. Mobile

genetic elements. 2012; 2:145-148.

29) Bruggemann H, Lomholt HB, Tettelin H, et al. CRISPR/cas loci of type II Propionibacterium acnes

confer immunity against acquisition of mobile elements present in type I P. acnes. PLoS One. 2012;

7:e34171.

30) Kasimatis G, Fitz-Gibbon S, Tomida S, et al. Analysis of complete genomes of Propionibacterium

acnes reveals a novel plasmid and increased pseudogenes in an acne associated strain. BioMed research

international. 2013; 2013:918320.

30
ACCEPTED MANUSCRIPT
O. Alexeyev

31) Holland C, Mak TN, Zimny-Arndt U, et al. Proteomic identification of secreted proteins of

Propionibacterium acnes. BMC Microbiol. 2010; 10:230.

32) Niazi SA, Clarke D, Do T, et al. Propionibacterium acnes and Staphylococcus epidermidis isolated

P T
from refractory endodontic lesions are opportunistic pathogens. J Clin Microbiol. 2010; 48:3859-3869.

RI
33) Lodes MJ, Secrist H, Benson DR, et al. Variable expression of immunoreactive surface proteins of

SC
Propionibacterium acnes. Microbiology. 2006; 152:3667-3681.

34) Tilles G. Acne pathogenesis: history of concepts. Dermatology. 2014; 229:1-46.

NU
35) Dreno B, Gollnick HP, Kang S, et al. Understanding innate immunity and inflammation in acne:

implications for management. J Eur Acad Dermatol Venereol. 2015; 29 Suppl 4:3-11.
MA
36) Suh DH, Kwon HH. What's new in the physiopathology of acne? Br J Dermatol. 2015; 172 Suppl

1:13-19.
ED

37) Dreno B. The changing faces of acne. Br J Dermatol. 2015; 172 Suppl 1:1-2.
PT

38) Beylot C, Auffret N, Poli F, et al. Propionibacterium acnes: an update on its role in the

pathogenesis of acne. J Eur Acad Dermatol Venereol. 2014; 28:271-278.


CE

39) Harvey A, Huynh TT. Inflammation and acne: putting the pieces together. Journal of drugs in
AC

dermatology : JDD. 2014; 13:459-463.

40) Williamson P, Kligman AM. A new method for the quantitative investigation of cutaneous

bacteria. J Invest Dermatol. 1965; 45:498-503.

41) McGinley KJ, Webster GF, Leyden JJ. Regional variations of cutaneous propionibacteria. Appl

Environ Microbiol. 1978; 35:62-66.

42) Jahns AC, Lundskog B, Ganceviciene R, et al. An increased incidence of Propionibacterium acnes

biofilms in acne vulgaris: a case-control study. Br J Dermatol. 2012; 167:50-58.

43) Grice EA, Segre JA. The skin microbiome. Nature reviews. 2011; 9:244-253.

31
ACCEPTED MANUSCRIPT
O. Alexeyev

44) Mollerup S, Friis-Nielsen J, Vinner L, et al. Propionibacterium acnes: Disease-Causing Agent or

Common Contaminant? Detection in Diverse Patient Samples by Next-Generation Sequencing. J Clin

Microbiol. 2016; 54:980-987.

P T
45) Alexeyev OA, Jahns AC. Sampling and detection of skin Propionibacterium acnes: current status.

RI
Anaerobe. 2012; 18:479-483.

SC
46) Gehse M, Hoffler U, Gloor M, et al. Propionibacteria in patients with acne vulgaris and in healthy

persons. Arch Dermatol Res. 1983; 275:100-104.

NU
47) Panpradist N, Toley BJ, Zhang X, et al. Swab sample transfer for point-of-care diagnostics:

characterization of swab types and manual agitation methods. PLoS One. 2014; 9:e105786.
MA
48) Harry Kathryn H, Turner Julie C , . MKT. Comparison of physical characteristics and collection and

elution performance of clinical swabs. African Journal of Microbiology Research. 2013; 7:4039-4048.
ED

49) Keyworth N, Millar MR, Holland KT. Swab-wash method for quantitation of cutaneous
PT

microflora. J Clin Microbiol. 1990; 28:941-943.

