Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 167
1.
Eur J Clin Microbiol Infect Dis ; 41(1): 169-173, 2022 Jan.
Article En | MEDLINE | ID: mdl-34535842

The purpose of this study was to investigate if the C. acnes present at the end of a primary shoulder arthroplasty could be responsible for shoulder arthroplasty infection. Prospective study includes patients undergoing primary shoulder arthroplasty from January 2015 until December 2018. From all the patients included, 5 to 12 tissue samples were obtained and were specifically cultured to detect the presence of C. acnes. DNA was extracted from the C acnes isolated colonies and Whole Genome Sequencing (WGS) analysis was done. A cohort of 156 patients was finally included. In twenty-seven patients, the C. acnes was present at the end of the primary surgery. Two of these patients developed a C. acnes periprosthetic shoulder infection at 6 and 4 months after the primary surgery. WGS of C. acnes isolated colonies showed that all the revision-surgery isolates clustered near to the corresponding primary-surgery isolates compared to the other independent bacterial colonies. (99.89% of similarity). C. acnes present at the end of the primary surgery can be the cause of early or delayed periprosthetic joint infections in shoulder arthroplasty.


Gram-Positive Bacterial Infections/microbiology , Postoperative Complications/microbiology , Propionibacterium acnes/isolation & purification , Shoulder Prosthesis/microbiology , Shoulder/microbiology , Aged , Aged, 80 and over , Arthroplasty/adverse effects , Female , Genome, Bacterial , Humans , Male , Middle Aged , Propionibacterium acnes/genetics , Propionibacterium acnes/growth & development , Prospective Studies , Shoulder/surgery
2.
BMC Microbiol ; 21(1): 165, 2021 06 03.
Article En | MEDLINE | ID: mdl-34082713

BACKGROUND: Lactobacillus rhamnosus GG (LGG) is the most widely used probiotic, but the mechanisms underlying its beneficial effects remain unresolved. Previous studies typically inoculated LGG in hosts with established gut microbiota, limiting the understanding of specific impacts of LGG on host due to numerous interactions among LGG, commensal microbes, and the host. There has been a scarcity of studies that used gnotobiotic animals to elucidate LGG-host interaction, in particular for gaining specific insights about how it modifies the metabolome. To evaluate whether LGG affects the metabolite output of pathobionts, we inoculated with LGG gnotobiotic mice containing Propionibacterium acnes, Turicibacter sanguinis, and Staphylococcus aureus (PTS). RESULTS: 16S rRNA sequencing of fecal samples by Ion Torrent and MinION platforms showed colonization of germ-free mice by PTS or by PTS plus LGG (LTS). Although the body weights and feeding rates of mice remained similar between PTS and LTS groups, co-associating LGG with PTS led to a pronounced reduction in abundance of P. acnes in the gut. Addition of LGG or its secretome inhibited P. acnes growth in culture. After optimizing procedures for fecal metabolite extraction and metabolomic liquid chromatography-mass spectrometry analysis, unsupervised and supervised multivariate analyses revealed a distinct separation among fecal metabolites of PTS, LTS, and germ-free groups. Variables-important-in-projection scores showed that LGG colonization robustly diminished guanine, ornitihine, and sorbitol while significantly elevating acetylated amino acids, ribitol, indolelactic acid, and histamine. In addition, carnitine, betaine, and glutamate increased while thymidine, quinic acid and biotin were reduced in both PTS and LTS groups. Furthermore, LGG association reduced intestinal mucosal expression levels of inflammatory cytokines, such as IL-1α, IL-1ß and TNF-α. CONCLUSIONS: LGG co-association had a negative impact on colonization of P. acnes, and markedly altered the metabolic output and inflammatory response elicited by pathobionts.


Gram-Positive Bacterial Infections/microbiology , Lacticaseibacillus rhamnosus/metabolism , Probiotics/administration & dosage , Animals , Cytokines/genetics , Cytokines/metabolism , Female , Firmicutes/growth & development , Firmicutes/physiology , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Gram-Positive Bacterial Infections/genetics , Gram-Positive Bacterial Infections/metabolism , Humans , Lacticaseibacillus rhamnosus/genetics , Male , Mice , Mice, Inbred C57BL , Propionibacterium acnes/growth & development , Propionibacterium acnes/physiology , Staphylococcus aureus/growth & development , Staphylococcus aureus/physiology
3.
Biomedica ; 40(4): 693-701, 2020 12 02.
Article En, Es | MEDLINE | ID: mdl-33275348

Introduction: Tea tree oil is an essential oil recognized for its antimicrobial properties. Objective: To evaluate the composition, features, and antimicrobial effect at 2% v/v, and its minimal inhibitory concentration (MIC) against Cutibacterium acnes (Propionibacterium acnes). Materials and methods: Three different batches of tea tree oil were evaluated. We characterized its chemotype by gas chromatography and its 2% v/v antimicrobial activity against C. acnes by agar diffusion assay (guide M11-A8 CLSI). Results: The three batches of oil had the chemotypes required by the ISO 4730 standard, which indicates that it is a high-quality product. Additionally, they had 30% to 40% of terpinen-4-ol, a compound that favors its antimicrobial activity. Antimicrobial activity against C. acnes for all batches had a concentration-dependent effect with microbial growth inhibitory activity in all assays at 2% v/v. The MIC obtained against C. acnes for all batches was 0.25% v/v. The antimicrobial activity of tea tree oil against this microorganism has been previously reported with a MIC between 0.05% and 1.25% v/v, a range that covers the one obtained in this study. Conclusion: These results show the high quality of the oil and its capacity as an antibacterial agent against C. acnes. New studies should be conducted to confirm its activity and that of its components in isolates of the microorganism from patients with acne vulgaris.


