Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 835
Filter
1.
Acta Derm Venereol ; 104: adv33206, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38751177

ABSTRACT

The relevance of the gut microbiota in some skin inflammatory diseases, including acne vulgaris, has been emphasized. Probiotics could play a role in the modulation of the microbiota, improving the clinical course of this disease. A 12-week randomized, double-blind, placebo-controlled, clinical trial with patients aged 12 to 30 years with acne vulgaris was conducted. The study product was a capsule composed of the probiotic Lacticaseibacillus rhamnosus (CECT 30031) and the cyanobacterium Arthrospira platensis (BEA_IDA_0074B). Patients with improvement in the Acne Global Severity Scale were 10/34 (29.41%) in the placebo group compared with 20/40 (50%) in the probiotic group (p = 0.03). A significant reduction (p = 0.03) in the number of non-inflammatory acne lesions was observed in the probiotic group (-18.60 [-24.38 to -12.82]) vs the placebo group (-10.54 [-17.43 to -3.66]). Regarding the number of total  lesions, a reduction almost reaching statistical significance (p = 0.06) was observed in the probiotic group (-27.94 [-36.35 to -19.53]) compared with the placebo group (-18.31 [-28.21 to -8.41]). In addition, patients with improvement attending the Global Acne Grading System were 7/34 (20.58%) in the placebo group vs 17/40 (42.50%) in the probiotic group (p = 0.02). The number of adverse events was similar in both groups. The probiotic used in this study was effective and well tolerated, and it should be considered for acne vulgaris patients.


Subject(s)
Acne Vulgaris , Lacticaseibacillus rhamnosus , Probiotics , Humans , Probiotics/administration & dosage , Probiotics/adverse effects , Probiotics/therapeutic use , Acne Vulgaris/microbiology , Acne Vulgaris/therapy , Acne Vulgaris/drug therapy , Acne Vulgaris/diagnosis , Double-Blind Method , Adolescent , Male , Young Adult , Female , Adult , Treatment Outcome , Child , Administration, Oral , Severity of Illness Index , Gastrointestinal Microbiome/drug effects , Time Factors
2.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38731983

ABSTRACT

Acne vulgaris is a prevalent skin disorder affecting many young individuals, marked by keratinization, inflammation, seborrhea, and colonization by Cutibacterium acnes (C. acnes). Ellagitannins, known for their antibacterial and anti-inflammatory properties, have not been widely studied for their anti-acne effects. Chestnut (Castanea sativa Mill., C. sativa), a rich ellagitannin source, including castalagin whose acne-related bioactivity was previously unexplored, was investigated in this study. The research assessed the effect of C. sativa leaf extract and castalagin on human keratinocytes (HaCaT) infected with C. acnes, finding that both inhibited IL-8 and IL-6 release at concentrations below 25 µg/mL. The action mechanism was linked to NF-κB inhibition, without AP-1 involvement. Furthermore, the extract displayed anti-biofilm properties and reduced CK-10 expression, indicating a potential role in mitigating inflammation, bacterial colonization, and keratosis. Castalagin's bioactivity mirrored the extract's effects, notably in IL-8 inhibition, NF-κB inhibition, and biofilm formation at low µM levels. Other polyphenols, such as flavonol glycosides identified via LC-MS, might also contribute to the extract's biological activities. This study is the first to explore ellagitannins' potential in treating acne, offering insights for developing chestnut-based anti-acne treatments pending future in vivo studies.


Subject(s)
Acne Vulgaris , Fagaceae , Hydrolyzable Tannins , Plant Extracts , Plant Leaves , Humans , Hydrolyzable Tannins/pharmacology , Fagaceae/chemistry , Acne Vulgaris/microbiology , Acne Vulgaris/drug therapy , Plant Extracts/pharmacology , Plant Extracts/chemistry , Plant Leaves/chemistry , Keratinocytes/drug effects , Keratinocytes/metabolism , NF-kappa B/metabolism , HaCaT Cells , Propionibacterium acnes/drug effects , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Interleukin-8/metabolism
3.
Z Naturforsch C J Biosci ; 79(3-4): 47-60, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38549398

