Next Article in Journal
Molecular Epidemiology of Penicillin-Susceptible Staphylococcus aureus Bacteremia in Australia and Reliability of Diagnostic Phenotypic Susceptibility Methods to Detect Penicillin Susceptibility
Next Article in Special Issue
Potential Association of Cutibacterium acnes with Sarcoidosis as an Endogenous Hypersensitivity Infection
Previous Article in Journal
Differed Growth Stage Dynamics of Root-Associated Bacterial and Fungal Community Structure Associated with Halophytic Plant Lycium ruthenicum
Previous Article in Special Issue
A Potential Predictive Role of the Scalp Microbiome Profiling in Patients with Alopecia Areata: Staphylococcus caprae, Corynebacterium, and Cutibacterium Species
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Cutibacterium acnes in Sarcoidosis: From Antigen to Treatable Trait?

by
Raisa Kraaijvanger
1 and
Marcel Veltkamp
1,2,*
1
Interstitial Lung Diseases Centre of Excellence, Department of Pulmonology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
2
Division of Hearth and Lungs, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
*
Author to whom correspondence should be addressed.
Microorganisms 2022, 10(8), 1649; https://doi.org/10.3390/microorganisms10081649
Submission received: 6 July 2022 / Revised: 4 August 2022 / Accepted: 8 August 2022 / Published: 15 August 2022
(This article belongs to the Special Issue Infection, Immunity, and Allergy Caused by Propionibacterium acnes)

Abstract

:
Cutibacterium acnes (C. acnes, formerly Propionibacterium acnes) is considered to be a non-pathogenic resident of the human skin, as well as mucosal surfaces. However, it also has been demonstrated that C. acnes plays a pathogenic role in diseases such as acne vulgaris or implant infections after orthopedic surgery. Besides a role in infectious disease, this bacterium also seems to harbor immunomodulatory effects demonstrated by studies using C. acnes to enhance anti-tumor activity in various cancers or vaccination response. Sarcoidosis is a systemic inflammatory disorder of unknown causes. Cultures of C. acnes in biopsy samples of sarcoidosis patients, its presence in BAL fluid, tissue samples as well as antibodies against this bacterium found in serum of patients with sarcoidosis suggest an etiological role in this disease. In this review we address the antigenic as well as immunomodulatory potential of C. acnes with a focus on sarcoidosis. Furthermore, a potential role for antibiotic treatment in patients with sarcoidosis will be explored.

1. Introduction

Cutibacterium acnes (formerly Propionibacterium acnes) (C. acnes) is an anaerobic-aerotolerant Gram-positive, rod shaped bacterium and commensal of the human skin, oral cavity, large intestine, conjunctiva and the external ear canal [1,2,3]. The fact that the human defense system allows C. acnes to colonize the body on several levels suggests that this bacterium, at least under normal circumstances, is not harmful for the human host. Based on serology, two phenotypes of C. acnes have been distinguished: type I and type II. Later on, phylogenetic analysis based on nucleotide sequencing of a nonribosomal housekeeping gene (recA) and a putative hemolysin/cytotoxin gene (tly) showed that type I and type II represented different groups and that type I could be subdivided in type IA1, IA2, IB and IC [4,5]. More recently, a third phenotype was identified and was designated as type III [6]. These different C. acnes strains differ in their induction of proinflammatory cytokines and their ability to induce an immune responses [2].

2. C. acnes and Different Diseases

Traditionally, C. acnes has been considered as a non-pathogenic resident of the human microbiota. Over the years, however, C. acnes has also been suspected to plays a role in various disorders such as acne vulgaris (Figure 1). Already in 1896 C. acnes was associated with acne vulgaris and now its etiological role is widely accepted, even though the precise mechanisms are yet to be fully elucidated [2]. C. acnes has a prominent part in the normal human skin flora, where it plays a critical role in the regulation of skin homeostasis and preventing colonization of other harmful pathogens [2,7]. Comparing healthy skin with skin affected by acne vulgaris revealed that there was no difference in the load of C. acnes [8]. Based on these data it was hypothesized that inflammatory skin disorders may not result from C. acnes acting as an opportunistic pathogen, but rather the imbalance in skin microbiota may play a critical role [7,8]. Treatment of acne vulgaris comes with complications. Current treatments are often difficult to tolerate or are inadequate to inhibit the inflammatory effect. Isotretinoin, for example, is a treatment extensively prescribed for the treatment of severe acne, but can cause depression and comes with an increased risk of birth defects when taken during pregnancy [9,10]. Moreover, the use of antibiotics as treatment for acne vulgaris may result in resistance development of the C. acnes bacterium [11]. In 2018, Wang et al. proposed the use of a vaccination with the C. acnes Christie-Atkins-Munch-Petersen (CAMP) factor, a virulence factor triggering cell death, to potentially decrease C. acnes-induced inflammation [12]. With the use of a mouse model it was shown that this vaccination increased protective immunity against C. acnes. In mice, a decreased C. acnes colonization and reduced production of a proinflammatory MIP-2 cytokine was shown after vaccination [12].
C. acnes not only seems to play a role in inflammation of the skin but also in infections of the heart, the gastrointestinal tract and also in orthopedic surgery, particularly in relation to implants [3,13,14]. In the last decade, an increased number of studies have found an association between C. acnes and a wide range of implant-associated infections (IAI) [15]. C. acnes is the cause of various post-surgical associated infections, including breast implants [16,17], ocular implants, neurosurgical shunts [18], spinal hardware, cardiovascular devices [13] and prosthetic joints [19]. Of all implant-associated infections caused by C. acnes, shoulder prosthetic joints are most often infected. This is probably due to greater colonization in the shoulder and armpit than for example in the hip or knee [14,20]. Once C. acnes is settled on the prosthesis it can form a biofilm, which needs to be fully eliminated in order to cure the infection. The formation of a biofilm is a pertinacious trait and often requires the full replacement of all implants [20].
Moreover, it has also been shown that C. acnes is associated with the inflammation of the prostate as well as prostate cancer. A study from Sweden showed that C. acnes could be cultured in 60% of the prostate cancer cases and in 26% of the cancer-free controls [21], whereas a French study detected only very few C. acnes positive samples in their cohort [22]. However, based on the biopsy procedure involving antibiotic prophylaxis, these results are difficult to interpret. It is now hypothesized that intraepithelial C. acnes infection in non-cancerous prostate glands resulting in excessive inflammation may contribute to the development of prostate cancer. Furthermore, in patients diagnosed with prostate cancer, this infiltration is associated with a worse prognosis [23,24,25].

3. Immunomodulatory Effect of C. acnes

3.1. Anti-Tumor Effect

In addition to the fact that C. acnes can be seen as an infectious agent, it is also capable of modulating the immune system resulting in an anti-tumor activity against various animal and human cancers [26,27,28]. The anti-tumor effect of C. acnes has been reported in multiple studies of various types of cancer, such as malignant melanoma [29] and breast cancer [28]. However, C. acnes is not the only bacterial strain which has an anti-tumor and immune-modulatory effect. Several bacteria such as Clostridium novyi [30], Bifidobacterium longum [31] and Salmonella typhi [32] have also been shown to have this effect.
C. acnes can modulate a variety of cellular functions of lymphocytes and macrophages which have been shown to be beneficial for an anti-tumor response (Figure 2). An important example of such modulation is the induction of a Th1 response, resulting in the activation of macrophages and natural killer (NK) cells [33,34,35]. Moreover, it has also been shown that C. acnes is able to skew a Th2 response towards a Th1 response. A Th2 environment is favorable in tumor formation based on the fact that there is less activity of cytotoxic T-cells and NK cells [36]. Using a mouse model for melanoma and breast cancer it was shown that C. acnes infection resulted in a switch in the immune response toward a Th1 response, both locally in the tumor as well as in the peripheral blood compartment. This resulted in an increased infiltration of T-cells into the tumor lesions of these mice [28,29]. Upon activation by the C. acnes, NK cells infiltrate the tumor after being attracted by tumor-secreted cytokines and cell-death signals. Dendritic cells, in their turn, leave the cancer tissue and migrate to the draining lymph nodes, where they present captured antigens to naïve and memory T-cells [37]. The phagocytosis of C. acnes by dendritic cells results in a Th1-type cytokine immune response and causes the production of IFN-γ, TNF-α and IL-12 [28].
The anti-tumor effect is not induced by all types of C. acnes. Mouse model studies showed that the injection of C. acnes type I resulted in survival of 60 to 100% of the cases, while type II did not show this same anti-tumor effect [38,39]. The difference in anti-tumor activity might be explained by the difference in the carbohydrate composition of the cell walls of the different types of C. acnes resulting in different immunological and biological activity [40]. Moreover, the anti-tumor effect of type I strains might also be due to the more potent resistance of the strains to degradation by phagocytes than type II strains [41]. Interestingly, C. acnes not only showed an anti-tumor effect, but also showed an anti-metastatic effect by inhibiting the spread and growth of metastasized tumor cells [42].