50) Pinkus H. Examination of the epidermis by the strip method of removing horny layers. I.
CE

Observations on thickness of the horny layer, and on mitotic activity after stripping. J Invest Dermatol.
AC

1951; 16:383-386.

51) Updegraff DM. A Cultural Method of Quantitatively Studying the Microorganisms in the Skin. J

Invest Dermatol. 1964; 43:129-137.

52) Holland KT, Roberts CD. A technique for sampling microorganisms from the pilo-sebaceous

ducts. J Appl Bacteriol. 1974; 37:289-296.

53) Forster M, Bolzinger MA, Rovere MR, et al. Confocal Raman microspectroscopy for evaluating

the stratum corneum removal by 3 standard methods. Skin Pharmacol Physiol. 2011; 24:103-112.

54) Dossi M, Storti G, Moscatelli D. Synthesis of Poly(Alkyl Cyanoacrylates) as Biodegradable

Polymers for Drug Delivery Applications. Macromol Symp. 2010; 289:124-128.


32
ACCEPTED MANUSCRIPT
O. Alexeyev

55) Plewig G. Follicular keratinization. J Invest Dermatol. 1974; 62:308-320.

56) Miura Y, Ishige I, Soejima N, et al. Quantitative PCR of Propionibacterium acnes DNA in samples

aspirated from sebaceous follicles on the normal skin of subjects with or without acne. J Med Dent Sci.

P T
2010; 57:65-74.

RI
57) Craft N, Li H. Response to the commentaries on the paper: Propionibacterium acnes strain

SC
populations in the human skin microbiome associated with acne. J Invest Dermatol. 2013; 133:2295-

2297.

NU
58) Puhvel SM, Reisner RM, Amirian DA. Quantification of bacteria in isolated pilosebaceous follicles

in normal skin. J Invest Dermatol. 1975; 65:525-531.


MA
59) Kellum RE. Isolation of human sebaceous glands. Arch Dermatol. 1966; 93:610-612.

60) Imamura S, Pochi PE, Strauss JS, et al. The localization and distribution of Corynebacterium acnes
ED

and its antigens in normal skin and in lesions of acne vulgaris. J Invest Dermatol. 1969; 53:143-150.
PT

61) Amann R, Fuchs BM. Single-cell identification in microbial communities by improved

fluorescence in situ hybridization techniques. Nature reviews. 2008; 6:339-348.


CE

62) Moter A, Gobel UB. Fluorescence in situ hybridization (FISH) for direct visualization of
AC

microorganisms. J Microbiol Methods. 2000; 41:85-112.

63) Alexeyev OA, Lundskog B, Ganceviciene R, et al. Pattern of tissue invasion by Propionibacterium

acnes in acne vulgaris. J Dermatol Sci. 2012; 67:63-66.

64) Jahns AC, Alexeyev OA. Three dimensional distribution of Propionibacterium acnes biofils in

human skin. Exp Dermatol. 2014; 23:687-689.

65) Alexeyev OA. Bacterial landscape of human skin: seeing the forest for the trees. Exp Dermatol.

2013; 22:443-446.

66) Lorenz MG, Wackernagel W. Bacterial gene transfer by natural genetic transformation in the

environment. Microbiol Rev. 1994; 58:563-602.


33
ACCEPTED MANUSCRIPT
O. Alexeyev

67) Trampuz A, Piper KE, Jacobson MJ, et al. Sonication of removed hip and knee prostheses for

diagnosis of infection. N Engl J Med. 2007; 357:654-663.

68) Numata S, Akamatsu H, Akaza N, et al. Analysis of facial skin-resident microbiota in Japanese

P T
acne patients. Dermatology. 2014; 228:86-92.

RI
69) McGinley KJ, Webster GF, Ruggieri MR, et al. Regional variations in density of cutaneous

SC
propionibacteria: correlation of Propionibacterium acnes populations with sebaceous secretion. J Clin

Microbiol. 1980; 12:672-675.

NU
70) Akaza N, Akamatsu H, Numata S, et al. Microorganisms inhabiting follicular contents of facial

acne are not only Propionibacterium but also Malassezia spp. J Dermatol. 2015.
MA
71) Leyden JJ, Mcginley KJ, Mills OH, et al. Propionibacterium Levels in Patients with and without

Acne-Vulgaris. Journal of Investigative Dermatology. 1975; 65:382-384.