ABSTRACT

Garcinia mangostana fruits are used traditionally for inflammatory skin conditions, including acne. In this study, an in silico approach was employed to predict the interactions of G. mangostana xanthones and benzophenones with three proteins involved in the pathogenicity of acne, namely the human JNK1, Cutibacterium acnes KAS III and exo-ß-1,4-mannosidase. Molecular docking analysis was performed using Autodock Vina. The highest docking scores and size-independent ligand efficiency values towards JNK1, C. acnes KAS III and exo-ß-1,4-mannosidase were obtained for garcinoxanthone T, gentisein/2,4,6,3',5'-pentahydroxybenzophenone and mangostanaxanthone VI, respectively. To the best of our knowledge, this is the first report of the potential of xanthones and benzophenones to interact with C. acnes KAS III. Molecular dynamics simulations using GROMACS indicated that the JNK1-garcinoxanthone T complex had the highest stability of all ligand-protein complexes, with a high number of hydrogen bonds predicted to form between this ligand and its target. Petra/Osiris/Molinspiration (POM) analysis was also conducted to determine pharmacophore sites and predict the molecular properties of ligands influencing ADMET. All ligands, except for mangostanaxanthone VI, showed good membrane permeability. Garcinoxanthone T, gentisein and 2,4,6,3',5'-pentahydroxybenzophenone were identified as the most promising compounds to explore further, including in experimental studies, for their anti-acne potential.


Subject(s)
Acne Vulgaris , Benzophenones , Garcinia mangostana , Molecular Docking Simulation , Xanthones , Xanthones/chemistry , Xanthones/pharmacology , Benzophenones/chemistry , Benzophenones/pharmacology , Garcinia mangostana/chemistry , Humans , Acne Vulgaris/drug therapy , Acne Vulgaris/microbiology , Molecular Dynamics Simulation , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 8/chemistry , Computer Simulation , Hydrogen Bonding
4.
Microb Pathog ; 189: 106598, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38423403

ABSTRACT

Propionibacterium acnes (P. acnes) is an anaerobic and gram-positive bacterium involved in the pathogenesis and inflammation of acne vulgaris. This study particularly focuses on the antimicrobial effect of Lacticaseibacillus paracasei LPH01 against P. acnes, a bacterium that causes acne vulgaris. Fifty-seven Lactobacillus strains were tested for their ability to inhibit P. acnes growth employing the Oxford Cup and double dilution methods. The cell-free supernatant (CFS) of L. paracasei LPH01 demonstrated a strong inhibitory effect, with an inhibition zone diameter of 24.65 ± 0.27 mm and a minimum inhibitory concentration of 12.5 mg/mL. Among the CFS, the fraction over 10 kDa (CFS-10) revealed the best antibacterial effect. Confocal laser scanning microscopes and flow cytometry showed that CFS-10 could reduce cell metabolic activity and cell viability and destroy the integrity and permeability of the cell membrane. A scanning electron microscope revealed that bacterial cells exhibited obvious morphological and ultrastructural changes, which further confirmed the damage of CFS-10 to the cell membrane and cell wall. Findings demonstrated that CFS-10 inhibited the conversion of triglycerides, decreased the production of free fatty acids, and down-regulated the extracellular expression of the lipase gene. This study provides a theoretical basis for the metabolite of L. paracasei LPH01 as a potential antibiotic alternative in cosmeceutical skincare products.


Subject(s)
Acne Vulgaris , Lacticaseibacillus paracasei , Humans , Propionibacterium acnes , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Acne Vulgaris/drug therapy , Acne Vulgaris/microbiology , Inflammation/drug therapy , Microbial Sensitivity Tests
5.
J Cosmet Dermatol ; 23(6): 2156-2169, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38406887

ABSTRACT

BACKGROUND: Acne is a common skin issue that typically occurs during adolescence. It causes long-lasting redness and swelling in the skin. An alternative approach to treating acne could involve using a cosmetic facial mask containing herbal ingredients such as Curcumin and Rosa Damascena extract for its antibacterial properties. AIMS: This study aims to create and try out a peel-off mask gel made from Curcumin and R. Damascena extract. This gel is intended to have the ability to kill bacteria such as Staphylococcus aureus, Escherichia coli, and Propionibacterium acnes and remove dead cells from the skin surface. METHODS: The peel-off mask was made using polyvinyl alcohol (PVA) in 8% and 10% as solidifier. The evaluation of peel-off masks comprises the examination of physiochemical and mechanical aspects. Furthermore, their longevity, effectiveness, and antibacterial properties are also considered. RESULTS: The white color, pleasant smell, and soft texture were the defining features of the peel-off gel mask. The changes in PVA affect the pH level, thickness, and how quickly the peel-off mask dries. The stability test found that the peel-off mask had no significant physical changes when exposed to freezing and thawing. However, there were some differences in color and separation when using the real-time method. A prepared peel-off mask containing 10% PVA and curcumin works best against P. acne. The amount of PVA in the formula affected the physical and chemical qualities, but it did not impact on the antibacterial abilities of the peel-off mask gel. The best formula that gives the best results uses 10% PVA + curcumin. CONCLUSIONS: Using the Curcumin and R. Damascena extract in the creation of the peel-off mask gel ensures its efficacy and safety for skin application.