3.2. C. acnes as a Vaccine Adjuvant

Based on its immunomodulatory capacity, C. acnes has also been studied regarding a potential beneficial role as a vaccine adjuvant in infectious disease (Figure 2). Due to the cytokine pattern mediated by C. acnes, its heat-killed suspension has been used as a Th1 response inducer to enhance the efficacy of a HIV vaccine [43]. It was hypothesized that the C. acnes modulation of the immune system probably occurs by direct action on antigen-presenting cells (APCs), such as macrophages, DCs and B lymphocytes [44].
In a mouse model, the soluble polysaccharide fraction of C. acnes also induced an enhanced vaccination response in experimental T. cruzi infection [45].
In equine respiratory infections, inactivated C. acnes is commercialized under the name of Neogen®Vet EqStim® (Neogen Coroporation, Lansing, MI, USA) and administered as either prophylactic treatment or to enhance recovery from respiratory infections [46,47]. A study showed a significant improvement in the recovery as well as a significant decrease in disease severity from respiratory diseases in horses treated with heat-killed C. acnes compared to untreated controls [48]. These results were later confirmed by a second study treating 25 horses with C. acnes of which 96% of the horses showed complete recovery or improvement compared to 35% of the control group [47]. When entering the blood stream, C. acnes is phagocytized by macrophages in the liver or spleen. The degradation of C. acnes is delayed and could be the reason for the immune-modulatory properties of this bacterium [46,49,50].
In all vaccination studies it appears that the induction of a Th1 response by C. acnes in particular is the most important factor for the adjuvant properties of this bacterium.

3.3. Innate Immunity

As mentioned previously, C. acnes can interact with cells of the immune system on different levels (Figure 2). Not only macrophages, but also lymphocytes and neutrophils react to the pathogenic factors released by C. acnes, such as proteases, lipases and chemotactic factors [51]. Interaction with C. acnes and the immune system results in the stimulation and production of anti-microbial peptides and various chemokines and cytokines [52,53,54]. Toll-like receptors (TLR) on the immune cells can recognize C. acnes both extracellularly and intracellularly. It was seen that these immune cells were directly activated via TLR-2 and TLR-9 [55,56]. C. acnes is able to survive intracellularly inside macrophages, enabling it to evade the immune response of the host [57]. C. acnes produces catalase in order to facilitate this intracellular localization. Furthermore, C. acnes has a cell wall with a high resistance to degrading and oxidizing enzymes, enabling intracellular persistence [58].

3.4. Adaptive Immunity

Initially, C. acnes was seen as an Th1-inducing agent due to the cytokine pattern that is induced, such as IL-1α, IL-6, TNF-α, IL-12 and IL-18, which result in the release of IFN-γ [44,59]. Furthermore, early inflammatory acne lesions were shown to have an activated Th1 lymphocyte response [60].
In addition, C. acnes was shown not only to induce the production of Th1-type cytokines, but also Th17-type cytokines, such as IL-17. IL-17 is a pro-inflammatory cytokine which plays a role in various autoimmune and inflammatory disorders [61]. However, Th17 cells were shown to play a role in protecting the host against extracellular bacteria and fungi [62].
In recent years it has become clear that C. acnes can promote a mixed Th17/Th1 response by inducing the secretion of IL-17A and IFN-γ. When studying the blood of patients suffering from acne vulgaris it was seen that both Th17 and Th17.1 cells were found in higher frequencies than in healthy individuals and may play a possible role in its pathogenesis [63,64]. In this disease, the Th17 cells were shown to be related to the activation of CD4+ Th-cells resulting in an increase in secreted IL-17 in acne-related strains [64,65]. The differentiation of Th subsets was shown to be tightly regulated by transcription factors, signaling transducers and activator of transcription (STAT). STAT4 was shown to regulate Th1 differentiation, whereas Th2 differentiation is dependent on STAT5a. STAT3 in its turn is responsible for the differentiation of Th17 differentiation [62]. As mentioned previously, C. acnes was also shown to have an adjuvant effect by modulating the immune system. This adjuvant effect is related to a direct effect of the bacterial components on APCs, especially on dendritic cells. When the bacterial components bind the APCs expression of costimulatory molecules and TLRs is modulated, which was shown to be essential for driving the T-cell response [44,59]. Moreover, B1 cells were also shown to play a role in the adjuvant effect of C. acnes. B1 cells are a sub-class of B cell lymphocytes and their importance in adaptive immune responses has been well established.
Figure 2. Proposed interaction between Cutibacterium acnes and the immune system resulting in different immunoregulatory effects. C. acnes has extensive immunostimulatory activity. It can stimulate different innate immune receptor such as TLR-2, TLR-4 and intracellular TLR-9 and NOD-like receptors. C. acnes can induce a Thelper-1 (Th1) cytokine milieu and subsequent Th1 response which is thought to be beneficial in enhancing both anti-tumor response and vaccination. Furthermore, C. acnes seems capable of skewing a Th2 response to a Th1 response. Recent data also suggest that C. acnes is involved in initiating a more Th17-based response. The intercellular C. acnes infection is thought to induce granuloma formation in sarcoidosis. The mechanism of this antigenic role of C. acnes was hypothesized by Eishi et al. (adapted with permission from [66]). However, C. acnes is also thought to play a mitogenic role to granuloma formation in sarcoidosis, mainly based on induction of a Th1/Th17 response.
Figure 2. Proposed interaction between Cutibacterium acnes and the immune system resulting in different immunoregulatory effects. C. acnes has extensive immunostimulatory activity. It can stimulate different innate immune receptor such as TLR-2, TLR-4 and intracellular TLR-9 and NOD-like receptors. C. acnes can induce a Thelper-1 (Th1) cytokine milieu and subsequent Th1 response which is thought to be beneficial in enhancing both anti-tumor response and vaccination. Furthermore, C. acnes seems capable of skewing a Th2 response to a Th1 response. Recent data also suggest that C. acnes is involved in initiating a more Th17-based response. The intercellular C. acnes infection is thought to induce granuloma formation in sarcoidosis. The mechanism of this antigenic role of C. acnes was hypothesized by Eishi et al. (adapted with permission from [66]). However, C. acnes is also thought to play a mitogenic role to granuloma formation in sarcoidosis, mainly based on induction of a Th1/Th17 response.
Microorganisms 10 01649 g002

4. The Role of C. acnes in Sarcoidosis

4.1. Sarcoidosis and Possible Antigens

Sarcoidosis is a multi-systemic granulomatous disorder which most often affects the lungs and peripheral lymph nodes, but the disease can also affect other organs [67,68]. In sarcoidosis so called ‘epithelioid cell granulomas’ are formed by a Th1/Th17.1 immune response against an antigen with a strong antigenicity [69]. The lung is constantly confronted with airborne substances including micro-organisms. As this organ is most often affected by sarcoidosis, many researchers have considered infection as a trigger and have tried to identify possible infectious agents in sarcoidosis [70]. Over the years, many possible infectious but also noninfectious agents such as metals or silica have been suggested, summarized in Table 1. Regarding infectious agents, most studies were performed addressing the possible role of either mycobacteria, aspergillus species or C. acnes as antigens in sarcoidosis [71].