ED

72) Kishishita M, Ushijima T, Ozaki Y, et al. Biotyping of Propionibacterium acnes isolated from
PT

normal human facial skin. Appl Environ Microbiol. 1979; 38:585-589.

73) Higaki S, Kitagawa T, Kagoura M, et al. Correlation between Propionibacterium acnes biotypes,
CE

lipase activity and rash degree in acne patients. J Dermatol. 2000; 27:519-522.
AC

74) Giannopoulos L, Papaparaskevas J, Refene E, et al. MLST typing of antimicrobial-resistant

Propionibacterium acnes isolates from patients with moderate to severe acne vulgaris. Anaerobe. 2015;

31:50-54.

75) Gloor M, Franke M. On the propionibacteria in the pilosebaceous ducts of uninvolved skin of

acne patients. Arch Dermatol Res. 1978; 262:125-129.

76) Lavker RM, Leyden JJ, McGinley KJ. The relationship between bacteria and the abnormal

follicular keratinization in acne vulgaris. J Invest Dermatol. 1981; 77:325-330.

77) Till AE, Goulden V, Cunliffe WJ, et al. The cutaneous microflora of adolescent, persisten and late-

onset acne patients does not differ. Br J Dermatol. 2000; 142:885-892.


34
ACCEPTED MANUSCRIPT
O. Alexeyev

78) Bek-Thomsen M, Lomholt HB, Kilian M. Acne is not associated with yet-uncultured bacteria. J

Clin Microbiol. 2008; 46:3355-3360.

79) Bek-Thomsen M, Lomholt HB, Scavenius C, et al. Proteome analysis of human sebaceous follicle

P T
infundibula extracted from healthy and acne-affected skin. PLoS One. 2014; 9:e107908.

RI
80) Marples RR, McGinley KJ, Mills OH. Microbiology of comedones in acne vulgaris. J Invest

SC
Dermatol. 1973; 60:80-83.

81) Smith MA, Waterworth PM. The Bacteriology of Acne Vulgaris in Relation to Its Treatment with

NU
Antibiotics. British Journal of Dermatology. 1961; 73:152-159.

82) Ganor S, Sacks TG. A comparison of the flora of the comedones of acne vulgaris and comedones
MA
in elderly people. Dermatologica. 1969; 138:1-9.

83) Shehadeh NH, Kligman AM. The Bacteriology of Acne. Arch Dermatol. 1963; 88:829-831.
ED

84) Nishijima S, Kurokawa I, Katoh N, et al. The bacteriology of acne vulgaris and antimicrobial
PT

susceptibility of Propionibacterium acnes and Staphylococcus epidermidis isolated from acne lesions. J

Dermatol. 2000; 27:318-323.


CE

85) Leeming JP, Holland KT, Cunliffe WJ. The pathological and ecological significance of
AC

microorganisms colonizing acne vulgaris comedones. J Med Microbiol. 1985; 20:11-16.

86) Cunliffe WJ, Holland DB, Jeremy A. Comedone formation: etiology, clinical presentation, and

treatment. Clin Dermatol. 2004; 22:367-374.

87) Puhvel SM, Amirian DA. Bacterial flora of comedones. Br J Dermatol. 1979; 101:543-548.

88) Leeming JP, Holland KT, Cuncliffe WJ. The microbial colonization of inflamed acne vulgaris

lesions. Br J Dermatol. 1988; 118:203-208.

89) Costerton JW, Stewart PS, Greenberg EP. Bacterial biofilms: a common cause of persistent

infections. Science. 1999; 284:1318-1322.

90) Watnick P, Kolter R. Biofilm, city of microbes. Journal of bacteriology. 2000; 182:2675-2679.
35
ACCEPTED MANUSCRIPT
O. Alexeyev

91) Olsen I. Biofilm-specific antibiotic tolerance and resistance. European journal of clinical

microbiology & infectious diseases : official publication of the European Society of Clinical Microbiology.

2015; 34:877-886.

P T
92) Jolivet-Gougeon A, Bonnaure-Mallet M. Biofilms as a mechanism of bacterial resistance. Drug

RI
discovery today Technologies. 2014; 11:49-56.