Subject(s)
Acne Vulgaris , Anti-Bacterial Agents , Antioxidants , Curcumin , Plant Extracts , Rosa , Staphylococcus aureus , Curcumin/pharmacology , Curcumin/administration & dosage , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Rosa/chemistry , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/administration & dosage , Antioxidants/pharmacology , Antioxidants/administration & dosage , Acne Vulgaris/drug therapy , Acne Vulgaris/microbiology , Staphylococcus aureus/drug effects , Propionibacterium acnes/drug effects , Polyvinyl Alcohol/chemistry , Escherichia coli/drug effects , Skin Cream/administration & dosage , Skin/drug effects , Skin/microbiology , Microbial Sensitivity Tests
6.
Australas J Dermatol ; 65(4): 319-327, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38419189

ABSTRACT

BACKGROUND: Numerous inflammatory skin diseases are associated with the gut microbiota. Studies of the association between gut microbiota and inflammatory skin diseases have yielded conflicting results owing to confounding factors, and the causal relationship between them remains undetermined. METHODS: Two-sample Mendelian randomization (MR) was used to examine the association between gut microbiota and four common inflammatory skin diseases: acne, psoriasis, urticaria and atopic dermatitis. The summary statistics of the gut microbiota from the largest available genome-wide association study meta-analysis (n = 13,266) conducted by the MiBioGen consortium along with the summary statistics of the four diseases were obtained from the FinnGen consortium. Causal relationships were assessed using the inverse variance weighted (IVW), weighted median, MR-Egger and maximum likelihood methods, and several sensitivity analyses were performed to ensure the accuracy of the results. Finally, reverse and multivariable MR analyses were performed to verify the robustness of the results. RESULTS: We found causal associations of Bacteroidaceae [odds ratio (OR), 2.25; 95% confidence interval (CI), 1.48-3.42; pivw = 0.0001], Allisonella (OR, 1.42; 95% CI, 1.18-1.70; pivw = 0.0002) and Bacteroides (OR, 2.25; 95% CI, 1.48-3.42; pivw = 0.0001) with acne, the Eubacterium fissicatena group with psoriasis (OR, 1.22; 95% CI, 1.10-1.35; pivw = 0.0002) and Intestinibacter with urticaria (OR, 1.28; 95% CI, 1.13-1.45; pivw = 0.0001). These results were corrected for a false discovery rate. Sensitivity analyses were performed to validate the robustness of the associations and reverse MR confirmed that the results were not influenced by the reverse effect. CONCLUSION: Our study revealed that some gut microbiota are risk factors for inflammatory skin diseases, providing new information on potential therapeutic targets. Additionally, a possible association with the gut-skin axis was confirmed. Further research is required to elucidate the mechanisms underlying these relationships.


Subject(s)
Acne Vulgaris , Dermatitis, Atopic , Gastrointestinal Microbiome , Mendelian Randomization Analysis , Psoriasis , Humans , Gastrointestinal Microbiome/genetics , Psoriasis/microbiology , Psoriasis/genetics , Acne Vulgaris/microbiology , Dermatitis, Atopic/microbiology , Urticaria/microbiology , Genome-Wide Association Study
7.
Sci Rep ; 14(1): 4529, 2024 02 24.
Article in English | MEDLINE | ID: mdl-38402320

ABSTRACT

The increasing prevalence of antibiotic resistance in Cutibacterium acnes (C. acnes) requires the search for alternative therapeutic strategies. Antimicrobial peptides (AMPs) offer a promising avenue for the development of new treatments targeting C. acnes. In this study, to design peptides with the specific inhibitory activity against C. acnes, we employed a deep learning pipeline with generators and classifiers, using transfer learning and pretrained protein embeddings, trained on publicly available data. To enhance the training data specific to C. acnes inhibition, we constructed a phylogenetic tree. A panel of 42 novel generated linear peptides was then synthesized and experimentally evaluated for their antimicrobial selectivity and activity. Five of them demonstrated their high potency and selectivity against C. acnes with MIC of 2-4 µg/mL. Our findings highlight the potential of these designed peptides as promising candidates for anti-acne therapeutics and demonstrate the power of computational approaches for the rational design of targeted antimicrobial peptides.