4.2. C. acnes as Antigen in Sarcoidosis

In the past decades, multiple studies have suggested that C. acnes might be a disease-causing antigen in a subgroup of patients with sarcoidosis. First of all, C. acnes has sporadically been cultured from the lymph nodes of patients with sarcoidosis [77,99]. However, as C. acnes is a commensal, Koch’s postulates cannot be applied, making it difficult to interpret these findings [100].
Secondly, C. acnes has been detected by immunohistochemical (IHC) staining in the tissues of various organs of patients with sarcoidosis, including the lungs [77,101,102,103], lymph nodes [77,101,104], skin [101] and heart [105].
In the case of the identification of the antigen in granulomas, possible antigens are more likely to be found in immature granulomas with many inflammatory cells than in mature granulomas in which the degradation process is almost completed and only a few lymphocytes are still present.
Thirdly, the DNA of C. acnes was shown to be amplified in a higher percentage of sarcoidosis patients than in the DNA of mycobacterium tuberculosis [106]. This was confirmed by Zhou et al., who combined data from various studies using meta-analysis and found that 78% of patients with sarcoidosis had C. acnes within the lesions [107]. However, the detection of DNA can also point to latent infection unrelated to the formation of sarcoidosis. This could be remedied with the use of a quantitative PCR, which can discriminate between a potential latent infection and a reactivated infection [71].
Finally, C. acnes elicits a specific immunoglobulin response in sarcoidosis patients. Increased local immunoglobulin production, especially IgG and IgA concentrations, were detected in BAL of sarcoidosis patients [108,109]. Schupp et al. were the first to show a B-cell-specific immune response to C. acnes in Caucasian sarcoidosis patients and showed that IgA production was increased only in early disease, especially Löfgren syndrome, while IgG was elevated in chronic disease as well [108]. Previously, Japanese studies already reported an increased Th1 immune response and increased IgG and IgA titers in PBMCs from sarcoidosis patients with presence of C. acnes [110,111,112].
Beijer et al. [88] tried to establish trigger-related phenotypes in Dutch sarcoidosis patients by measuring immunological sensitization with the use of enzyme-linked immune absorbent spot (ELISpot) assays. In case of immunological sensitization for C. acnes, 3.5% of the sarcoidosis patients showed a positive ELISpot for C. acnes catalase. Looking at the clinical phenotype, patients with a positive ELISpot for C. acnes catalase showed a trend towards more cutaneous involvement of the sarcoidosis and were significantly younger at diagnosis compared to patients without a positive C. acnes catalase ELISpot [84]. However, contrary to what was previously found in the immunoglobulin response studies [108,110,111], the immunological sensitization of C. acnes was much lower in Dutch sarcoidosis patients compared to the control group of obstructive sleep apnea (OSA) patients (3.5% versus 15%). Beijer et al., suggested that this lower percentage could reflect the homing of the cells to the lungs. However, it is not clear why the T cell response was lower for the sarcoidosis patients [88]. Based on these results one may argue that in order to study C. acnes sensitization in sarcoidosis, peripheral blood may not be the correct compartment.

4.3. A Possible Role of C. acnes as a Mitogen in Sarcoidosis

Traditionally, in the quest for the cause of sarcoidosis, the main focus has been the antigenic potential of microorganisms or inorganic materials studied. However, studies on the role of C. acnes have been hampered by the fact that this bacterium is a human commensal and indeed was found to be the most common commensal in lung tissue and mediastinal lymph nodes in patient without sarcoidosis [113]. In previous studies detecting C. acnes in sarcoidosis, available lung tissue after resection for lung cancer or granulomatous tissue of patients with active mycobacterial infection were used as a controls [67].
Data on whether C. acnes could also be present in non-infectious granulomatous pulmonary diseases such as hypersensitivity pneumonitis (HP) or vasculitis were not available. However, recently Beijer et al. demonstrated that C. acnes could also be detected in the granulomatous tissue of patients with HP and Eosinophilic granulomatosis with polyangiitis (EGPA). Interestingly, in light of the potential of C. acnes to skew an immune response towards a more Th-1 response they categorized HP patients into a group where C. acnes was present in the granulomas and a group without the presence of C. acnes in the granulomas. When comparing the amount of lymphocytosis in BAL fluid between both groups it was found that the presence of C. acnes in the granulomas of patients with HP was associated with a significantly higher amount of lymphocytes in BAL fluid (Figure 3), further supporting the hypothesis that the role of C. acnes could be mitogenic in different granulomatous pulmonary disorders [114].

5. Can the Presence of C. acnes in Patients with Sarcoidosis Be Seen as Treatable Trait?

Unfortunately, there is still no curative treatment for sarcoidosis [68]. Immunosuppressive drugs, such as steroids or disease-modifying antirheumatic drugs (DMARDs) are often prescribed aiming for a reduction in symptoms and the prevention of organ damage. However, when using immunosuppressive therapy, patients often experience many burdensome side effects [115,116,117]. There is a need for more personalized treatment in sarcoidosis, which could potentially be accomplished by identifying (non)infectious agents in individual sarcoidosis patients. In the case of C. acnes, one could speculate that a subgroup of sarcoidosis patients might benefit from therapy with antibiotics. The potential clinical relevance was already suggested in a study using a mouse model: in a group of 14 mice, 4 demonstrated parenchymal non-caseating granulomas after the mice were sensitized for C. acnes. However, if these mice were pre-treated with azithromycin, known to be bactericidal to C. acnes, only one of the mice developed a small number of granulomas after sensitization with this bacterium [118].
Moreover, for sarcoidosis patients there are already multiple case reports describing successful treatment with antibiotics of C. acnes-related sarcoidosis, especially in Japanese sarcoidosis patients. In 2008, a case of a 47-year-old female suffering from sarcoidosis affecting several parts of the body was described in whom treatment with minocycline resulted in an reduction in muscular sarcoidosis and a significant decrease in the serum Angiotensin-convertering enzyme (ACE) level [119]. Furthermore, in 2014 a case of a 78-year-old woman with C. acnes-associated sarcoidosis was described in which treatment with clarithromycin was shown to induce a resolution of the sarcoid granulomas and was found to be effective for treatment of the sarcoidosis in this particular patient [120]. However, no randomized controlled trials investigating the potential effect of antibiotic treatment in a patient with sarcoidosis where C. acnes is actually present in their granulomatous tissue have been carried out. In 2018 the J-Acnes trial was initiated, where patients with cardiac sarcoidosis are treated with antibacterial drugs in addition to the immunosuppressive therapy [121]. The results of this trial are eagerly awaited.
Recently, the PHENOSAR study was initiated in 2022 (ClinicalTrials.gov, Identifier: NCT05291468). This will be the first trial in which targeted therapy in sarcoidosis is based on the presence of C. acnes in granulomatous tissue. In this trial a combination of two antibiotics, azithromycin and doxycycline, will be used to minimize the chance of resistance development of the C. acnes bacterium. A total of four groups will be formed which will receive either antibiotics or a placebo, as shown in Figure 4.
Patients will receive treatment/placebo for 13 weeks after which the burden of inflammation is determined by comparing PET/CT-scan and serum biomarkers ACE and IL-2R of baseline and 13 weeks. Hopefully this study will provide data in order to determine whether the presence of C. acnes in patients with sarcoidosis could be seen as a treatable trait for this disease.

6. Conclusions

The human commensal C. acnes seems to be a jack of all trades with regard to possible influences on innate immunity, anti-tumor response, enhancement of vaccine effectivity and etiology of various diseases such as acne vulgaris, prostate cancer and sarcoidosis.
Over the past decades C. acnes has been identified as a possible cause of sarcoidosis; however, the discussion about causality vs. association with this disease is still not settled. Recent data suggest that C. acnes might also have a more mitogenic role in sarcoidosis but future studies are definitely needed to clarify the exact role of C. acnes in the pathogenesis of sarcoidosis.

Author Contributions

Conceptualization: R.K. and M.V., methodology: R.K. and M.V., investigation: R.K. and M.V., resources: R.K. and M.V., writing—original draft preparation: R.K., writing—review and editing: M.V., visualization: R.K. and M.V., supervision: M.V., Funding acquisition: M.V. All authors have read and agreed to the published version of the manuscript.