SC
93) Høiby N, Bjarnsholt T, Givskov M, et al. Antiobiotic resistance of bacterial biofilms. Int J

Antimicrob Agents. 2010; 35:322-332.

NU
94) Conlon BP, Rowe SE, Lewis K. Persister cells in biofilm associated infections. Advances in

experimental medicine and biology. 2015; 831:1-9.


MA
95) Parsek MR, Singh PK. Bacterial biofilms: an emerging link to disease pathogenesis. Annu Rev

Microbiol. 2003; 57:677-701.


ED

96) Mihai MM, Holban AM, Giurcaneanu C, et al. Microbial biofilms: impact on the pathogenesis of
PT

periodontitis, cystic fibrosis, chronic wounds and medical device-related infections. Current topics in

medicinal chemistry. 2015; 15:1552-1576.


CE

97) Vlassova N, Han A, Zenilman JM, et al. New horizons for cutaneous microbiology: the role of
AC

biofilms in dermatological disease. Br J Dermatol. 2011; 165:751-759.

98) Tunney MM, Patrick S, Curran MD, et al. Detection of prosthetic hip infection at revision

arthroplasty by immunofluorescence microscopy and PCR amplification of the bacterial 16S rRNA gene.

Journal of Clinical Microbiology. 1999; 37:3281-3290.

99) Ramage G, Tunney MM, Patrick S, et al. Formation of Propionibacterium acnes biofilms on

orthopaedic biomaterials and their susceptibility to antimicrobials. Biomaterials. 2003; 24:3221-3227.

100) Bruggemann H, Henne A, Hoster F, et al. The complete genome sequence of Propionibacterium

acnes, a commensal of human skin. Science. 2004; 305:671-673.

36
ACCEPTED MANUSCRIPT
O. Alexeyev

101) Bruggemann H. Insights in the pathogenic potential of Propionibacterium acnes from its

complete genome. Semin Cutan Med Surg. 2005; 24:67-72.

102) Bayston R, Ashraf W, Barker-Davies R, et al. Biofilm formation by Propionibacterium acnes on

P T
biomaterials in vitro and in vivo: impact on diagnosis and treatment. Journal of biomedical materials

RI
research Part A. 2007; 81:705-709.

SC
103) Bjerkan G, Witso E, Bergh K. Sonication is superior to scraping for retrieval of bacteria in biofilm

on titanium and steel surfaces in vitro. Acta Orthopaedica. 2009; 80:245-250.

NU
104) Coenye T, Brackman G, Rigole P, et al. Eradication of Propionibacterium acnes biofilms by plant

extracts and putative identification of icariin, resveratrol and salidroside as active compounds.
MA
Phytomedicine : international journal of phytotherapy and phytopharmacology. 2012; 19:409-412.

105) Tunney MM, Dunne N, Einarsson G, et al. Biofilm formation by bacteria isolated from retrieved
ED

failed prosthetic hip implants in an in vitro model of hip arthroplasty antibiotic prophylaxis. Journal of
PT

orthopaedic research : official publication of the Orthopaedic Research Society. 2007; 25:2-10.

106) Coenye T, Peeters E, Nelis HJ. Biofilm formation by Propionibacterium acnes is associated with
CE

increased resistance to antimicrobial agents and increased production of putative virulence factors.
AC

Research in Microbiology. 2007; 158:386-392.

107) Jahns AC, Lundskog B, Berg J, et al. Microbiology of folliculitis: a histological study of 39 cases.

APMIS. 2014; 122:25-32.

108) Jahns AC, Lundskog B, Nosek D, et al. Microbiology of folliculitis decalvans: a histological study of

37 patients. J Eur Acad Dermatol Venereol. 2015; 29:1025-1026.

109) Jahns AC, Killasli H, Nosek D, et al. Microbiology of hidradenitis suppurativa (acne inversa): a

histological study of 27 patients. APMIS. 2014; 122:804-809.

37
ACCEPTED MANUSCRIPT
O. Alexeyev

110) Graham GM, Farrar MD, Cruse-Sawyer JE, et al. Proinflammatory cytokine production by human

keratinocytes stimulated with Propionibacterium acnes and P. acnes GroEL. Br J Dermatol. 2004;

150:421-428.