Introducción. El aceite del árbol de té es un aceite esencial reconocido por sus propiedades antimicrobianas. Objetivos. Evaluar la composición, características y efecto antimicrobiano del aceite al 2 % del árbol de té y su concentración inhibitoria mínima (CIM) contra Cutibacterium acnes (Propionibacterium acnes). Materiales y métodos. Se evaluó el quimiotipo en tres lotes diferentes de este aceite mediante cromatografía de gases, así como su actividad antimicrobiana en concentración al 2 % v/v y la CIM contra C. acnes mediante ensayo de difusión en agar (guía M11-A8 CLSI). Resultados. Los lotes evaluados presentaron los quimiotipos ajustados a la norma ISO 4730, lo que indicó la alta calidad del producto. Los lotes contenían de 30 a 40 % de terpinen-4-ol, compuesto que favorece la actividad antimicrobiana, la cual presentó en todos los lotes un efecto dependiente de la concentración contra C. acnes, con una inhibición del crecimiento microbiano en concentración al 2 % v/v en todas las pruebas. La concentración inhibitoria mínima fue de 0,25 % v/v. La actividad antimicrobiana del aceite del árbol de té contra este microorganismo ya ha sido reportada con una concentración inhibitoria mínima entre 0,05 y 1,25 % v/v, rango que cobija la obtenida en este estudio. Conclusiones. Los resultados evidenciaron la gran calidad de este producto y su capacidad como agente antibacteriano contra C. acnes. Se deben hacer estudios con otros aislamientos del microorganismo provenientes de pacientes con acné vulgar para confirmar su actividad general y la de cada uno de sus componentes.


Anti-Infective Agents, Local/chemistry , Anti-Infective Agents, Local/pharmacology , Propionibacterium acnes/drug effects , Tea Tree Oil/pharmacology , Anti-Infective Agents, Local/standards , Color , Flame Ionization , Microbial Sensitivity Tests , Propionibacterium acnes/growth & development , Refractometry , Tea Tree Oil/administration & dosage , Tea Tree Oil/chemistry , Tea Tree Oil/standards , Terpenes/analysis
4.
Int J Mol Sci ; 21(14)2020 Jul 19.
Article En | MEDLINE | ID: mdl-32707723

The probiotic activity of skin Staphylococcus epidermidis (S. epidermidis) bacteria can elicit diverse biological functions via the fermentation of various carbon sources. Here, we found that polyethylene glycol (PEG)-8 Laurate, a carbon-rich molecule, can selectively induce the fermentation of S. epidermidis, not Cutibacterium acnes (C. acnes), a bacterium associated with acne vulgaris. The PEG-8 Laurate fermentation of S. epidermidis remarkably diminished the growth of C. acnes and the C. acnes-induced production of pro-inflammatory macrophage-inflammatory protein 2 (MIP-2) cytokines in mice. Fermentation media enhanced the anti-C. acnes activity of a low dose (0.1%) clindamycin, a prescription antibiotic commonly used to treat acne vulgaris, in terms of the suppression of C. acnes colonization and MIP-2 production. Furthermore, PEG-8 Laurate fermentation of S. epidermidis boosted the activity of 0.1% clindamycin to reduce the sizes of C. acnes colonies. Our results demonstrated, for the first time, that the PEG-8 Laurate fermentation of S. epidermidis displayed the adjuvant effect on promoting the efficacy of low-dose clindamycin against C. acnes. Targeting C. acnes by lowering the required doses of antibiotics may avoid the risk of creating drug-resistant C. acnes and maintain the bacterial homeostasis in the skin microbiome, leading to a novel modality for the antibiotic treatment of acne vulgaris.


Clindamycin/administration & dosage , Laurates/metabolism , Polyethylene Glycols/metabolism , Propionibacteriaceae/drug effects , Staphylococcus epidermidis/metabolism , Acne Vulgaris/drug therapy , Acne Vulgaris/microbiology , Animals , Anti-Bacterial Agents/administration & dosage , Fermentation , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/microbiology , Humans , In Vitro Techniques , Mice , Mice, Inbred ICR , Probiotics/metabolism , Propionibacteriaceae/growth & development , Propionibacterium acnes/drug effects , Propionibacterium acnes/growth & development , Skin/drug effects , Skin/metabolism , Skin/microbiology
5.
BMC Microbiol ; 20(1): 198, 2020 07 07.
Article En | MEDLINE | ID: mdl-32635901