Funding

This study is part of the TopZorg grant funded by ZonMw (nr 10070012010004).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The study did not report any data.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Grice, E.A.; Segre, J.A. The skin microbiome. Nat. Rev. Microbiol. 2011, 9, 244–253. [Google Scholar] [CrossRef] [PubMed]
  2. Perry, A.; Lambert, P. Propionibacterium acnes: Infection beyond the skin. Expert Rev. Anti-Infect. Ther. 2011, 9, 1149–1156. [Google Scholar] [CrossRef] [PubMed]
  3. Portillo, M.E.; Corvec, S.; Borens, O.; Trampuz, A. Propionibacterium acnes: An underestimated pathogen in implant-associated infections. BioMed Res. Int. 2013, 2013, 804391. [Google Scholar] [CrossRef] [PubMed]
  4. McDowell, A.; Valanne, S.; Ramage, G.; Tunney, M.M.; Glenn, J.V.; McLorinan, G.C.; Bhatia, A.; Maisonneuve, J.-F.; Lodes, M.; Persing, D.H.; et al. Propionibacterium acnes types I and II represent phylogenetically distinct groups. J. Clin. Microbiol. 2005, 43, 326–334. [Google Scholar] [CrossRef]
  5. McDowell, A.; Nagy, I.; Magyari, M.; Barnard, E.; Patrick, S. The opportunistic pathogen Propionibacterium acnes: Insights into typing, human disease, clonal diversification and CAMP factor evolution. PLoS ONE 2013, 8, 70897. [Google Scholar] [CrossRef]
  6. McDowell, A.; Perry, A.L.; Lambert, P.A.; Patrick, S. A new phylogenetic group of Propionibacterium acnes. J. Med. Microbiol. 2008, 57, 218–224. [Google Scholar] [CrossRef]
  7. Dréno, B.; Pécastaings, S.; Corvec, S.; Veraldi, S.; Khammari, A.; Roques, C. Cutibacterium acnes (Propionibacterium acnes) and acne vulgaris: A brief look at the latest updates. J. Eur. Acad. Dermatol. Venereol. 2018, 32, 5–14. [Google Scholar] [CrossRef]
  8. Mayslich, C.; Grange, P.A.; Dupin, N. Cutibacterium acnes as an opportunistic pathogen: An update of its virulence-associated factors. Microorganisms 2021, 9, 303. [Google Scholar] [CrossRef]
  9. Layton, A.M.; Dreno, B.; Gollnick, H.P.M.; Zouboulis, C.C. A review of the European Directive for prescribing systemic isotretinoin for acne vulgaris. J. Eur. Acad. Dermatol. Venereol. 2006, 20, 773–776. [Google Scholar] [CrossRef]
  10. Mondal, D.; Shenoy, S.R.; Mishra, S. Retinoic acid embryopathy. Int. J. Appl. Basic Med. Res. 2017, 7, 264–265. [Google Scholar] [CrossRef]
  11. Dessinioti, C.; Katsambas, A.D. The role of Propionibacterium acnes in acne pathogenesis: Facts and controversies. Clin. Dermatol. 2010, 28, 2–7. [Google Scholar] [CrossRef]
  12. Wang, Y.; Hata, T.R.; Tong, Y.L.; Kao, M.-S.; Zouboulis, C.C.; Gallo, R.L.; Huang, C.-M. The anti-inflammatory activities of propionibacterium acnes CAMP factor-targeted acne vaccines. J. Investig. Dermatol. 2018, 138, 2355–2364. [Google Scholar] [CrossRef]
  13. Achermann, Y.; Goldstein, E.J.C.; Coenye, T.; Shirtliff, M.E. Propionibacterium acnes: From commensal to opportunistic biofilm-associated implant pathogen. Clin. Microbiol. Rev. 2014, 27, 419–440. [Google Scholar] [CrossRef]
  14. Boisrenoult, P. Cutibacterium acnes prosthetic joint infection: Diagnosis and treatment. Orthop. Traumatol. Surg. Res. 2018, 104, S19–S24. [Google Scholar] [CrossRef]
  15. Brüggemann, H.; Salar-Vidal, L.; Gollnick, H.P.M.; Lood, R. A Janus-faced bacterium: Host-beneficial and -detrimental roles of cutibacterium acnes. Front. Microbiol. 2021, 12, 673845. [Google Scholar] [CrossRef]
  16. Del Pozo, J.L.; Tran, N.V.; Petty, P.M.; Johnson, C.H.; Walsh, M.F.; Bite, U.; Clay, R.P.; Mandrekar, J.N.; Piper, K.E.; Steckelberg, J.M.; et al. Pilot study of association of bacteria on breast implants with capsular contracture. J. Clin. Microbiol. 2009, 47, 1333–1337. [Google Scholar] [CrossRef]
  17. Ajdic, D.; Zoghbi, Y.; Gerth, D.; Panthaki, Z.J.; Thaller, S. The relationship of bacterial biofilms and capsular contracture in breast implants. Aesthetic Surg. J. 2016, 36, 297–309. [Google Scholar] [CrossRef]
  18. Conen, A.; Walti, L.N.; Merlo, A.; Fluckiger, U.; Battegay, M.; Trampuz, A. Characteristics and treatment outcome of cerebrospinal fluid shunt–associated infections in adults: A retrospective analysis over an 11-year period. Clin. Infect. Dis. 2008, 47, 73–82. [Google Scholar] [CrossRef]
  19. Piper, K.E.; Jacobson, M.J.; Cofield, R.H.; Sperling, J.W.; Sanchez-Sotelo, J.; Osmon, D.R.; McDowell, A.; Patrick, S.; Steckelberg, J.M.; Mandrekar, J.N.; et al. Microbiologic diagnosis of prosthetic shoulder infection by use of implant sonication. J. Clin. Microbiol. 2009, 47, 1878–1884. [Google Scholar] [CrossRef]
  20. Levy, P.Y.; Fenollar, F.; Stein, A.; Borrione, F.; Cohen, E.; Lebail, B.; Raoult, D. Propionibacterium acnes postoperative shoulder arthritis: An emerging clinical entity. Clin. Infect. Dis. 2008, 46, 1884–1886. [Google Scholar] [CrossRef]
  21. Davidsson, S.; Mölling, P.; Rider, J.; Unemo, M.; Karlsson, M.G.; Carlsson, J.; Andersson, S.-O.; Elgh, F.; Söderquist, B.; Andrén, O. Frequency and typing of Propionibacterium acnes in prostate tissue obtained from men with and without prostate cancer. Infect. Agents Cancer 2016, 11, 26. [Google Scholar] [CrossRef]
  22. Bidaud, A.-L.; Karam, G.; Kandel-Aznar, C.; D’Epenoux, L.R.; Guillouzouic, A.; Bémer, P.; Leroy, A.-G.; Corvec, S. Low prevalence of Cutibacterium acnes in prostatic tissue biopsies in a French hospital. Anaerobe 2020, 66, 102286. [Google Scholar] [CrossRef]
  23. Erlandsson, A.; Carlsson, J.; Lundholm, M.; Fält, A.; Andersson, S.-O.; Andrén, O.; Davidsson, S. M2 macrophages and regulatory T cells in lethal prostate cancer. Prostate 2019, 79, 363–369. [Google Scholar] [CrossRef]
  24. Davidsson, S.; Andren, O.; Ohlson, A.-L.; Carlsson, J.; Andersson, S.-O.; Giunchi, F.; Rider, J.R.; Fiorentino, M. FOXP3+ regulatory T cells in normal prostate tissue, postatrophic hyperplasia, prostatic intraepithelial neoplasia, and tumor histological lesions in men with and without prostate cancer. Prostate 2018, 78, 40–47. [Google Scholar] [CrossRef]
  25. Idorn, M.; Køllgaard, T.; Kongsted, P.; Sengeløv, L.; Straten, P.T. Correlation between frequencies of blood monocytic myeloid-derived suppressor cells, regulatory T cells and negative prognostic markers in patients with castration-resistant metastatic prostate cancer. Cancer Immunol. Immunother. 2014, 63, 1177–1187. [Google Scholar] [CrossRef]
  26. Roszkowski, W.; Ko, H.; Beuth, J.; Jeljaszewicz, J. Immunomodulation by propionibacteria. Zent. Bakteriol. 1990, 274, 289–298. [Google Scholar] [CrossRef]
  27. Michalak-Stoma, A.; Tabarkiewicz, J.; Olender, A.; Juszkiewicz-Borowiec, M.; Stoma, F.; Pietrzak, A.; Pozarowski, P.; Bartkowiak-Emeryk, M. The effect of Propionibacterium acnes on maturation of dendritic cells derived from acne patients’ peripherial blood mononuclear cells. Folia Histochem. Cytobiol. 2008, 46, 535–539. [Google Scholar] [CrossRef]
  28. Talib, W.H.; Saleh, S. Propionibacterium acnes augments antitumor, anti-angiogenesis and immunomodulatory effects of melatonin on breast cancer implanted in mice. PLoS ONE 2015, 10, e0124384. [Google Scholar] [CrossRef]