P T
111) Ingham E, Eady EA, Goodwin CE, et al. Pro-inflammatory levels of interleukin-1 alpha-like

RI
bioactivity are present in the majority of open comedones in acne vulgaris. J Invest Dermatol. 1992;

SC
98:895-901.

112) Walters CE, Ingham E, Eady EA, et al. In vitro modulation of keratinocyte-derived interleukin-1

NU
alpha (IL-1 alpha) and peripheral blood mononuclear cell-derived IL-1 beta release in response to

cutaneous commensal microorganisms. Infect Immun. 1995; 63:1223-1228.


MA
113) Jeremy AH, Holland DB, Roberts SG, et al. Inflammatory events are involved in acne lesion

initiation. J Invest Dermatol. 2003; 121:20-27.


ED

114) Agak GW, Qin M, Nobe J, et al. Propionibacterium acnes Induces an IL-17 Response in Acne
PT

Vulgaris that Is Regulated by Vitamin A and Vitamin D. J Invest Dermatol. 2014; 134:366-373.

115) Kistowska M, Meier B, Proust T, et al. Propionibacterium acnes promotes Th17 and Th17/Th1
CE

responses in acne patients. J Invest Dermatol. 2015; 135:110-118.


AC

116) Kim J, Ochoa MT, Krutzik SR, et al. Activation of toll-like receptor 2 in acne triggers inflammatory

cytokine responses. Journal of immunology. 2002; 169:1535-1541.

117) Jugeau S, Tenaud I, Knol AC, et al. Induction of toll-like receptors by Propionibacterium acnes. Br

J Dermatol. 2005; 153:1105-1113.

118) Qin M, Pirouz A, Kim MH, et al. Propionibacterium acnes Induces IL-1beta secretion via the

NLRP3 inflammasome in human monocytes. J Invest Dermatol. 2014; 134:381-388.

119) Kistowska M, Gehrke S, Jankovic D, et al. IL-1beta drives inflammatory responses to

propionibacterium acnes in vitro and in vivo. J Invest Dermatol. 2014; 134:677-685.

38
ACCEPTED MANUSCRIPT
O. Alexeyev

120) Lee SE, Kim JM, Jeong SK, et al. Protease-activated receptor-2 mediates the expression of

inflammatory cytokines, antimicrobial peptides, and matrix metalloproteinases in keratinocytes in

response to Propionibacterium acnes. Arch Dermatol Res. 2010; 302:745-756.

P T
121) Norris JF, Cunliffe WJ. A histological and immunocytochemical study of early acne lesions. Br J

RI
Dermatol. 1988; 118:651-659.

SC
122) Trivedi NR, Gilliland KL, Zhao W, et al. Gene array expression profiling in acne lesions reveals

marked upregulation of genes involved in inflammation and matrix remodeling. J Invest Dermatol. 2006;

NU
126:1071-1079.
MA
ED
PT
CE
AC

39
ACCEPTED MANUSCRIPT
O. Alexeyev

Figure 1.

PT
RI
SC
NU
MA
ED
PT
CE
AC

40
ACCEPTED MANUSCRIPT
O. Alexeyev

Figure 2.

PT
RI
SC
NU
MA
ED
PT
CE
AC

41
ACCEPTED MANUSCRIPT
O. Alexeyev

Table 1.

P T
P. acnes population Sampling method

RI
Swab Scrape Cyanoacrylate gel Biopsy

SC
Superficial stratum + + + -a
corneum

NU
Within stratum corneum - + + +
Infudibulum - -
MA + +
Lower hair follicle - - - +
a
Likely removed during pre-treatment with antiseptics.
ED
PT
CE
AC

42
ACCEPTED MANUSCRIPT
O. Alexeyev

Table 2.

P T
Physiological state Phenotype

RI
Viable (high metabolic Can proliferate and form colonies on culture media
activity) (if available)

SC
Dormant (low metabolic Temporarily lost or markedly diminished capability
activity) to proliferate and form colonies on culture media

NU
Non-viable (dead) Permanently lost capability to proliferate and form
colonies on culture media
MA
ED
PT
CE
AC

43

You might also like