Subject(s)
Acne Vulgaris , Anti-Infective Agents , Deep Learning , Humans , Antimicrobial Peptides , Phylogeny , Anti-Infective Agents/pharmacology , Acne Vulgaris/microbiology , Propionibacterium acnes , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use
8.
J Drugs Dermatol ; 23(2): SF37896s4-SF378969s10, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38306149

ABSTRACT

Antibiotics, topical and oral, are a cornerstone in the treatment of acnes vulgaris specifically by targeting the skin bacterium Cutibacterium acnes. Billions of individuals have received antibiotics as part of their treatment resulting in a worldwide pandemic of antibiotic resistance not only for C. acnes but also many other pathogens. With the increasing prevalence of acne and exponentially increasing utilization of antibiotics, prescribers must urgently embrace the notion of antibiotic stewardship to maintain the efficacy of acne treatments while attenuating the rise of resistance. This paper serves as an update on C. acnes resistance to antibiotics commonly employed in the treatment of acne and the necessity of implementing benzoyl peroxide in the treatment regimen as monotherapy or combination antibiotic therapies for overcoming and preventing resistance. J Drugs Dermatol. 2024;23:1(Suppl 2):s4-10.


Subject(s)
Acne Vulgaris , Antimicrobial Stewardship , Humans , Drug Resistance, Bacterial , Acne Vulgaris/diagnosis , Acne Vulgaris/drug therapy , Acne Vulgaris/microbiology , Anti-Bacterial Agents , Benzoyl Peroxide/therapeutic use , Propionibacterium acnes
9.
Microbiol Spectr ; 12(2): e0283823, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38197658

ABSTRACT

Acne vulgaris caused by antibiotic-resistant Cutibacterium acnes (C. acnes) infection is difficult to treat conventionally. Phages have been suggested as a potential solution, but research on the mechanism of phage treatment is inadequate. This research investigates the underlying molecular mechanisms of phage φPaP11-13 attenuating C. acnes-induced inflammation in rat models. We found that rats infected with C. acnes had higher average ear thickness, greater enrichment of inflammatory cells as shown by hematoxylin-eosin (HE) staining, and fewer TUNEL (TdT-mediated dUTP Nick-End Labeling)-positive keratinocytes visualized by IF staining. Moreover, an increase of IGF-1 and IGF-1 receptor (IGF-1r) was detected using the immunohistochemical (IHC) staining method, Western blot (WB), and quantitative real-time PCR (qRT-PCR) when infected with C. acnes, which was decreased after the application of phage φPaP11-13. By applying the IGF-1 antibody, it was demonstrated that the severity of C. acnes-induced inflammation was relevant to the expression of IGF-1. Through WB and qRT-PCR, activation of the PI3K/Akt pathway and a down-regulation of the BAD-mediated apoptosis pathway were discovered after C. acnes infection. Subsequently, it was shown that the activation of the PI3K/Akt pathway against BAD-mediated apoptosis pathway was alleviated after applying phage φPaP11-13. Furthermore, applying the IGF-1r inhibitor, Pan-PI3K inhibitor, and Akt inhibitor reversed the changing trends of BAD induced by C. acnes and phage φPaP11-13. This study demonstrates that one of the critical mechanisms underlying the attenuation of acne vulgaris by phage φPaP11-13 is lysing C. acnes and regulating keratinocyte apoptosis via the PI3K/Akt signaling pathway.IMPORTANCECutibacterium acnes infection-induced acne vulgaris may cause severe physical and psychological prognosis. However, the overuse of antibiotics develops drug resistance, bringing challenges in treating Cutibacterium acnes. Bacteriophages are currently proven effective in MDR (multiple drug-resistant) Cutibacterium acnes, but there is a significant lack of understanding of phage therapy. This study demonstrated a novel way of curing acne vulgaris by using phages through promoting cell death of excessive keratinocytes in acne lesions by lysing Cutibacterium acnes. However, the regulation of this cell cycle has not been proven to be directly mediated by phages. The hint of ternary relation among "phage-bacteria-host" inspires huge interest in future phage therapy studies.