  29. Tsuda, K.; Yamanaka, K.; Linan, W.; Miyahara, Y.; Akeda, T.; Nakanishi, T.; Kitagawa, H.; Kakeda, M.; Kurokawa, I.; Shiku, H.; et al. Intratumoral injection of Propionibacterium acnes suppresses malignant melanoma by enhancing Th1 immune responses. PLoS ONE 2011, 6, e29020. [Google Scholar] [CrossRef]
  30. Dang, L.H.; Bettegowda, C.; Huso, D.L.; Kinzler, K.W.; Vogelstein, B. Combination bacteriolytic therapy for the treatment of experimental tumors. Proc. Natl. Acad. Sci. USA 2001, 98, 15155–15160. [Google Scholar] [CrossRef]
  31. Talib, W.H.; Mahasneh, A.M. Combination of Ononis hirta and Bifidobacterium longum decreases syngeneic mouse mammary tumor burden and enhances immune response. J. Cancer Res. Ther. 2012, 8, 417–423. [Google Scholar] [CrossRef] [PubMed]
  32. Lee, C.-H.; Wu, C.-L.; Shiau, A.-L. Toll-like receptor 4 mediates an antitumor host response induced by Salmonella choleraesuis. Clin. Cancer Res. 2008, 14, 1905–1912. [Google Scholar] [CrossRef] [PubMed]
  33. Buck, G.E.; Kelly, M.T. Investigation of the component of Propionibacterium acnes (Corynebacterium parvum) responsible for macrophage activation. Infect. Immun. 1980, 27, 620–627. [Google Scholar] [CrossRef] [PubMed]
  34. Howard, J.G.; Christie, G.H.; Scott, M.T. Biological effects of Corynebacterium parvum: IV. Adjuvant and inhibitory activities on B lymphocytes. Cell. Immunol. 1972, 7, 290–301. [Google Scholar] [CrossRef]
  35. Lichtenstein, A.; Bick, A.; Cantrell, J.; Zighelboim, J. Augmentation of NK activity by Corynebacterium parvum fractions in vivo and in vitro. Int. J. Immunopharmacol. 1983, 5, 137–144. [Google Scholar] [CrossRef]
  36. Disis, M.L. Immune regulation of cancer. J. Clin. Oncol. 2010, 28, 4531–4538. [Google Scholar] [CrossRef]
  37. Nakano, A.; Yoneyama, H.; Ueha, S.; Kitabatake, M.; Ishikawa, S.; Kawase, I.; Sugiyama, H.; Matsushima, K. Intravenous administration of MIP-1α with intra-tumor injection of P. acnes shows potent anti-tumor effect. Int. Immunopharmacol. 2007, 7, 845–857. [Google Scholar] [CrossRef]
  38. Bast, R.C., Jr.; Knapp, R.C.; Mitchell, A.K.; Thurston, J.G.; Tucker, R.W.; Schlossman, S.F. Immunotherapy of a murine ovarian carcinoma with Corynebacterium parvum and specific heteroantiserum. I. Activation of peritoneal cells to mediate antibody-dependent cytotoxicity. J. Immunol. 1979, 123, 1945–1951. [Google Scholar]
  39. Knapp, R.C.; Berkowitz, R.S. Corynebacterium parvum as an immunotherapeutic agent in an ovarian cancer model. Am. J. Obstet. Gynecol. 1977, 128, 782–786. [Google Scholar] [CrossRef]
  40. Berek, J.S.; Cantrell, J.L.; Lichtenstein, A.K.; Hacker, N.F.; Knox, R.M.; Nieberg, R.K.; Poth, T.; Elashoff, R.M.; Lagasse, L.D.; Zighelboim, J. Immunotherapy with biochemically dissociated fractions of Propionibacterium acnes in a murine ovarian cancer model. Cancer Res. 1984, 44, 1871–1875. [Google Scholar]
  41. Roszkowski, W.; Ko, H.L.; Szmigielski, S.; Jeljaszewicz, J.; Pulverer, G. The correlation of susceptibility of different Propionibacterium strains to macrophage killing and antitumor activity. Med. Microbiol. Immunol. 1980, 169, 1–8. [Google Scholar] [CrossRef]
  42. Fujiwara, H.; Tsuchida, T.; Tsuji, Y.; Hamaoka, T. Preventive effect of Propionibacterium acnes on metastasis in mice rendered tolerant to tumor-associated transplantation antigens. GANN Jpn. J. Cancer Res. 1980, 71, 692–698. [Google Scholar]
  43. Teixeira, D.; Ishimura, M.E.; Apostolico, J.; Viel, J.M.; Passarelli, V.C.; Cunha-Neto, E.; Rosa, D.; Longo-Maugéri, I.M. Propionibacterium acnes enhances the immunogenicity of HIVBr18 human immunodeficiency virus-1 vaccine. Front. Immunol. 2018, 9, 177. [Google Scholar] [CrossRef]
  44. Squaiella-Baptistão, C.C.; Teixeira, D.; Mussalem, J.S.; Ishimura, M.E.; Longo-Maugéri, I.M. Modulation of Th1/Th2 immune responses by killed Propionibacterium acnes and its soluble polysaccharide fraction in a type I hypersensitivity murine model: Induction of different activation status of antigen-presenting cells. J. Immunol. Res. 2015, 2015, 132083. [Google Scholar] [CrossRef]
  45. Mussalem, J.S.; Squaiella-Baptistão, C.C.; Teixeira, D.; Yendo, T.M.; Thies, F.G.; Popi, A.F.; Mariano, M.; Longo-Maugéri, I.M. Adjuvant effect of killed Propionibacterium acnes on mouse peritoneal B-1 lymphocytes and their early phagocyte differentiation. PLoS ONE 2012, 7, e33955. [Google Scholar] [CrossRef]
  46. Paillot, R. A systematic review of the immune-modulators Parapoxvirus ovis and Propionibacterium acnes for the prevention of respiratory disease and other infections in the horse. Vet. Immunol. Immunopathol. 2013, 153, 1–9. [Google Scholar] [CrossRef]
  47. Vail, C.D.; Nestved, A.J.; Rollins, J.B.; Huff, G.K.; Hartgrove, T.B.; Evans, D.R.; Clapper, J.J.; Van Kampen, K.R.; Peters, B.A.; Hay, C.A. Adjunct treatment of equine respiratory disease complex (ERDC) with the Propionibacterium acnes, immunostimulant, EqStim®. J. Equine Vet. Sci. 1990, 10, 399–400. [Google Scholar] [CrossRef]
  48. Evans, D.R.; Brent Rollins, J.; Huff, G.K.; Hartgrove, T.B.; Van Kampen, K.R. Inactivated Propionibacterium acnes (immunoregulin) as adjunct to conventional therapy in the treatment of equine respiratory diseases. Equine Pract. 1998, 10, 17–21. [Google Scholar]
  49. Tchaptchet, S.; Gumenscheimer, M.; Kalis, C.; Freudenberg, N.; Hölscher, C.; Kirschning, C.J.; Lamers, M.; Galanos, C.; Freudenberg, M.A. TLR9-dependent and independent pathways drive activation of the immune system by Propionibacterium acnes. PLoS ONE 2012, 7, e39155. [Google Scholar] [CrossRef]
  50. Kalis, C.; Gumenscheimer, M.; Freudenberg, N.; Tchaptchet, S.; Fejer, G.; Heit, A.; Akira, S.; Galanos, C.; Freudenberg, M.A. Requirement for TLR9 in the immunomodulatory activity of Propionibacterium acnes. J. Immunol. 2005, 174, 4295–4300. [Google Scholar] [CrossRef]
  51. Holland, C.; Mak, T.N.; Zimny-Arndt, U.; Schmid, M.; Meyer, T.F.; Jungblut, P.R.; Brüggemann, H. Proteomic identification of secreted proteins of Propionibacterium acnes. BMC Microbiol. 2010, 10, 230. [Google Scholar] [CrossRef]
  52. Chen, Q.; Koga, T.; Uchi, H.; Hara, H.; Terao, H.; Moroi, Y.; Urabe, K.; Furue, M. Propionibacterium acnes-induced IL-8 production may be mediated by NF-κB activation in human monocytes. J. Dermatol. Sci. 2002, 29, 97–103. [Google Scholar] [CrossRef]
  53. Nagy, I.; Pivarcsi, A.; Koreck, A.; Széll, M.; Urbán, E.; Kemény, L. Distinct strains of Propionibacterium acnes induce selective human β-defensin-2 and interleukin-8 expression in human keratinocytes through toll-like receptors. J. Investig. Dermatol. 2005, 124, 931–938. [Google Scholar] [CrossRef]
  54. Yu, Y.; Champer, J.; Agak, G.W.; Kao, S.; Modlin, R.L.; Kim, J. Different Propionibacterium acnes phylotypes induce distinct immune responses and express unique surface and secreted proteomes. J. Investig. Dermatol. 2016, 136, 2221–2228. [Google Scholar] [CrossRef]
  55. Kim, J.; Ochoa, M.-T.; Krutzik, S.R.; Takeuchi, O.; Uematsu, S.; Legaspi, A.J.; Brightbill, H.D.; Holland, D.; Cunliffe, W.J.; Akira, S.; et al. Activation of toll-like receptor 2 in acne triggers inflammatory cytokine responses. J. Immunol. 2002, 169, 1535–1541. [Google Scholar] [CrossRef]