BACKGROUND: Bacteria involved in ruminal formation of trans-10 intermediates are unclear. Therefore, this study aimed at identifying rumen bacteria that produce trans-10 intermediates from 18-carbon unsaturated fatty acids. RESULTS: Pure cultures of 28 rumen bacterial species were incubated individually in the presence of 40 µg/mL 18:3n-3, 18:2n-6 or trans-11 18:1 under control or lactate-enriched (200 mM Na lactate) conditions for 24 h. Of the 28 strains, Cutibacterium acnes (formerly Propionibacterium acnes) was the only bacterium found to produce trans-10 intermediates from 18:3n-3 and 18:2n-6, irrespective of the growth condition. To further assess the potential importance of this species in the trans-11 to trans-10 shift, different biomass ratios of Butyrivibrio fibrisolvens (as a trans-11 producer) and C. acnes were incubated in different growth media (control, low pH and 22:6n-3 enriched media) containing 40 µg/mL 18:2n-6. Under control conditions, a trans-10 shift, defined in the current study as trans-10/trans-11 ≥ 0.9, occurred when the biomass of C. acnes represented between 90 and 98% of the inoculum. A low pH or addition of 22:6n-3 inhibited cis-9, trans-11 CLA and trans-10, cis-12 CLA formation by B. fibrisolvens and C. acnes, respectively, whereby C. acnes seemed to be more tolerant. This resulted in a decreased biomass of C. acnes required at inoculation to induce a trans-10 shift to 50% (low pH) and 90% (22:6n-3 addition). CONCLUSIONS: Among the bacterial species studied,C. acnes was the only bacterium that have the metabolic ability to produce trans-10 intermediates from 18:3n-3 and 18:2n-6. Nevertheless, this experiment revealed that it is unlikely that C. acnes is the only or predominant species involved in the trans-11 to trans-10 shift in vivo.


Propionibacterium acnes/growth & development , Rumen/microbiology , Trans Fatty Acids/analysis , Animals , Bacteriological Techniques , Biomass , Gas Chromatography-Mass Spectrometry , Hydrogen-Ion Concentration , Propionibacterium acnes/isolation & purification , Propionibacterium acnes/metabolism , Trans Fatty Acids/metabolism
6.
J Microbiol Biotechnol ; 30(9): 1379-1386, 2020 Sep 28.
Article En | MEDLINE | ID: mdl-32627753

Acne is a chronic skin disease that typically occurs in the teens and twenties, and its symptoms vary according to age, sex, diet, and lifestyle. The condition is characterized by hyperproliferation of keratinocytes in the epidermis, sebum overproduction, excessive growth of Propionibacterium acnes, and P. acnes-induced skin inflammation. Interleukin (IL)-1α and IL-6 are predominant in the inflammatory lesions of acne vulgaris. These cytokines induce an inflammatory reaction in the skin in the presence of pathogens or stresses. Moreover, IL-1α accelerates the production of keratin 16, which is typically expressed in wounded or aberrant skin, leading to abnormalities in architecture and hyperkeratinization. Orobol (3',4',5,7-tetrahydroxyisoflavone) is a metabolite of genistein that inhibited the P. acnes-induced increases in IL-6 and IL-1α levels in human keratinocytes (HaCaTs) more effectively compared with salicylic acid. In addition, orobol decreased the IL-1α and IL-6 mRNA levels and inhibited the phosphorylation of inhibitor of kappa-B kinase, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha, and mitogen-activated protein kinase induced by P. acnes. Finally, the expression of Ki67 was decreased by orobol. Thus, orobol ameliorated the inflammation and hyperkeratinization induced by heat-killed P. acnes and thus has potential for use in functional foods and cosmetics.


Anti-Inflammatory Agents/pharmacology , Flavonoids/pharmacology , Keratinocytes/drug effects , Propionibacterium acnes/drug effects , Anti-Inflammatory Agents/chemistry , Cell Proliferation/drug effects , Cytokines/metabolism , Flavonoids/chemistry , Genistein/chemistry , Genistein/pharmacology , HaCaT Cells , Humans , Inflammation , Interleukins/metabolism , Keratinocytes/immunology , Keratinocytes/microbiology , Keratinocytes/pathology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Phosphorylation/drug effects , Propionibacterium acnes/growth & development , Transcription Factor AP-1/metabolism
7.
J Med Microbiol ; 69(5): 689-696, 2020 May.
Article En | MEDLINE | ID: mdl-32375980