Subject(s)
Acne Vulgaris , Bacteriophages , Animals , Rats , Insulin-Like Growth Factor I/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Keratinocytes/metabolism , Keratinocytes/pathology , Acne Vulgaris/microbiology , Propionibacterium acnes/metabolism , Inflammation/metabolism , Apoptosis
10.
J Dermatolog Treat ; 35(1): 2299107, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38164791

ABSTRACT

Objectives:We aimed to explore the potential role of omega-3 (ω-3) fatty acids on acne vulgaris by modulating gut microbiota.Materials and Methods:We randomly divided the untreated acne patients into two groups with or without ω-3 fatty acids intervention for 12 weeks. The Sprague Dawley (SD) rats with acne model were given isotretinoin, ω-3 fatty acids or their combination respectively. Then the colonic contents samples of the drug intervention SD rats were transferred to the pseudo sterile rats with acne model. The severity of the disease was assessed by the Global Acne Grading System (GAGS) score of the patients, and the swelling rate of auricle and the pathological section of the rat with acne model. The 16S rDNA gene sequencing was performed to detect the alteration of the gut microbiota.Results:ω-3 fatty acids could increase the diversity of the gut microbiota and regulate the flora structure positively both in the patients and rats, increase the abundance of butyric acid producing bacteria and GAGS score in the patients, and alleviate the inflammation and comedones of rats.Conclusion:Supplementation of ω-3 fatty acids could alleviate the inflammation of acne vulgaris by increasing the abundance of butyric acid producing bacteria.


Subject(s)
Acne Vulgaris , Fatty Acids, Omega-3 , Gastrointestinal Microbiome , Animals , Humans , Rats , Acne Vulgaris/microbiology , Adjuvants, Immunologic , Butyrates/therapeutic use , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Inflammation/drug therapy , Rats, Sprague-Dawley
11.
Sci Rep ; 14(1): 1521, 2024 01 17.
Article in English | MEDLINE | ID: mdl-38233540

ABSTRACT

Acne vulgaris is a type of chronic skin disorder caused by Propionibacterium acnes (P. acnes). Neutrophil extrinsic traps (NETs) play key role in many types of inflammatory skin diseases. Adipose-derived stem cells (ADSCs) was reported modulate immune responses and neutrophil activity. Here, we explored the potential role of ADSCs and the potential mechanism associated with neutrophil extracellular traps (NETs) in relieving acne vulgaris. In the P. acnes-infected ear skin model, histological staining was used to evaluate the inflammatory infiltration and NET formation in control, P. acnes, and P. acnes + ADSCs groups. Besides, western blot was used to detect the expression levels of cit-H3, MPO, and Nrf2 in ear tissue. In vitro, the immunofluorescence staining of MPO and cit-H3, and SYTOX green staining were performed to measure the NET formation. CCK-8 assay, EdU staining, and wound healing assay were used to detect the proliferation and migration abilities of keratinocytes. ELISA assay was utilized to detect the secretion of inflammatory cytokines. In P. acnes-infected ear skin, ADSC treatment significantly attenuated inflammation and NET formation via activating Nrf2 signaling pathway. In vitro, the conditioned medium of ADSCs reduced the formation of P. acne-induced NETs. Besides, ADSCs could inhibit that the NETs efficiently promoted the proliferation, migration, and inflammatory cytokine secretion of keratinocytes. Our study suggested that ADSCs could attenuate P. acne-related inflammation by inhibiting NET formation. This study provides a novel therapeutic perspective of ADSCs in combating acne vulgaris.


Subject(s)
Acne Vulgaris , Extracellular Traps , Humans , Extracellular Traps/metabolism , NF-E2-Related Factor 2/metabolism , Acne Vulgaris/microbiology , Inflammation , Stem Cells/metabolism , Propionibacterium acnes/metabolism
12.
J Invest Dermatol ; 144(1): 116-124.e4, 2024 01.
Article in English | MEDLINE | ID: mdl-37478901

ABSTRACT

Cutibacterium acnes is a commensal bacterium on the skin that is generally well-tolerated, but different strain types have been hypothesized to contribute to the disease acne vulgaris. To understand how some strain types might contribute to skin inflammation, we generated a repository of C. acnes isolates from skin swabs of healthy subjects and subjects with acne and assessed their strain-level identity and capacity to stimulate cytokine release. Phylotype II K-type strains were more frequent on healthy and nonlesional skin of subjects with acne than those isolated from lesions. Phylotype IA-1 C-type strains were increased on lesional skin compared with those on healthy skin. The capacity to induce cytokines from cultured monocyte-derived dendritic cells was opposite to this action on sebocytes and keratinocytes and did not correlate with the strain types associated with the disease. Whole-genome sequencing revealed a linear plasmid in high-inflammatory isolates within similar strain types that had different proinflammatory responses. Single-cell RNA sequencing of mouse skin after intradermal injection showed that strains containing this plasmid induced a higher inflammatory response in dermal fibroblasts. These findings revealed that C. acnes strain type is insufficient to predict inflammation and that carriage of a plasmid could contribute to disease.