  56. Tanabe, T.; Ishige, I.; Suzuki, Y.; Aita, Y.; Furukawa, A.; Ishige, Y.; Uchida, K.; Suzuki, T.; Takemura, T.; Ikushima, S.; et al. Sarcoidosis and NOD1 variation with impaired recognition of intracellular Propionibacterium acnes. Biochim. Biophys. Acta 2006, 1762, 794–801. [Google Scholar] [CrossRef]
  57. Fischer, N.; Mak, T.N.; Shinohara, D.B.; Sfanos, K.S.; Meyer, T.F.; Brüggemann, H. Deciphering the intracellular fate of Propionibacterium acnes in macrophages. BioMed Res. Int. 2013, 2013, 603046. [Google Scholar] [CrossRef]
  58. Yamamoto, K.; Uchida, K.; Furukawa, A.; Tamura, T.; Ishige, Y.; Negi, M.; Kobayashi, D.; Ito, T.; Kakegawa, T.; Hebisawa, A.; et al. Catalase expression of Propionibacterium acnes may contribute to intracellular persistence of the bacterium in sinus macrophages of lymph nodes affected by sarcoidosis. Immunol. Res. 2019, 67, 182–193. [Google Scholar] [CrossRef]
  59. Mussalem, J.S.; Vasconcelos, J.R.C.; Squaiella, C.C.; Ananias, R.Z.; Braga, E.G.; Rodrigues, M.M.; Longo-Maugéri, I.M. Adjuvant effect of the Propionibacterium acnes and its purified soluble polysaccharide on the immunization with plasmidial DNA containing a Trypanosoma cruzi gene. Microbiol. Immunol. 2006, 50, 253–263. [Google Scholar] [CrossRef]
  60. Mouser, P.E.; Seaton, E.D.; Chu, A.C.; Baker, B.S. Propionibacterium acnes-reactive T helper-1 cells in the skin of patients with acne vulgaris. J. Investig. Dermatol. 2003, 121, 1226–1228. [Google Scholar] [CrossRef]
  61. Kistowska, M.; Meier, B.; Proust, T.; Feldmeyer, L.; Cozzio, A.; Kuendig, T.; Contassot, E.; French, L.E. Propionibacterium acnes promotes Th17 and Th17/Th1 responses in acne patients. J. Investig. Dermatol. 2015, 135, 110–118. [Google Scholar] [CrossRef] [PubMed]
  62. Zhu, J.; Yamane, H.; Paul, W.E. Differentiation of effector CD4 T cell populations. Annu. Rev. Immunol. 2010, 28, 445–489. [Google Scholar] [CrossRef] [PubMed]
  63. Agak, G.W.; Qin, M.; Nobe, J.; Kim, M.-H.; Krutzik, S.R.; Tristan, G.R.; Elashoff, D.; Garbán, H.J.; Kim, J. Propionibacterium acnes induces an IL-17 response in Acne vulgaris that is regulated by vitamin A and vitamin D. J. Investig. Dermatol. 2014, 134, 366–373. [Google Scholar] [CrossRef] [PubMed]
  64. Agak, G.W.; Kao, S.; Ouyang, K.; Qin, M.; Moon, D.; Butt, A.; Kim, J. Phenotype and antimicrobial activity of Th17 cells induced by Propionibacterium acnes strains associated with healthy and acne skin. J. Investig. Dermatol. 2018, 138, 316–324. [Google Scholar] [CrossRef]
  65. Kelhälä, H.-L.; Palatsi, R.; Fyhrquist, N.; Lehtimäki, S.; Väyrynen, J.; Kallioinen, M.; Kubin, M.E.; Greco, D.; Tasanen, K.; Alenius, H.; et al. IL-17/Th17 pathway is activated in acne lesions. PLoS ONE 2014, 9, e105238. [Google Scholar] [CrossRef]
  66. Eishi, Y. Etiologic link between sarcoidosis and Propionibacterium acnes. Respir. Investig. 2013, 51, 56–68. [Google Scholar] [CrossRef]
  67. Eishi, Y.; Suga, M.; Ishige, I.; Kobayashi, D.; Yamada, T.; Takemura, T.; Takizawa, T.; Koike, M.; Kudoh, S.; Costabel, U.; et al. Quantitative analysis of mycobacterial and propionibacterial DNA in lymph nodes of Japanese and European patients with sarcoidosis. J. Clin. Microbiol. 2002, 40, 198–204. [Google Scholar] [CrossRef]
  68. Costabel, U.; Hunninghake, G.W. ATS/ERS/WASOG statement on sarcoidosis. Eur. Respir. J. 1999, 14, 735–737. [Google Scholar] [CrossRef]
  69. Pagán, A.J.; Ramakrishnan, L. The formation and function of granulomas. Annu. Rev. Immunol. 2018, 36, 639–665. [Google Scholar] [CrossRef]
  70. Nishiwaki, T.; Yoneyama, H.; Eishi, Y.; Matsuo, N.; Tatsumi, K.; Kimura, H.; Kuriyama, T.; Matsushima, K. Indigenous pulmonary Propionibacterium acnes primes the host in the development of sarcoid-like pulmonary granulomatosis in mice. Am. J. Pathol. 2004, 165, 631–639. [Google Scholar] [CrossRef]
  71. Yamaguchi, T.; Costabel, U.; McDowell, A.; Guzman, J.; Uchida, K.; Ohashi, K.; Eishi, Y. Immunohistochemical detection of potential microbial antigens in granulomas in the diagnosis of sarcoidosis. J. Clin. Med. 2021, 10, 983. [Google Scholar] [CrossRef]
  72. Hawkins, C.; Shaginurova, G.; Shelton, D.A.; Herazo-Maya, J.D.; Oswald-Richter, K.A.; Rotsinger, J.E.; Young, A.; Celada, L.J.; Kaminski, N.; Sevin, C.; et al. Local and systemic CD4+ T cell exhaustion reverses with clinical resolution of pulmonary sarcoidosis. J. Immunol. Res. 2017, 2017, 3642832. [Google Scholar] [CrossRef]
  73. Rotsinger, J.E.; Celada, L.J.; Polosukhin, V.V.; Atkinson, J.B.; Drake, W.P. Molecular analysis of sarcoidosis granulomas reveals antimicrobial targets. Am. J. Respir. Cell Mol. Biol. 2016, 55, 128–134. [Google Scholar] [CrossRef]
  74. Chen, E.S.; Wahlström, J.; Song, Z.; Willett, M.H.; Wikén, M.; Yung, R.C.; West, E.E.; McDyer, J.F.; Zhang, Y.; Eklund, A.; et al. T cell responses to mycobacterial catalase-peroxidase profile a pathogenic antigen in systemic sarcoidosis. J. Immunol. 2014, 181, 8784–8796. [Google Scholar] [CrossRef]
  75. Oswald-Richter, K.; Sato, H.; Hajizadeh, R.; Shepherd, B.E.; Sidney, J.; Sette, A.; Newman, L.S.; Drake, W.P. Mycobacterial ESAT-6 and katG are recognized by sarcoidosis CD4+ T cells when presented by the American sarcoidosis susceptibility allele, DRB1*1101. J. Clin. Immunol. 2010, 30, 157–166. [Google Scholar] [CrossRef]
  76. Suzuki, Y.; Uchida, K.; Takemura, T.; Sekine, M.; Tamura, T.; Furukawa, A.; Hebisawa, A.; Sakakibara, Y.; Awano, N.; Amano, T.; et al. Propionibacterium acnes-derived insoluble immune complexes in sinus macrophages of lymph nodes affected by sarcoidosis. PLoS ONE 2018, 13, e0192408. [Google Scholar] [CrossRef]
  77. Negi, M.; Takemura, T.; Guzman, J.; Uchida, K.; Furukawa, A.; Suzuki, Y.; Iida, T.; Ishige, I.; Minami, J.; Yamada, T.; et al. Localization of Propionibacterium acnes in granulomas supports a possible etiologic link between sarcoidosis and the bacterium. Mod. Pathol. 2012, 25, 1284–1297. [Google Scholar] [CrossRef]
  78. Ishihara, M.; Ohno, S.; Ono, H.; Isogai, E.; Kimura, K.; Isogai, H.; Aoki, K.; Ishida, T.; Suzuki, K.; Kotake, S.; et al. Seroprevalence of anti-Borrelia antibodies among patients with confirmed sarcoidosis in a region of Japan where Lyme borreliosis is endemic. Graefe’s Arch. Clin. Exp. Ophthalmol. 1998, 236, 280–284. [Google Scholar] [CrossRef]
  79. Clarke, E.L.; Lauder, A.P.; Hofstaedter, C.E.; Hwang, Y.; Fitzgerald, A.S.; Imai, I.; Biernat, W.; Rękawiecki, B.; Majewska, H.; Dubaniewicz, A.; et al. Microbial lineages in sarcoidosis. A metagenomic analysis tailored for low–microbial content samples. Am. J. Respir. Crit. Care Med. 2018, 197, 225–234. [Google Scholar] [CrossRef]
  80. Terčelj, M.; Stopinšek, S.; Ihan, A.; Salobir, B.; Simčič, S.; Wraber, B.; Rylander, R. In vitro and in vivo reactivity to fungal cell wall agents in sarcoidosis. Clin. Exp. Immunol. 2011, 166, 87–93. [Google Scholar] [CrossRef]
  81. Suchankova, M.; Paulovicova, E.; Majer, I.; Tedlova, E.; Novosadova, H.; Tibenska, E.; Tedla, M.; Bucova, M. Increased antifungal antibodies in bronchoalveolar lavage fluid and serum in pulmonary sarcoidosis. Scand. J. Immunol. 2015, 81, 259–264. [Google Scholar] [CrossRef] [PubMed]