Introduction. Rhein (4, 5-dihydroxyanthraquinone-2-carboxylic acid) has various properties, including anti-inflammatory, antioxidant and anticancer activities. However, the mechanism underlying the role of rhein in antimicrobial activity remains largely unknown.Aim. This study aims to identify potential natural compounds of rhein that are capable of inhibiting Cutibacterium acnes and elucidate the effects of rhein on NADH dehydrogenase-2 activity in C. acnes.Methodology. The anti-C. acnes activity of compounds was analysed using minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), the paper disc diffusion test and the checkerboard dilution test. To check whether rhein was inhibitory, putative type II NADH dehydrogenase (NDH-2) of C. acnes was analysed, cloned and expressed in Escherichia coli, and then NDH-2 purification was assessed with Ni-NTA before rhein inhibition of NADH dehydrogenase-2 activity was checked with ferricyanide [K3Fe(CN)6] as a substrate.Results. The results showed that the MIC of rhein against C. acnes was 6.25 µg ml-1, while the MBC was 12.5 µg ml-1, and there was a 38 mm inhibition zone in the paper disc diffusion test. Rhein showed an additive two- to fourfold reduction of the MIC value with four antibiotics on the checkerboard dilution test. The purified NADH dehydrogenase gene product showed a size of approximately 51 kDa and had a V max of 23 µmol and a K m of 280 µm. The inhibitory effect of rhein against NADH dehydrogenase-2 activity was non-competitive with ferricyanide [K3Fe(CN)6] with a K i value of 3.5-4.5 µm.Conclusion. This study provided evidence of the inhibitory effects of rhein on the growth of C. acnes by blocking of NADH dehydrogenase-2 activity. This mechanism of inhibitory activity in the reduction of ROS formation and ATP productivity should be further tested in C. acnes and the question of whether rhein inhibits the natural growth of C. acnes should be investigated.


Anthraquinones/pharmacology , Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Gram-Positive Bacterial Infections/microbiology , NADH Dehydrogenase/antagonists & inhibitors , Propionibacterium acnes/drug effects , Propionibacterium acnes/enzymology , Anthraquinones/therapeutic use , Anti-Bacterial Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Enzyme Activation/drug effects , Enzyme Inhibitors/therapeutic use , Gram-Positive Bacterial Infections/drug therapy , Humans , Kinetics , Microbial Sensitivity Tests , Microbial Viability/drug effects , Propionibacterium acnes/growth & development , Reactive Oxygen Species/metabolism , Recombinant Proteins
8.
Sci Rep ; 10(1): 7491, 2020 05 04.
Article En | MEDLINE | ID: mdl-32367064

Glycolic acid is the smallest alpha hydroxy acid and widely used for skincare applications, including to treat acne vulgaris. Oftentimes, high concentrations of glycolic acid (~20-50 vol%) are incorporated into chemical peels to reduce acne-related inflammation while there is an outstanding need to determine to what extent glycolic acid can potently inhibit Cutibacterium acnes (formerly known as Propionibacterium acnes), which is a Gram-positive bacterium implicated in acne pathogenesis. Herein, we report that glycolic acid exhibits pH-dependent antibacterial activity against C. acnes and mechanistic studies identified that the nonionic form of glycolic acid is more active than the anionic form. The degree of antibacterial activity, including minimum bactericidal concentration (MBC), of glycolic acid was evaluated in the pH range of 3 to 4.5, and the greatest potency was observed at pH 3. In light of skincare formulation needs, we selected the pH 3.5 condition for further testing and determined that glycolic acid kills C. acnes cells by disrupting bacterial cell membranes. While most conventional treatments involve high concentrations of glycolic acid (>20%), our findings support the potential of developing anti-acne formulations with glycolic acid concentrations as low as 0.2% and with pH conditions that are suitable for over-the-counter applications.


Anti-Bacterial Agents/pharmacology , Glycolates/pharmacology , Propionibacterium acnes/growth & development , Acne Vulgaris/drug therapy , Acne Vulgaris/microbiology , Humans , Hydrogen-Ion Concentration
9.
PDA J Pharm Sci Technol ; 74(5): 592-601, 2020.
Article En | MEDLINE | ID: mdl-31843987

Mammalian cell lines constitute an important part in the manufacture of therapeutic proteins. A contamination-free operation of a mammalian cell bioreactor demands constant attention to details. Cutibacterium acnes, a slow-growing bacterium, is a common skin flora and is often associated with mammalian cell bioreactor contaminations. This paper reviews the literature published about such C. acnes contaminations and presents three hypothetical contamination scenarios based on the author's experience in the hope of fostering early detection of contamination events. Bioreactor process parameters such as unusual fluctuation in dissolved oxygen, high levels of ammonia, and microscopic examination have been identified as useful tools to detect slow-growing bacteria contamination.


Bacteriological Techniques , Bioreactors/microbiology , Cell Culture Techniques/instrumentation , Equipment Contamination , Propionibacterium acnes/isolation & purification , Animals , Cell Line , Humans , Kinetics , Propionibacterium acnes/growth & development
10.
Int J Mol Sci ; 20(14)2019 Jul 17.
Article En | MEDLINE | ID: mdl-31319552

Acne is an inflammatory skin disorder in puberty with symptoms including papules, folliculitis, and nodules. Propionibacterium acnes (P. acnes) is the main anaerobic bacteria that cause acne. It is known to proliferate within sebum-blocked skin hair follicles. P. acnes activates monocytic cell immune responses to induce the expression of proinflammatory cytokines. Although the anti-inflammatory function of the Laurus nobilis (L. nobilis) extract (LNE) on several immunological disorders have been reported, the effect of LNE in P. acnes-mediated skin inflammation has not yet been explored. In the present study, we examined the ability of the LNE to modulate the P. acnes-induced inflammatory signaling pathway, and evaluated its mechanism. LNE significantly suppressed the expression of P. acnes-mediated proinflammatory cytokines, such as IL-1ß, IL-6, and NLRP3. We also found that LNE inhibited the inflammatory transcription factor NF-κB in response to P. acnes. In addition, eucalyptol, which is the main constituent of LNE, consistently inhibited P. acnes-induced inflammatory signaling pathways. Moreover, LNE significantly ameliorated P. acnes-induced inflammation in a mouse model of acne. We suggest for the first time that LNE hold therapeutic value for the improvement of P. acnes-induced skin inflammation.


Acne Vulgaris/drug therapy , Anti-Inflammatory Agents/pharmacology , Eucalyptol/pharmacology , Gram-Positive Bacterial Infections/drug therapy , Laurus/chemistry , Plant Extracts/pharmacology , Propionibacterium acnes/growth & development , Acne Vulgaris/metabolism , Acne Vulgaris/microbiology , Acne Vulgaris/pathology , Animals , Anti-Inflammatory Agents/chemistry , Cell Line , Eucalyptol/chemistry , Gram-Positive Bacterial Infections/metabolism , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/pathology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Mice , Plant Extracts/chemistry
11.
Article En | MEDLINE | ID: mdl-31058101

Microbiota is a crucial player in gynecologic health, in which bacteria can shift to a dysbiotic state triggering a pathogenic process. Based on an ecological understanding of the problem, the aim of this study is to select a potential probiotic strain to improve female reproductive tract based on its capacity to initially lower pH and to promote the reduction of pathogenic bacteria. Based on this rationale, strain Lactobacillus rhamnosus BPL005 was initially selected for its capacity to reduce in vitro pH levels and produce organic acids. Subsequently, strain L. rhamnosus BPL005 (CECT 8800) was demonstrated to have a protective role on endometrial infections in an in vitro model of bacterial colonization of primary endometrial epithelial cells with Atopobium vaginae, Gardnerella vaginalis, Propionibacterium acnes, and Streptococcus agalactiae. In this model, BPL005 when co-cultured with those pathogens was shown to lower pH and to produce organic acids, being lactic acid the most relevant. The co-cultivation of strain L. rhamnosus BPL005 with tested reference pathogens produced a significant reduction in P. acnes and St. agalactiae levels and a non-significant reduction in A. vaginae and G. vaginalis. The colonization of L. rhamnosus BPL005 in the culture decreased IL-6, IL-8, and MCP-1, heightened in the presence of pathogens, and increased IL-1RA and IL-1 beta. Finally, safety was evaluated showing no signs of cytotoxicity, irritation in vaginal tests, or allergic contact dermatitis potential through the Local Lymph Node Assay. Overall, these results show the potential of L. rhamnosus BPL005 strain as a probiotic in gynecological health.


Antibiosis , Genitalia, Female/microbiology , Lacticaseibacillus rhamnosus/growth & development , Lacticaseibacillus rhamnosus/isolation & purification , Microbiota , Probiotics/isolation & purification , Reproductive Tract Infections/microbiology , Actinobacteria/growth & development , Carboxylic Acids/metabolism , Cells, Cultured , Epithelial Cells/microbiology , Female , Gardnerella vaginalis/growth & development , Humans , Hydrogen-Ion Concentration , Lacticaseibacillus rhamnosus/metabolism , Propionibacterium acnes/growth & development , Streptococcus agalactiae/growth & development
12.
Microbiologyopen ; 8(9): e00841, 2019 09.
Article En | MEDLINE | ID: mdl-30950214

Cutibacterium acnes, former Proprionibacterium acnes, is a heterogeneous species including acneic bacteria such as the RT4 strain, and commensal bacteria such as the RT6 strain. These strains have been characterized by metagenomic analysis but their physiology was not investigated until now. Bacteria were grown in different media, brain heart infusion medium (BHI), reinforced clostridial medium (RCM), and in sebum like medium (SLM) specifically designed to reproduce the lipid rich environment of the sebaceous gland. Whereas the RT4 acneic strain showed maximal growth in SLM and lower growth in RCM and BHI, the RT6 non acneic strain was growing preferentially in RCM and marginally in SLM. These differences were correlated with the lipophilic surface of the RT4 strain and to the more polar surface of the RT6 strain. Both strains also showed marked differences in biofilm formation activity which was maximal for the RT4 strain in BHI and for the RT6 strain in SLM. However, cytotoxicity of both strains on HaCaT keratinocytes remained identical and limited. The RT4 acneic strain showed higher inflammatory potential than the RT6 non acneic strain, but the growth medium was without significant influence. Both bacteria were also capable to stimulate ß-defensine 2 secretion by keratinocytes but no influence of the bacterial growth conditions was observed. Comparative proteomics analysis was performed by nano LC-MS/MS and revealed that whereas the RT4 strain only expressed triacylglycerol lipase, the principal C. acnes virulence factor, when it was grown in SLM, the RT6 strain expressed another virulence factor, the CAMP factor, exclusively when it was grown in BHI and RCM. This study demonstrates the key influence of growth conditions on virulence expression by C. acnesand suggest that acneic and non acneic strains are related to different environmental niches.


Adaptation, Physiological , Propionibacterium acnes/growth & development , Propionibacterium acnes/metabolism , Sebum/microbiology , Bacterial Proteins/analysis , Cell Line , Culture Media/chemistry , Humans , Keratinocytes/immunology , Keratinocytes/microbiology , Propionibacterium acnes/chemistry , Proteome/analysis , Virulence Factors/analysis
13.
Br J Pharmacol ; 176(13): 2321-2335, 2019 07.
Article En | MEDLINE | ID: mdl-30927447

BACKGROUND AND PURPOSE: Propionibacterium acnes is a Gram-positive bacterium associated with the skin disorder acne. In this study, as fatty acids are considered to be important in the life habitat of P. acnes, we tested our lipopeptide library in an attempt to create potent P. acnes-specific antimicrobial agents. EXPERIMENTAL APPROACH: The antimicrobial activity of various lipopeptides was determined by measuring their minimal inhibitory concentration (MIC). Lipids from P. acnes were used to explore their mode of action. RAW264.7 cells stimulated with LPS and P. acnes respectively were used to measure their anti-inflammatory activity. Mice ears injected with P. acnes were used to assess the antimicrobial and anti-inflammatory effects of the peptides tested in vivo. KEY RESULTS: The most potent candidate, C16-KWKW, was observed to be more active against P. acnes than against other non-targeted bacterial strains, such as Streptococcus mutans, Staphylococcus aureus, and Escherichia coli. The mode of action of C16-KWKW was observed to be through interference with the integrity of the bacterial membrane, thereby impairing membrane permeability and causing leakage of inner contents of bacterial cells. Furthermore, C16-KWKW inhibited the expression of pro-inflammatory cytokines, such as IL-1ß, TNF-α, and inducible NOS stimulated by both LPS and P. acnes, thus showing potential anti-inflammatory activity, which was further verified in the in vivo animal studies. CONCLUSIONS AND IMPLICATIONS: C16-KWKW is a lipopeptide displaying both anti-P. acnes and anti-inflammatory effects in vitro and in vivo and shows potential as a treatment for acne vulgaris induced by P. acnes.


Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Lipopeptides/pharmacology , Propionibacterium acnes/drug effects , Acne Vulgaris/drug therapy , Animals , Cell Line , Cyclooxygenase 2/genetics , Female , Humans , Interleukin-1beta/genetics , Mice , Microbial Sensitivity Tests , NF-kappa B/genetics , Nitric Oxide Synthase Type II/genetics , Propionibacterium acnes/growth & development , Tumor Necrosis Factor-alpha/genetics
14.
Microbiologyopen ; 8(3): e00659, 2019 03.
Article En | MEDLINE | ID: mdl-29911330

Increasing popularity of preservative-free cosmetics necessitates in-depth research, specifically as bacteria can react to local factors by important metabolic changes. In this respect, investigating the effect of cosmetic preparations on pathogenic strains of commensal species such as acneic forms of Cutibacterium acnes (former Propionibacterium acnes) and bacteria behaving both as commensals and opportunistic pathogens such as Staphylococcus aureus is of major interest. In this study, we studied the effect of commonly used cosmetics, Uriage™ thermal water (UTW) and a rhamnose-rich polysaccharide (PS291® ) on RT4 and RT5 acneic strains of C. acnes and a cutaneous strain of S. aureus. UTW affected the growth kinetic of acneic C. acnes essentially by increasing its generation time and reducing its biomass, whereas only the S. aureus final biomass was decreased. PS291 had more marginal effects. Both compounds showed a marked antibiofilm activity on C. acnes and S. aureus. For S. aureus that appeared essentially due to inhibition of initial adhesion. Cosmetics did not modify the metabolic activity of bacteria. Both C. acnes and S. aureus showed marked hydrophobic surface properties. UTW and PS291 had limited effect on C. acnes but increased the hydrophobic character of S. aureus. This work underlines the effect of cosmetics on cutaneous bacteria and the potential limitations of preservative-free products.


Biofilms/drug effects , Biofilms/growth & development , Cosmetics/metabolism , Propionibacterium acnes/drug effects , Propionibacterium acnes/growth & development , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Bacterial Adhesion/drug effects , Metabolism/drug effects
15.
Nat Commun ; 9(1): 5075, 2018 12 12.
Article En | MEDLINE | ID: mdl-30542056

Acne vulgaris is a highly heritable common, chronic inflammatory disease of the skin for which five genetic risk loci have so far been identified. Here, we perform a genome-wide association study of 3823 cases and 16,144 controls followed by meta-analysis with summary statistics from a previous study, with a total sample size of 26,722. We identify 20 independent association signals at 15 risk loci, 12 of which have not been previously implicated in the disease. Likely causal variants disrupt the coding region of WNT10A and a P63 transcription factor binding site in SEMA4B. Risk alleles at the 1q25 locus are associated with increased expression of LAMC2, in which biallelic loss-of-function mutations cause the blistering skin disease epidermolysis bullosa. These findings indicate that variation affecting the structure and maintenance of the skin, in particular the pilosebaceous unit, is a critical aspect of the genetic predisposition to severe acne.


Acne Vulgaris/genetics , Acne Vulgaris/pathology , Genetic Predisposition to Disease/genetics , Hair Follicle/growth & development , Female , Genetic Variation/genetics , Genome-Wide Association Study , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/pathology , Humans , Laminin/biosynthesis , Laminin/genetics , Male , Membrane Proteins/metabolism , Propionibacterium acnes/growth & development , Semaphorins/genetics , Skin/pathology , Wnt Proteins/genetics
16.
Int J Mol Sci ; 19(11)2018 Nov 03.
Article En | MEDLINE | ID: mdl-30400322

Kaempferia parviflora, referred to as black ginger, has traditionally been used as a health-promoting alternative medicine. In this study, we examined the anti-inflammatory, sebostatic, and anti-Propionibacterium acnes activities of K. parviflora extract. The extract significantly down-regulated the expression of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2), and pro-inflammatory cytokine tumor necrosis factor alpha (TNF-α) level. Moreover, the phosphorylation of IĸBα and nuclear factor-kappa B (NF-κB), and the enhanced nuclear translocation of NF-κB p65 in lipopolysaccharide-stimulated murine macrophage-like cell line (RAW 264.7) cells were markedly decreased by the extract. Notably, the main component of K. parviflora, 5,7-dimethoxyflavone, also modulated the expression of iNOS and NF-κB signal molecules in P. acnes-stimulated human keratinocyte (HaCaT) cells. Additionally, K. parviflora extract inhibited the lipogenesis of sebocytes, as evidenced by a reduced level of triglyceride and lipid accumulation in the sebocytes. The sebostatic effect was also confirmed by a reduced expression of peroxisome proliferation-activating receptors (PPAR-γ) and oil-red O staining in sebocytes. Taken together, this study suggests for the first time that K. parviflora extract could be developed as a potential natural anti-acne agent with anti-inflammatory, sebostatic, and anti-P. acnes activity.


Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Flavonoids/pharmacology , Keratinocytes/drug effects , Propionibacterium acnes/drug effects , Zingiberaceae/chemistry , Animals , Anti-Bacterial Agents/isolation & purification , Anti-Inflammatory Agents/isolation & purification , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Flavonoids/isolation & purification , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Mice , Microbial Sensitivity Tests , NF-KappaB Inhibitor alpha/genetics , NF-KappaB Inhibitor alpha/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Plant Extracts/chemistry , Propionibacterium acnes/growth & development , RAW 264.7 Cells , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
17.
Immunopharmacol Immunotoxicol ; 40(5): 415-422, 2018 Oct.
Article En | MEDLINE | ID: mdl-30422030

Objective: Acne is a chronic skin disease that involves four key pathogenic factors: excess sebum production, ductal epidermal hyperproliferation, Propionibacterium acnes (P. acnes) colonization, and skin inflammation. Mangostins are well-known for their anti-bacterial and anti-inflammatory effects, suggesting that mangostins may have therapeutic potential for acne. The present study aimed to explore the anti-acne effects of mangostins from the perspective of multiple pathogenic mechanisms of acne. Methods: The effects of α- and γ-mangostins on the growth of P. acnes and lipase activity were analyzed. Their effects on P. acnes-induced keratinocyte proliferation were examined by CCK-8. The expression of inflammatory genes and activation of NF-κB and MAPK signaling pathways were detected by quantitative real-time PCR and western blotting, respectively. Results: Alpha- and γ-mangostins not only inhibited the growth of P. acnes, but also reduced the proliferation of keratinocytes induced by heat-killed P. acnes. Furthermore, α- and γ-mangostins were able to suppress P. acnes-induced expression of pro-inflammatory cytokines, including TNF-α, IL-1ß, and IL-6 in keratinocytes by inhibiting the activation of NF-κB and MAPK signaling pathways. Discussion and conclusions: Mangostins appeared to possess multiple anti-acne activities, including the inhibition of P. acnes growth, regulation of keratinocytes proliferation, and attenuation of skin inflammatory reaction. Hence, mangostins might be developed into a potential therapeutic agent for the treatment of acne.


Anti-Bacterial Agents/pharmacology , Keratinocytes/drug effects , Propionibacterium acnes/drug effects , Xanthones/pharmacology , Acne Vulgaris/immunology , Acne Vulgaris/microbiology , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/genetics , Cytokines/immunology , Dose-Response Relationship, Drug , Gene Expression/drug effects , Humans , Inflammation , Keratinocytes/immunology , Keratinocytes/microbiology , Lipase/metabolism , Microbial Sensitivity Tests , Propionibacterium acnes/enzymology , Propionibacterium acnes/growth & development
18.
Int J Antimicrob Agents ; 52(6): 942-946, 2018 Dec.
Article En | MEDLINE | ID: mdl-30144502

The aim of this study was to investigate the ability of Candida albicans and Cutibacterium acnes to grow together as a polymicrobial biofilm in vitro and to examine the influence of C. acnes on C. albicans susceptibility to micafungin. Mature 72-h-old single-species biofilms of C. albicans and polymicrobial biofilms involving both C. albicans and C. acnes were formed in brain-heart infusion and were observed by scanning electronic microscopy. Moreover, 24-h-old single-species and polymicrobial biofilms were treated for 24 h with micafungin (concentrations ranging from 0.75 mg/L to 12 mg/L) and the antibiofilm activity of micafungin was evaluated on fungal cells by flow cytometry following addition of propidium iodide. The results showed that C. albicans and C. acnes formed a polymicrobial biofilm in the tested conditions and that bacterial presence did not modify fungal viability. Micafungin induced a fungal mortality rate ranging from 70-95% in C. albicans single-species biofilms and from 35-40% in C. acnes-C. albicans polymicrobial biofilms. Mortality induced by micafungin was significantly reduced (P < 0.05 for micafungin at 6 mg/L and P < 0.001 for other micafungin concentrations) in polymicrobial conditions compared with single-species biofilms. In conclusion, this study showed that C. albicans and C. acnes are able to form polymicrobial biofilms together in a synergistic way and that this organisation increases yeast resistance to micafungin.


Antifungal Agents/pharmacology , Biofilms/growth & development , Candida albicans/drug effects , Micafungin/pharmacology , Microbial Interactions , Microbial Viability/drug effects , Propionibacterium acnes/metabolism , Antifungal Agents/metabolism , Biofilms/drug effects , Candida albicans/growth & development , Flow Cytometry , Humans , Micafungin/metabolism , Microbiological Techniques , Microscopy, Electron, Scanning , Propionibacterium acnes/growth & development
19.
Curr Probl Dermatol ; 54: 115-122, 2018.
Article En | MEDLINE | ID: mdl-30130780

Acne is based on a complex, multifactorial pathophysiology beginning with a microcomedo. Comedogenesis involves follicular hyperproliferation and disturbed keratinization, hyperseborrhea and hyperplasia of sebaceous glands as well as disturbances in skin microbiome. Acne is treated with antibiotics, retinoids, keratolytics, hormonal and anti-inflammatory agents. Efficacy and side effects of given medications are well known. The uppermost layer of the stratum corneum is acidic. The low pH provides protection by slowing down the growth of some bacteria. Increase of skin surface pH leads to impaired barrier function, disturbances in skin microbiome and inflammation. Acne-predisposed skin is in a constant state of subclinical inflammation. Subclinical inflammation may be linked to changes in skin surface pH and disturbances of the stratum corneum, allowing microorganisms to stimulate the production of pro-inflammatory cytokines. Here, based on the current literature, the possible link between the skin surface pH, epidermal barrier function and acne is reviewed.


Acne Vulgaris/metabolism , Skin/metabolism , Acne Vulgaris/etiology , Acne Vulgaris/therapy , Aging/metabolism , Animals , Female , Humans , Hydrogen-Ion Concentration , Male , Propionibacterium acnes/growth & development , Propionibacterium acnes/pathogenicity , Sex Characteristics , Skin/chemistry , Skin Pigmentation
20.
Lett Appl Microbiol ; 67(5): 497-505, 2018 Nov.
Article En | MEDLINE | ID: mdl-30099746

A healthy skin provides a protective barrier against pathogenic micro-organisms. Recent studies have shown that probiotics, as those of Bifidobacterium genus, could act beneficially in dermatology, both when ingested and by topical use. In the present study, we evaluated by in vitro antagonism assays and using two skin cell lines the potential of four strains of Bifidobacterium spp. Among the four bifidobacteria, Bifidobacterium longum 51A was the only one able to inhibit the growth of the eight pathogenic indicators tested. Production of some cytokines and extracellular matrix proteins was determined when ccc or inactivated cells of the bifidobacteria were incubated with keratinocyte and/or fibroblast cell cultures. Significant results were observed only for IL-6, IL-8 and IL-18 production, and inactivated Bifidobacterium pseudolongum 1191A was the only one which significantly stimulated collagen production, whereas lumican was stimulated by treatments with live Bifidobacterium bifidum 1622A , B. longum 51A and B. pseudolongum 1191A . Highest adhesion and internalization capabilities were observed with B. bifidum 1622A and Bifidobacterium breve 1101A . Concluding, B. longum 51A was highlighted for its antagonistic capacity and B. bifidum 1622A and B. pseudolongum 1191A for stimulating the production of cytokines and proteins of the extracellular matrix. SIGNIFICANCE AND IMPACT OF THE STUDY: The skin is the first line of defence against invasive micro-organisms, and its local microbiota provides additional protective functions based on antagonism against pathogenic micro-organisms and immunomodulation. Based on in vitro assays using Bifidobacterium spp. we demonstrated the antagonistic potential, as well as capacity in stimulating the production of cytokines and proteins of the extracellular matrix that these bacteria may exert on skin cells. This positive influence suggests the use of a consortium of these bifidobacteria in a topical product for dermatological treatments.


Antibiosis/physiology , Bifidobacterium/metabolism , Cytokines/metabolism , Extracellular Matrix Proteins/metabolism , Probiotics/metabolism , Skin/microbiology , Bifidobacterium/classification , Candida albicans/growth & development , Cell Line , Humans , Malassezia/growth & development , Propionibacterium acnes/growth & development , Staphylococcus aureus/growth & development , Staphylococcus epidermidis/growth & development
...