Subject(s)
Acne Vulgaris , Dermatitis , Animals , Mice , Humans , Skin/microbiology , Acne Vulgaris/microbiology , Propionibacterium acnes/genetics , Plasmids/genetics , Inflammation , Cytokines/genetics
14.
Int J Dermatol ; 63(3): 306-310, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37743606

ABSTRACT

Antibiotics have constituted the mainstay of acne therapy despite acne being classified as an inflammatory disorder. The indiscriminate usage of antibiotics over the years has thus fueled the issue of antimicrobial resistance. Cutibacterium acnes (C. acnes) can acquire resistance due to chromosomal mutation or genetic acquisition. C. acnes can transfer resistance to other resident flora, complicating the management of skin and soft tissue infections. It can also transfer resistant strains to other body sites and to immunocompromised and elderly patients thus putting them at risk of serious infections. Recent studies have highlighted the physiologic role of C. acnes in maintaining the normal homeostasis of the skin microbiome. The role of Malassezia in causation of acne has piqued interest in recent times. The efficacy of antibiotics in acne is attributed to their para-antibiotic, anti-inflammatory action rather than antimicrobial action. Thus, usage of low-dose antibiotics and alternatives to antibiotics has been advocated. Some alternative therapies showing efficacy in acne are probiotics, oral zinc, precision therapy using succinic acid, bacteriophages, and anti-biofilm therapy like myrtacin, topical azelaic acid, and salicylic acid. Using isotretinoin in early stages of acne can reduce the incidence of scarring and alleviate the need for antibiotics. Thus, a gradual shift from antibiotics to alternative therapies in acne is the need of the hour.


Subject(s)
Acne Vulgaris , Anti-Bacterial Agents , Humans , Aged , Anti-Bacterial Agents/therapeutic use , Acne Vulgaris/drug therapy , Acne Vulgaris/microbiology , Isotretinoin/therapeutic use , Skin , Salicylic Acid/therapeutic use , Propionibacterium acnes
15.
Sci Rep ; 13(1): 21576, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38062074

ABSTRACT

Emodin, a natural anthraquinone derivative, possesses anti-proliferative and anti-inflammatory properties in skin diseases. However, little information is available on the efficacy of emodin in treating acne vulgaris (acne). This study aims to investigate the protective effects and potential mechanisms of emodin as an anti-acne agent. In vitro, SZ95 sebocytes was chose to establish an acneigenic cellular model. We found that emodin effectively inhibited proliferation, induced cell cycle arrest and apoptosis of SZ95 sebocytes in a dose-dependent manner. To evaluate the lipid-lowering potential of emodin, we examined the levels of lipid contents and lipogenic transcription factors, and found that both lipid production and protein expression of PPARγ, LXR α/ß, and SREBP-1 were decreased after treatment with emodin. Furthermore, our results revealed that emodin inhibited sebaceous lipogenesis induced by insulin-like growth factor 1 (IGF-1), which was accompanied by a potent inhibition of the phosphoinositide-3-kinase (PI3K)/Akt/forkhead box protein O1 (FoxO1) pathway. In detail, emodin augmented the inhibitory effect of isotretinoin and PI3K inhibitor LY294002, while attenuating the activation of IGF-1 on PI3K/Akt/FoxO1 pathway. In addition, emodin could decrease the secretion of pro-inflammatory cytokines IL-6 and IL-8, and suppress the expression of NLRP3, capase-1, IL-1ß, and IL-18 in SZ95 sebocytes exposed to Cutibacterium acnes. Overall, our study provides preliminary evidence supporting the anti-growth, anti-lipogenic and anti-inflammatory properties of emodin, indicating the potential therapeutic application of emodin for acne treatment.


Subject(s)
Acne Vulgaris , Emodin , Humans , Lipogenesis , Sebaceous Glands/metabolism , Insulin-Like Growth Factor I/metabolism , Emodin/pharmacology , Emodin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Phosphatidylinositol 3-Kinases/metabolism , Acne Vulgaris/microbiology , Cell Proliferation , Phosphatidylinositol 3-Kinase/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism , Lipids/pharmacology
16.
Int J Mol Sci ; 24(21)2023 Nov 04.
Article in English | MEDLINE | ID: mdl-37958945

ABSTRACT

The skin is a tightly regulated, balanced interface that maintains our integrity through a complex barrier comprising physical or mechanical, chemical, microbiological, and immunological components. The skin's microbiota affect various properties, one of which is the establishment and maintenance of the physical barrier. This is achieved by influencing multiple processes, including keratinocyte differentiation, stratum corneum formation, and regulation of intercellular contacts. In this review, we summarize the potential contribution of Cutibacterium acnes to these events and outline the contribution of bacterially induced barrier defects to the pathogenesis of acne vulgaris. With the combined effects of a Westernized lifestyle, microbial dysbiosis, epithelial barrier defects, and inflammation, the development of acne is very similar to that of several other multifactorial diseases of barrier organs (e.g., inflammatory bowel disease, celiac disease, asthma, atopic dermatitis, and chronic rhinosinusitis). Therefore, the management of acne requires a complex approach, which should be taken into account when designing novel treatments that address not only the inflammatory and microbial components but also the maintenance and strengthening of the cutaneous physical barrier.


Subject(s)
Acne Vulgaris , Dermatitis, Atopic , Microbiota , Humans , Skin/pathology , Acne Vulgaris/microbiology , Epidermis/pathology , Dermatitis, Atopic/pathology
17.
J Innate Immun ; 15(1): 822-835, 2023.
Article in English | MEDLINE | ID: mdl-37903473

ABSTRACT

INTRODUCTION: CircRNAs are closely related to many human diseases; however, their role in acne remains unclear. This study aimed to determine the role of hsa_circ_0102678 in regulating inflammation of acne. METHODS: First, microarray analysis was performed to study the expression of circRNAs in acne. Subsequently, RNase R digestion assay and fluorescence in situ hybridization assay were utilized to confirm the characteristics of hsa_circ_0102678. Finally, qRT-PCR, Western blotting analysis, immunoprecipitation, luciferase reporter assay, circRNA probe pull-down assay, biotin-labeled miRNA pull-down assay, RNA immunoprecipitation assay, and m6A dot blot assay were utilized to reveal the functional roles of hsa_circ_0102678 on inflammation induced by C. acnes biofilm in human primary keratinocytes. RESULTS: Our investigations showed that the expression of hsa_circ_0102678 was significantly decreased in acne tissues, and hsa_circ_0102678 was a type of circRNAs, which was mainly localized in the cytoplasm of primary human keratinocytes. Moreover, hsa_circ_0102678 remarkably affected the expression of IL-8, IL-6, and TNF-α, which induced by C. acnes biofilm. Importantly, mechanistic studies indicated that the YTHDC1 could bind directly to hsa_circ_0102678 and promote the export of N6-methyladenosine-modified hsa_circ_0102678 to the cytoplasm. Besides, hsa_circ_0102678 could bind to miR-146a and sponge miR-146a to promote the expression of IRAK1 and TRAF6. CONCLUSION: Our findings revealed a previously unknown process by which hsa_circ_0102678 promoted keratinocyte inflammation induced by C. acnes biofilm via regulating miR-146a/TRAF6 and IRAK1 axis.


Subject(s)
Acne Vulgaris , Intracellular Signaling Peptides and Proteins , Nerve Tissue Proteins , Propionibacteriaceae , RNA Splicing Factors , RNA, Circular , Humans , Propionibacteriaceae/physiology , Acne Vulgaris/immunology , Acne Vulgaris/microbiology , Cells, Cultured , Keratinocytes/immunology , Keratinocytes/microbiology , RNA, Circular/genetics , Down-Regulation , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Biological Transport, Active , RNA Splicing Factors/metabolism , Nerve Tissue Proteins/metabolism
18.
Exp Dermatol ; 32(12): 2102-2111, 2023 12.
Article in English | MEDLINE | ID: mdl-37846925

ABSTRACT

Acne is a chronic disease that often persists for years. Skin microbial communities play an essential role in the development of acne. However, limited information is available about the dynamic patterns of skin microbiota in acne. This study aimed to characterize microbial community changes in skin pores and surfaces of acne patients with varying disease time. In this study, a total of 70 skin samples from 22 subjects were collected and sequenced using 16S rRNA amplicon sequencing. Although microbial compositions in skin pores were similar over time, significant differences in microbial structure were observed on the skin surface, with the dominance of Cutibacterium in the first 3 years and replacement by Staphylococcus in 4-6 years. Lactobacillus and Acinetobacter were more abundant in the normal group and continuingly decreased with disease time on the skin surface. Microbial networks further revealed substantial increases in microbial interactions in the 4-6 years group in both skin surfaces and pores. These results demonstrate that the skin microbiota alters with the disease duration and may provide a potential guide in redirecting skin microbiota towards healthy states.


Subject(s)
Acne Vulgaris , Microbiota , Humans , RNA, Ribosomal, 16S/genetics , Cross-Sectional Studies , Acne Vulgaris/microbiology , Skin/microbiology , Microbiota/genetics , Cohort Studies
19.
Sci Rep ; 13(1): 17412, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37833344

ABSTRACT

Prodigiosin, a red pigment produced by Hahella chejuensis, a marine-derived microorganism, has several biological functions, including antimicrobial activity and inflammatory relief. In this study, the antibacterial activity of prodigiosin against skin microorganisms was explored. Paper disc assay on skin bacterial cells revealed that Cutibacterium acnes related to acne vulgaris highly susceptible to prodigiosin. MIC (Minimal Inhibitory Concentration) and MBC (Minimal Bactericidal Concentration) were determined on Cutibacterium species. The RNA-seq analysis of prodigiosin-treated C. acnes cells was performed to understand the antibacterial mechanism of prodigiosin. Among changes in the expression of hundreds of genes, the expression of a stress-responsive sigma factor encoded by sigB increased. Conversely, the gene expression of cell wall biosynthesis and energy metabolism was inhibited by prodigiosin. Specifically, the expression of genes related to the metabolism of porphyrin, a pro-inflammatory metabolite, was significantly reduced. Therefore, prodigiosin could be used to control C. acnes. Our study provided new insights into the antimicrobial mechanism of prodigiosin against C. acnes strains.


Subject(s)
Acne Vulgaris , Prodigiosin , Humans , Prodigiosin/pharmacology , Transcriptome , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Acne Vulgaris/microbiology , Microbial Sensitivity Tests , Propionibacterium acnes/genetics
20.
Nanoscale ; 15(33): 13728-13739, 2023 Aug 25.
Article in English | MEDLINE | ID: mdl-37577823

ABSTRACT

Acne vulgaris is widely regarded as the most prevalent skin disorder characterized by painful, inflammatory skin lesions that are primarily attributed to the pathogenic actions of Cutibacterium acnes (C. acnes). To improve the clinical management of this disease, there is a pressing clinical demand to develop innovative antibacterial therapies that utilize novel mechanisms. The current research aimed to discover the antibacterial efficacy of narasin (NAR), a polyether ionophore, against drug-resistant acne bacteria. In addition, the study aimed to formulate self-nanomicellizing solid dispersions (SNMSD), utilizing Soluplus® (SOL), as a drug delivery system to incorporate NAR and selectively target the lipophilic C. acnes abundant environments within the skin. Furthermore, the study aimed to investigate the ex vivo deposition and permeation of NAR into the various layers of the skin using full-thickness porcine ear skin as a model skin. By encapsulating NAR within spherical polymeric micelles (dn < 80 nm) aqueous solubility was significantly increased by approximately 100-fold (from <40 µg mL-1 to 4600 µg mL-1). Following optimization, the micelle solution was integrated into a gel formulation (containing 0.2% w/v NAR) and evaluated for stability over 4 weeks at room temperature (drug content >98%). Results from drug deposition and permeation experiments demonstrated that the deposition of NAR from the NAR-micelle solution and its gel formulation into the lipophilic stratum corneum (19 835.60 ± 6237.89 ng cm-2 and 40 601.14 ± 3736.09 ng cm-2) and epidermis (19 347 ± 1912.98 ng cm-2 and 18 763.54 ± 580.77 ng cm-2) was superior to that of NAR in solution, which failed to penetrate any skin layers. In conclusion, the outcomes of this study provide evidence that NAR exhibits promising activity against antimicrobial resistant strains of C. acnes (MIC range ≤0.008-0.062) and that micelle nanocarriers can improve the aqueous solubility of poorly water-soluble drugs. Furthermore, our results highlight the ability of nanomicelles to enable selective and targeted drug delivery to the lipophilic skin layers.


Subject(s)
Acne Vulgaris , Micelles , Animals , Swine , Acne Vulgaris/drug therapy , Acne Vulgaris/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Nanotechnology
SELECTION OF CITATIONS
SEARCH DETAIL
...