  82. Greaves, S.A.; Ravindran, A.; Santos, R.G.; Chen, L.; Falta, M.T.; Wang, Y.; Mitchell, A.M.; Atif, S.M.; Mack, D.G.; Tinega, A.N.; et al. CD4+ T cells in the lungs of acute sarcoidosis patients recognize an Aspergillus nidulans epitope. J. Exp. Med. 2021, 218, e20210785. [Google Scholar] [CrossRef]
  83. Lim, C.X.; Weichhart, T. A fungal antigenic driver for Löfgren’s syndrome sarcoidosis. J. Exp. Med. 2021, 218, e20211572. [Google Scholar] [CrossRef]
  84. Beijer, E.; Meek, B.; Bossuyt, X.; Peters, S.; Vermeulen, R.C.H.; Kromhout, H.; Veltkamp, M. Immunoreactivity to metal and silica associates with sarcoidosis in Dutch patients. Respir. Res. 2020, 21, 141. [Google Scholar] [CrossRef]
  85. Fireman, E.; Bar Shai, A.; Alcalay, Y.; Ophir, N.; Kivity, S.; Stejskal, V. Identification of metal sensitization in sarcoid-like metal-exposed patients by the MELISA® lymphocyte proliferation testA pilot study. J. Occup. Med. Toxicol. 2016, 11, 18. [Google Scholar] [CrossRef]
  86. Li, L.; Silveira, L.J.; Hamzeh, N.; Gillespie, M.; Mroz, P.M.; Mayer, A.S.; Fingerlin, T.E.; Maier, L.A. Beryllium-induced lung disease exhibits expression profiles similar to sarcoidosis. Eur. Respir. J. 2016, 47, 1797–1808. [Google Scholar] [CrossRef]
  87. Rossman, M.D.; Kreider, M.E. Is chronic beryllium disease sarcoidosis of known etiology? Sarcoidosis Vasc. Diffus. Lung Dis. 2003, 20, 104–109. [Google Scholar]
  88. Beijer, E.; Kraaijvanger, R.; Roodenburg, C.; Grutters, J.C.; Meek, B.; Veltkamp, M. Simultaneous testing of immunological sensitization to multiple antigens in sarcoidosis reveals an association with inorganic antigens specifically related to a fibrotic phenotype. Clin. Exp. Immunol. 2020, 203, 115–124. [Google Scholar] [CrossRef]
  89. Drent, M.; Bomans, P.; Van Suylen, R.; Lamers, R.; Bast, A.; Wouters, E. Association of man-made mineral fibre exposure and sarcoidlike granulomas. Respir. Med. 2000, 94, 815–820. [Google Scholar] [CrossRef]
  90. Üzmezoǧlu, B.; Şimşek, C.; Gülgösteren, S.; Gebeşoǧlu, B.; Sari, G.; Çelik, D. Sarcoidosis in iron-steel industry: Mini case series. Sarcoidosis Vasc. Diffus. Lung Dis 2017, 34, 365–372. [Google Scholar]
  91. Newman, L.S. Metals that cause sarcoidosis. Semin. Respir. Infect. 1998, 13, 212–220. [Google Scholar] [PubMed]
  92. Tomioka, H.; Kaneda, T.; Katsuyama, E.; Kitaichi, M.; Moriyama, H.; Suzuki, E. Elemental analysis of occupational granulomatous lung disease by electron probe microanalyzer with wavelength dispersive spectrometer: Two case reports. Respir. Med. Case Rep. 2016, 18, 66–72. [Google Scholar] [CrossRef] [PubMed]
  93. Prezant, D.J.; Dhala, A.; Goldstein, A.; Janus, D.; Ortiz, F.; Aldrich, T.K.; Kelly, K.J. The incidence, prevalence, and severity of sarcoidosis in New York City firefighters. Chest 1999, 116, 1183–1193. [Google Scholar] [CrossRef] [PubMed]
  94. Kern, D.G.; Neill, M.A.; Wrenn, D.S.; Varone, J.C. Investigation of a unique time-space cluster of sarcoidosis in firefighters. Am. Rev. Respir. Dis. 1993, 148, 974–980. [Google Scholar] [CrossRef]
  95. Kajdasz, D.K.; Lackland, D.T.; Mohr, L.C.; Judson, M.A. A current assessment of rurally linked exposures as potential risk factors for sarcoidosis. Ann. Epidemiol. 2001, 11, 111–117. [Google Scholar] [CrossRef]
  96. Grunewald, J.; Kaiser, Y.; Ostadkarampour, M.; Rivera, N.V.; Vezzi, F.; Lötstedt, B.; Olsen, R.-A.; Sylwan, L.; Lundin, S.; Käller, M.; et al. T-cell receptor–HLA-DRB1 associations suggest specific antigens in pulmonary sarcoidosis. Eur. Respir. J. 2016, 47, 898–909. [Google Scholar] [CrossRef]
  97. Wahlström, J.; Dengjel, J.; Winqvist, O.; Targoff, I.; Persson, B.; Duyar, H.; Rammensee, H.-G.; Eklund, A.; Weissert, R.; Grunewald, J. Autoimmune T cell responses to antigenic peptides presented by bronchoalveolar lavage cell HLA-DR molecules in sarcoidosis. Clin. Immunol. 2009, 133, 353–363. [Google Scholar] [CrossRef]
  98. Wahlström, J.; Dengjel, J.; Persson, B.; Duyar, H.; Rammensee, H.-G.; Stevanoviδc, S.; Eklund, A.; Weissert, R.; Grunewald, J. Identification of HLA-DR–bound peptides presented by human bronchoalveolar lavage cells in sarcoidosis. J. Clin. Investig. 2007, 117, 3576–3582. [Google Scholar] [CrossRef]
  99. De Brouwer, B.; Veltkamp, M.; Wauters, C.A.; Grutters, J.C.; Janssen, R. Propionibacterium acnes isolated from lymph nodes of patients with sarcoidosis. Sarcoidosis Vasc. Diffus. Lung Dis 2015, 32, 271–274. [Google Scholar]
  100. Ishige, I.; Eishi, Y.; Takemura, T.; Kobayashi, I.; Nakata, K.; Tanaka, I.; Nagaoka, S.; Iwai, K.; Watanabe, K.; Takizawa, T.; et al. Propionibacterium acnes is the most common bacterium commensal in peripheral lung tissue and mediastinal lymph nodes from subjects without sarcoidosis. Sarcoidosis Vasc. Diffus. Lung Dis. Off. J. WASOG 2005, 22, 33–42. [Google Scholar]
  101. Beijer, E.; Seldenrijk, K.; Eishi, Y.; Uchida, K.; Damen, J.; Grutters, J.C.; Veltkamp, M. Presence of Propionibacterium acnes in granulomas associates with a chronic disease course in Dutch sarcoidosis patients. ERJ Open Res. 2021, 7, 00486–02020. [Google Scholar] [CrossRef]
  102. Isshiki, T.; Matsuyama, H.; Sakamoto, S.; Honma, N.; Mikami, T.; Shibuya, K.; Eishi, Y.; Homma, S. Development of Propionibacterium acnes-associated sarcoidosis during etanercept therapy. Intern. Med. 2019, 58, 1473–1477. [Google Scholar] [CrossRef]
  103. Suzuki, T.; Fujita, A.; Takamori, M.; Murata, K.; Wada, A.; Miyamoto, M.; Yamamoto, Y.; Sakashita, K.; Tada, Y.; Suzuki, Y.; et al. Implication of immunohistochemistry for propionibacterium acnes in differential diagnosis of necrotizing granuloma. J. Pulm. Respir. Med. 2016, 6, 2–4. [Google Scholar] [CrossRef]
  104. Kinoshita, Y.; Ishii, H.; Eishi, Y.; Uchida, K.; Yoshimura, M.; Iwasaki, A.; Fujita, M.; Nabeshima, K.; Watanabe, K. Histological differences between sarcoidosis and lung cancer-related sarcoid reaction. Respir. Investig. 2020, 58, 421–424. [Google Scholar] [CrossRef]
  105. Asakawa, N.; Uchida, K.; Sakakibara, M.; Omote, K.; Noguchi, K.; Tokuda, Y.; Kamiya, K.; Hatanaka, K.C.; Matsuno, Y.; Yamada, S.; et al. Immunohistochemical identification of Propionibacterium acnes in granuloma and inflammatory cells of myocardial tissues obtained from cardiac sarcoidosis patients. PLoS ONE 2017, 12, e0179980. [Google Scholar] [CrossRef]
  106. Ishige, I.; Usui, Y.; Takemura, T.; Eishi, Y.; Suga, M.; Ishige, I.; Kobayashi, D.; Yamada, T.; Takemura, T.; Takizawa, T.; et al. Quantitative PCR of mycobacterial and propionibacterial DNA in lymph nodes of Japanese patients with sarcoidosis. Lancet 1999, 354, 198–204. [Google Scholar] [CrossRef]
  107. Zhou, Y.; Hu, Y.; Li, H. Role of Propionibacterium acnes in sarcoidosis: A meta-analysis. Sarcoidosis Vasc. Diffus. Lung Dis. Off. J. WASOG 2013, 30, 262–267. [Google Scholar]
  108. Schupp, J.C.; Tchaptchet, S.; Lützen, N.; Engelhard, P.; Müller-Quernheim, J.; Freudenberg, M.A.; Prasse, A. Immune response to Propionibacterium acnes in patients with sarcoidosisIn vivo and in vitro. BMC Pulm. Med. 2015, 15, 75. [Google Scholar] [CrossRef]
  109. Vandenplas, O.; Depelchin, S.; Delaunois, L.; Delwiche, J.P.; Sibille, Y. Bronchoalveolar lavage immunoglobulin A and G and antiproteases correlate with changes in diffusion indices during the natural course of pulmonary sarcoidosis. Eur. Respir. J. 1994, 7, 1856–1864. [Google Scholar] [CrossRef]
  110. Furusawa, H.; Suzuki, Y.; Miyazaki, Y.; Inase, N.; Eishi, Y. Th1 and Th17 immune responses to viable Propionibacterium acnes in patients with sarcoidosis. Respir. Investig. 2012, 50, 104–109. [Google Scholar] [CrossRef]
  111. Yorozu, P.; Furukawa, A.; Uchida, K.; Akashi, T.; Kakegawa, T.; Ogawa, T.; Minami, J.; Suzuki, Y.; Awano, N.; Furusawa, H.; et al. Propionibacterium acnes catalase induces increased Th1 immune response in sarcoidosis patients. Respir. Investig. 2015, 53, 161–169. [Google Scholar] [CrossRef]
  112. Ebe, Y.; Ikushima, S.; Yamaguchi, T.; Kohno, K.; Azuma, A.; Sato, K.; Ishige, I.; Usui, Y.; Takemura, T.; Eishi, Y. Proliferative response of peripheral blood mononuclear cells and levels of antibody to recombinant protein from Propionibacterium acnes DNA expression library in Japanese patients with sarcoidosis. Sarcoidosis Vasc. Diffus. Lung Dis. Off. J. WASOG 2000, 17, 256–265. [Google Scholar]
  113. Yamane, H.; Tachibana, I.; Takeda, Y.; Saito, Y.; Tamura, Y.; He, P.; Suzuki, M.; Shima, Y.; Yoneda, T.; Hoshino, S.; et al. Propionibacterium acnes-induced hepatic granuloma formation is impaired in mice lacking tetraspanin CD9. J. Pathol. 2005, 206, 486–492. [Google Scholar] [CrossRef]
  114. Beijer, E.; Seldenrijk, K.; Meek, B.; Damen, J.; Quanjel, M.J.R.; Grutters, J.C.; Veltkamp, M. Detection of Cutibacterium acnes in granulomas of patients with either hypersensitivity pneumonitis or vasculitis reveals that its presence is not unique for sarcoidosis. ERJ Open Res. 2021, 7, 00930-2020. [Google Scholar] [CrossRef]
  115. Khan, N.A.; Donatelli, C.V.; Tonelli, A.R.; Wiesen, J.; Neto, M.L.R.; Sahoo, D.; Culver, D.A. Toxicity risk from glucocorticoids in sarcoidosis patients. Respir. Med. 2017, 132, 9–14. [Google Scholar] [CrossRef] [PubMed]
  116. Judson, M.A.; Chaudhry, H.; Louis, A.; Lee, K.; Yucel, R. The effect of corticosteroids on quality of life in a sarcoidosis clinic: The results of a propensity analysis. Respir. Med. 2015, 109, 526–531. [Google Scholar] [CrossRef]
  117. Gottlieb, J.E.; Israel, H.L.; Steiner, R.M.; Triolo, J.; Patrick, H. Outcome in sarcoidosis. Chest 1997, 111, 623–631. [Google Scholar] [CrossRef] [PubMed]
  118. Minami, J.; Eishi, Y.; Ishige, Y.; Kobayashi, I.; Ishige, I.; Kobayashi, D.; Ando, N.; Uchida, K.; Ikeda, S.; Sorimachi, N.; et al. Pulmonary granulomas caused experimentally in mice by a recombinant trigger-factor protein of Propionibacterium acnes. J. Med. Dent. Sci. 2003, 50, 265–274. [Google Scholar]
  119. Miyazaki, E.; Ando, M.; Fukami, T.; Nureki, S.I.; Eishi, Y.; Kumamoto, T. Minocycline for the treatment of sarcoidosis: Is the mechanism of action immunomodulating or antimicrobial effect? Clin. Rheumatol. 2008, 27, 1195–1197. [Google Scholar] [CrossRef]
  120. Takemori, N.; Nakamura, M.; Kojima, M.; Eishi, Y. Successful treatment in a case of Propionibacterium acnes-associated sarcoidosis with clarithromycin administration: A case report. J. Med. Case Rep. 2014, 8, 15. [Google Scholar] [CrossRef]
  121. Ishibashi, K.; Eishi, Y.; Tahara, N.; Asakura, M.; Sakamoto, N.; Nakamura, K.; Takaya, Y.; Nakamura, T.; Yazaki, Y.; Yamaguchi, T.; et al. Japanese antibacterial drug management for cardiac sarcoidosis (J-ACNES): A multicenter, open-label, randomized, controlled study. J. Arrhythmia 2018, 34, 520–526. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Known and suspected disorders associated with Cutibacterium acnes. There is convincing scientific evidence that C. acnes is involved in the development of acne vulgaris and prosthetic infections. Furthermore, associations have been found between the occurrence of prostate cancer or sarcoidosis and the presence of C. acnes, for which a causal role has not yet been demonstrated.
Figure 1. Known and suspected disorders associated with Cutibacterium acnes. There is convincing scientific evidence that C. acnes is involved in the development of acne vulgaris and prosthetic infections. Furthermore, associations have been found between the occurrence of prostate cancer or sarcoidosis and the presence of C. acnes, for which a causal role has not yet been demonstrated.
Microorganisms 10 01649 g001
Figure 3. Lymphocyte percentage in the BAL fluid of HP patients with and without the presence of C. acnes in granulomas. Asterisk represents significant difference. BAL: Broncho alveolar lavage, HP: Hypersensitivity pneumonitis. The results of this study are described in [114].
Figure 3. Lymphocyte percentage in the BAL fluid of HP patients with and without the presence of C. acnes in granulomas. Asterisk represents significant difference. BAL: Broncho alveolar lavage, HP: Hypersensitivity pneumonitis. The results of this study are described in [114].
Microorganisms 10 01649 g003
Figure 4. Flow chart of the PHENOSAR trial. Patients are stratified based on the presence or absence of Cutibacterium acnes in their granulomas. After this stratification, patients are randomized to receive either 500 mg Azithromycin 3 times a week and daily doxycycline, or a placebo for 13 weeks. CA+ = sarcoidosis patients with the presence of C. acnes in granulomatous tissue. CA− = sarcoidosis patients with no C. acnes in their granulomatous tissue. PET/CT = Positive Electron Tomography/Computer Tomography.
Figure 4. Flow chart of the PHENOSAR trial. Patients are stratified based on the presence or absence of Cutibacterium acnes in their granulomas. After this stratification, patients are randomized to receive either 500 mg Azithromycin 3 times a week and daily doxycycline, or a placebo for 13 weeks. CA+ = sarcoidosis patients with the presence of C. acnes in granulomatous tissue. CA− = sarcoidosis patients with no C. acnes in their granulomatous tissue. PET/CT = Positive Electron Tomography/Computer Tomography.
Microorganisms 10 01649 g004
Table 1. Possible antigens in sarcoidosis.
Table 1. Possible antigens in sarcoidosis.
Possible AntigensLiterature (References Are in Parentheses)
Infectious agents
Mycobacterium tuberculosis[72,73,74,75]
Cutibacterium acnes[76,77]
Borrelia[78]
Fungi[79,80,81]
Aspergillus nidulans[82,83]
Non-infectious environmental antigens
 Beryllium[84,85,86,87,88]
 Aluminum[84,88,89]
 Silica[85,88,89,90]
 copper[91]
 titanium[85,89,91,92]
 Combustible wood[93,94,95]
Autoantigens
 vimentin[96,97,98]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kraaijvanger, R.; Veltkamp, M. The Role of Cutibacterium acnes in Sarcoidosis: From Antigen to Treatable Trait? Microorganisms 2022, 10, 1649. https://doi.org/10.3390/microorganisms10081649

AMA Style

Kraaijvanger R, Veltkamp M. The Role of Cutibacterium acnes in Sarcoidosis: From Antigen to Treatable Trait? Microorganisms. 2022; 10(8):1649. https://doi.org/10.3390/microorganisms10081649

Chicago/Turabian Style

Kraaijvanger, Raisa, and Marcel Veltkamp. 2022. "The Role of Cutibacterium acnes in Sarcoidosis: From Antigen to Treatable Trait?" Microorganisms 10, no. 8: 1649. https://doi.org/10.3390/microorganisms10081649

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop