Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 196
1.
Yakugaku Zasshi ; 144(5): 497-501, 2024.
Article Ja | MEDLINE | ID: mdl-38692923

Signal-transducing adaptor protein-2 (STAP-2) is a unique scaffold protein that regulates several immunological signaling pathways, including LIF/LIF receptor and LPS/TLR4 signals. STAP-2 is required for Fas/FasL-dependent T cell apoptosis and SDF-1α-induced T cell migration. Conversely, STAP-2 modulates integrin-mediated T cell adhesion, suggesting that STAP-2 is essential for several negative and positive T cell functions. However, whether STAP-2 is involved in T cell-antigen receptor (TCR)-mediated T cell activation is unknown. STAP-2 deficiency was recently reported to suppress TCR-mediated T cell activation by inhibiting LCK-mediated CD3ζ and ZAP-70 activation. Using STAP-2 deficient mice, it was demonstrated that STAP-2 is required for the pathogenesis of Propionibacterium acnes-induced granuloma formation and experimental autoimmune encephalomyelitis. Here, detailed functions of STAP-2 in TCR-mediated T cell activation, and how STAP-2 affects the pathogenesis of T cell-mediated inflammation and immune diseases, are reviewed.


Adaptor Proteins, Signal Transducing , Lymphocyte Activation , Receptors, Antigen, T-Cell , Signal Transduction , T-Lymphocytes , ZAP-70 Protein-Tyrosine Kinase , Animals , Receptors, Antigen, T-Cell/physiology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Humans , Adaptor Proteins, Signal Transducing/physiology , Adaptor Proteins, Signal Transducing/metabolism , Mice , ZAP-70 Protein-Tyrosine Kinase/metabolism , ZAP-70 Protein-Tyrosine Kinase/physiology , Propionibacterium acnes/physiology , Propionibacterium acnes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/etiology , Inflammation/immunology , Apoptosis , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Cell Movement , Cell Adhesion , CD3 Complex , Chemokine CXCL12/physiology , Chemokine CXCL12/metabolism
2.
J Eur Acad Dermatol Venereol ; 38(4): 657-664, 2024 Apr.
Article En | MEDLINE | ID: mdl-37777343

Acne vulgaris is a chronic inflammatory skin disease with a complex pathogenesis. Traditionally, the primary pathophysiologic factors in acne have been thought to be: (1) altered sebum production, (2) inflammation, (3) excess keratinization and (4) colonization with the commensal Cutibacterium acnes. However, the role of C. acnes has been unclear, since virtually all adults have C. acnes on their skin yet not all develop acne. In recent years, understanding of the role of C. acnes has expanded. It is still acknowledged to have an important place in acne pathogenesis, but evidence suggests that an imbalance of individual C. acnes phylotypes and an alteration of the skin microbiome trigger acne. In addition, it is now believed that Staphylococcus epidermidis is also an actor in acne development. Together, C. acnes and S. epidermidis maintain and regulate homeostasis of the skin microbiota. Antibiotics, which have long been a staple of acne therapy, induce cutaneous dysbiosis. This finding, together with the long-standing public health edict to spare antibiotic use when possible, highlights the need for a change in acne management strategies. One fertile direction of study for new approaches involves dermocosmetic products that can support epidermal barrier function and have a positive effect on the skin microbiome.


Acne Vulgaris , Dermatitis , Microbiota , Humans , Acne Vulgaris/therapy , Skin/microbiology , Dysbiosis , Anti-Bacterial Agents , Propionibacterium acnes/physiology
3.
Folia Microbiol (Praha) ; 67(6): 825-835, 2022 Dec.
Article En | MEDLINE | ID: mdl-35711021

Acne is a frequently presented dermatological condition brought about by an interplay among inflammation, increased sebum production, hyperkeratinisation, and predominantly Propionibacterium acnes (renamed as Cutibacterium acnes) proliferation, leading to debilitating psychological scars. However, it has been shown that it is the loss of microbial diversity in the skin and the imbalance among C. acnes phylotypes that brings about acne rather than the C. acnes species as a whole. Interestingly, recent evidence suggests that other microorganisms may be implicated, such as the fungi Malassezia and the bacteria Cutibacterium granulosum. A plethora of scientific evidence suggests that the gut microbiome is implicated in the overall health and physiology of the host; studies show that the gut microbiome of acne patients is distinct and depicts less microbial diversity compared to individuals without acne. Herein, using the key terms: acne, C. acnes, IGF-1, sebum, and gut microbiome, we carried out a review of the literature, using Google Scholar and PubMed, and discussed the role of the gut and skin microbiome in relation to acne, as a narrative review. The role of hormones, diet, sebum, and stress in relation to the gut microbiome was also investigated. Therapeutic implications and the use of pre-/postbiotics are also deliberated upon. In this light, future research should investigate the relationship between the gut microbiome and the agreed upon factors of acne pathology, potentially leading to the discovery of novel acne treatments with milder side effects.


Acne Vulgaris , Gastrointestinal Microbiome , Microbiota , Humans , Acne Vulgaris/drug therapy , Acne Vulgaris/microbiology , Propionibacterium acnes/physiology , Skin/microbiology
4.
Bioengineered ; 13(5): 12446-12461, 2022 05.
Article En | MEDLINE | ID: mdl-35587595

The aims of this study were to investigate the outcomes of low- and high-virulence bacterial cervical intervertebral discs (IVDs) infection and its association with cervical IVDs degeneration in rats. A total of 75 clean grade male rats were used to establish the corresponding animal models of low and high virulent bacterial cervical disc infection via an anterior cervical approach, with injection of Propionibacterium acnes (P. acnes) and Staphylococcus epidermidis (S. epidermidis) with a 29 G needle to cervical IVDs. Specimens were collected for evaluation of Blood routine (Blood-RT), histological staining, and gene expression assays after a magnetic resonance imaging (MRI) scan. There were no statistical differences in all groups in white blood cells (WBC) at 2 and 6 weeks postoperatively (P = 0.136). The highest percentage of neutrophils was found in the S. epidermidis group at 2 weeks postoperatively (P = 0.043). MRI and histology showed that at 6 weeks postoperatively, the puncture group and P. acnes group had similar disc degeneration. In the S. epidermidis group, the disc and subchondral bone structure had been destroyed and bony fusion had occurred after the discitis. The upregulation of pro-inflammatory factor expression had the strongest effect of S. epidermidis on the early stage, while the upregulation in the puncture and P. acnes groups was more persistent. P. acnes infection of the cervical IVDs can lead to degenerative changes, whereas S. epidermidis infection leads to the manifestation of septic discitis. The correlation between P. acnes infection and cervical IVDs degeneration found in clinical studies was confirmed.


Discitis , Gram-Positive Bacterial Infections , Intervertebral Disc Degeneration , Intervertebral Disc , Animals , Discitis/complications , Discitis/diagnostic imaging , Discitis/pathology , Gram-Positive Bacterial Infections/complications , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/pathology , Intervertebral Disc/diagnostic imaging , Intervertebral Disc/microbiology , Intervertebral Disc/pathology , Intervertebral Disc Degeneration/diagnostic imaging , Male , Propionibacterium acnes/physiology , Rats , Virulence
5.
Bol Med Hosp Infant Mex ; 79(2): 83-90, 2022.
Article En | MEDLINE | ID: mdl-35468121

Acne is a chronic inflammatory disease of the pilosebaceous unit with multifactorial etiology. Abnormal proliferation of keratinocytes, altered sebum production, inflammation of the sebaceous follicle, and colonization by Cutibacterium acnes have been traditionally implicated. However, the diet has also been highlighted in the pathogenesis because of its direct relation with some biochemical markers and the transcription of specific genes associated with sebaceous gland activity, inflammation, and bacterial proliferation, which together promote the development of the disease, affect the severity of the condition, and modify its response to treatment.


El acné es una enfermedad inflamatoria crónica de la unidad pilosebácea de etiología multifactorial, en la que clásicamente se han implicado la proliferación anormal de queratinocitos, la producción alterada de sebo, la inflamación del folículo sebáceo y la colonización por Cutibacterium acnes. Sin embargo, también destaca la dieta en la patogenia al relacionarse directamente con la alteración de algunos marcadores bioquímicos y transcripción de ciertos genes que se asocian con la actividad de la glándula sebácea, la inflamación y la proliferación bacteriana, que en conjunto promueven el desarrollo de la enfermedad, afectan la gravedad del cuadro y modifican su respuesta al tratamiento.


Acne Vulgaris , Acne Vulgaris/microbiology , Acne Vulgaris/pathology , Diet , Humans , Inflammation/complications , Propionibacterium acnes/physiology , Sebum
6.
Adv Exp Med Biol ; 1367: 137-154, 2022.
Article En | MEDLINE | ID: mdl-35286695

Acne vulgaris results from a complex interaction between environment and genetic factors. While colonization of the pilosebaceous unit with Propionibacterium was previously considered to be the main cause of acne, the contribution of host-related factors that allow the growth of the bacteria and its immune response against bacterial components are now considered to be more important. Many of these host characteristics have a genetic base that is either involved in the regulation of the immune responses or the steroid hormones metabolisms. This chapter aims to explore the functions of these genes and their role in the pathogenesis of acne.


Acne Vulgaris , Propionibacterium acnes , Acne Vulgaris/genetics , Acne Vulgaris/microbiology , Humans , Immunogenetics , Propionibacterium acnes/physiology , Skin/pathology
7.
J Antimicrob Chemother ; 77(4): 1012-1019, 2022 03 31.
Article En | MEDLINE | ID: mdl-35086150

OBJECTIVES: Cutibacterium acnes is one of the common multifactorial causes that play an important role in the pathophysiology of acne vulgaris. We aimed to develop novel antimicrobial peptides for reduction of the hypercolonization. METHODS: Six cationic peptides were derived by de novo designation. The antimicrobial and cytotoxic activities of peptides were investigated. The peptide conformation was determined by circular dichroism spectrometry. The antimicrobial effects of peptides were evaluated using scanning electron microscopy (SEM), transmission electron microscopy (TEM) and DNA-binding ability assay. RESULTS: Among designed peptides, WSKK11 and WSRR11 were effective antimicrobials against C. acnes at MICs of 128 and 64 mg/L, respectively. The MICs of WSKK11 against Staphylococcus epidermidis, Staphylococcus aureus and Candida albicans were 8, 8 and 32 mg/L, while those of WSRR11 were 64, 32 and 32 mg/L, respectively. WSKK11 and WSRR11 were less toxic to human erythrocytes (<2%) and not toxic to macrophages, keratinocytes and fibroblasts up to 512 mg/L. WSKK11 and WSRR11 mostly revealed the conformation of the undefined or random coil structures under mimicked environmental conditions. The peptides affected cell surfaces and cell membranes of C. acnes as well as possibly translocating through the cell membrane, observed by a combination of SEM and TEM, respectively. WSKK11 and WSRR11 had the ability to bind bacterial DNA. CONCLUSIONS: The two novel antimicrobial peptides WSKK11 and WSRR11 are members of a new class of antimicrobial agents that could deal with acne problems. Therefore, the antimicrobial peptides may be promising novel active agents for dermatological, beauty and cosmeceutical applications.


Acne Vulgaris , Antimicrobial Peptides , Acne Vulgaris/drug therapy , Anti-Bacterial Agents/pharmacology , Humans , Microbial Sensitivity Tests , Propionibacterium acnes/physiology
8.
Front Immunol ; 12: 740540, 2021.
Article En | MEDLINE | ID: mdl-34650562

Acne is a multifactorial disease driven by physiological changes occurring during puberty in the pilosebaceous unit (PSU) that leads to sebum overproduction and a dysbiosis involving notably Cutibacterium acnes. These changes in the PSU microenvironment lead to a shift from a homeostatic to an inflammatory state. Indeed, immunohistochemical analyses have revealed that inflammation and lymphocyte infiltration can be detected even in the infraclinical acneic stages, highlighting the importance of the early stages of the disease. In this study, we utilized a robust multi-pronged approach that included flow cytometry, confocal microscopy, and bioinformatics to comprehensively characterize the evolution of the infiltrating and resident immune cell populations in acneic lesions, beginning in the early stages of their development. Using a discovery cohort of 15 patients, we demonstrated that the composition of immune cell infiltrate is highly dynamic in nature, with the relative abundance of different cell types changing significantly as a function of clinical lesion stage. Within the stages examined, we identified a large population of CD69+ CD4+ T cells, several populations of activated antigen presenting cells, and activated mast cells producing IL-17. IL-17+ mast cells were preferentially located in CD4+ T cell rich areas and we showed that activated CD4+ T cells license mast cells to produce IL-17. Our study reveals that mast cells are the main IL-17 producers in the early stage of acne, underlying the importance of targeting the IL-17+ mast cell/T helper cell axis in therapeutic approaches.


Acne Vulgaris/immunology , Gram-Positive Bacterial Infections/immunology , Mast Cells/immunology , Propionibacterium acnes/physiology , Th17 Cells/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Line , Computational Biology , Disease Progression , Flow Cytometry , Humans , Immunologic Memory , Interleukin-17/metabolism , Lectins, C-Type/metabolism , Lymphocyte Activation , Skin/pathology
9.
Int J Mol Sci ; 22(13)2021 Jun 29.
Article En | MEDLINE | ID: mdl-34209998

Acne vulgaris, which is mostly associated with the colonization of Cutibacterium acnes (C. acnes), is a common skin inflammatory disease in teenagers. However, over the past few years, the disease has extended beyond childhood to chronically infect approximately 40% of adults. While antibiotics have been used for several decades to treat acne lesions, antibiotic resistance is a growing crisis; thus, finding a new therapeutic target is urgently needed. Studies have shown that phage therapy may be one alternative for treating multi-drug-resistant bacterial infections. In the present study, we successfully isolated a C. acnes phage named TCUCAP1 from the skin of healthy volunteers. Morphological analysis revealed that TCUCAP1 belongs to the family Siphoviridae with an icosahedral head and a non-contractile tail. Genome analysis found that TCUCAP1 is composed of 29,547 bp with a G+C content of 53.83% and 56 predicted open reading frames (ORFs). The ORFs were associated with phage structure, packing, host lysis, DNA metabolism, and additional functions. Phage treatments applied to mice with multi-drug-resistant (MDR) C.-acnes-induced skin inflammation resulted in a significant decrease in inflammatory lesions. In addition, our attempt to formulate the phage into hydroxyethyl cellulose (HEC) cream may provide new antibacterial preparations for human infections. Our results demonstrate that TCUCAP1 displays several features that make it an ideal candidate for the control of C. acnes infections.


Acne Vulgaris/therapy , Phage Therapy/methods , Propionibacterium acnes/virology , Siphoviridae/classification , Whole Genome Sequencing/methods , Acne Vulgaris/microbiology , Animals , Base Composition , Cellulose/chemistry , Disease Models, Animal , Drug Compounding , Drug Resistance, Multiple, Bacterial , Genome Size , Genome, Viral , Healthy Volunteers , Humans , Injections, Intradermal , Mice , Open Reading Frames , Phylogeny , Propionibacterium acnes/physiology , Siphoviridae/genetics , Siphoviridae/isolation & purification , Skin/virology
10.
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
11.
Int Immunopharmacol ; 96: 107557, 2021 Jul.
Article En | MEDLINE | ID: mdl-33812252

Quercetin is a well-known antioxidant and a plant polyphenolic of flavonoid group found in many fruits, leaves, and vegetables. Propionibacterium acnes is a key skin pathogen involved in the progression of acne inflammation. Although quercetin has been applied to treat various inflammatory diseases, the effects of quercetin on P. acnes-induced skin inflammation have not been explored. This study investigated the effects of quercetin on P. acnes-induced inflammatory skin disease in vitro and in vivo. The results showed that quercetin suppressed the production of pro-inflammatory cytokines in P. acnes-stimulated HaCaT, THP-1 and RAW 264.7 cells. Additionally, quercetin reduced the production of TLR-2 and the phosphorylation of p38, ERK and JNK MAPKs in P. acnes-stimulated HaCaT and THP-1 cells. It also suppressed MMP-9 mRNA levels in two cell lines exposed to P. acnes in vitro. In the case of in vivo, P. acnes was intradermally injected into the ears of mice and it resulted in cutaneous erythema, swelling, and a granulomatous response. Treatment with quercetin markedly reduced ear thickness and swelling. These results suggested that quercetin can be a potential therapeutic agent against P. acnes-induced skin inflammation and may have diverse pharmaceutical and cosmetics applications.


Anti-Inflammatory Agents/therapeutic use , Gram-Positive Bacterial Infections/drug therapy , Inflammation/drug therapy , Keratinocytes/physiology , Propionibacterium acnes/physiology , Quercetin/therapeutic use , Skin/immunology , Animals , Gene Expression Regulation , Humans , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , RAW 264.7 Cells , Signal Transduction , THP-1 Cells
12.
Eur J Dermatol ; 31(1): 32-40, 2021 Feb 01.
Article En | MEDLINE | ID: mdl-33648912

BACKGROUND: Acne vulgaris is a common pilosebaceous disease associated with Propionibacterium acnes (P. acnes). Resolution of comedones may occur in association with shrunken sebaceous glands (SGs) containing de-differentiated cells, however the role of P. acnes is unclear. OBJECTIVES: To investigate the effects of P. acnes on aryl hydrocarbon receptor (AhR) activation, lipogenesis and differentiation in cultured immortalized human SZ95 sebocytes. MATERIALS & METHODS: Cultured sebocytes were incubated with formalin-killed (f-) P. acnes (f-P. acnes) at different ratios of multiplicity of infection. The mRNA levels of the AhR downstream cytochrome P450 (CYP) genes were measured by quantitative RT-PCR, nuclear translocation of AhR by western blot and immunofluorescence, lipogenesis and keratinization by gene set enrichment analysis (GSEA), lipid related analysis by Oil red O staining and Nile red staining, and sebaceous differentiation-related gene expression by western blot. RESULTS: f-P. acnes upregulated CYPs mRNA levels and induced translocation of AhR protein from the cytoplasm into the nucleus. GSEA revealed downregulation of lipogenesis and upregulation of keratinization. f-P. acnes inhibited linoleic acid-induced neutral lipid synthesis and expression of sebocyte markers, keratin 7 and mucin1/EMA, but increased expression of keratinocyte markers, keratin 10 and involucrin, which were abolished by AhR gene silencing. Inhibition of lipogenesis-related genes, such as sterol response element-binding protein, was also observed. CONCLUSION: f-P. acnes inhibits lipogenesis and induces terminal differentiation of sebocytes, into keratinocyte-like cells, via activation of the AhR pathway in vitro, suggesting that follicular P. acnes is not only acnegenic but also promotes acne remission through feedback regulation of sebum production.


Cell Differentiation/physiology , Formaldehyde/pharmacology , Propionibacterium acnes/drug effects , Propionibacterium acnes/physiology , Receptors, Aryl Hydrocarbon/physiology , Sebaceous Glands/cytology , Cells, Cultured , Humans
13.
Int J Mol Sci ; 22(5)2021 Feb 26.
Article En | MEDLINE | ID: mdl-33652921

Previously, we proposed the hypothesis that similarities in the inflammatory response observed in acne vulgaris and degenerative disc disease (DDD), especially the central role of interleukin (IL)-1ß, may be further evidence of the role of the anaerobic bacterium Cutibacterium (previously Propionibacterium) acnes in the underlying aetiology of disc degeneration. To investigate this, we examined the upregulation of IL-1ß, and other known IL-1ß-induced inflammatory markers and neurotrophic factors, from nucleus-pulposus-derived disc cells infected in vitro with C. acnes for up to 48 h. Upon infection, significant upregulation of IL-1ß, alongside IL-6, IL-8, chemokine (C-C motif) ligand 3 (CCL3), chemokine (C-C motif) ligand 4 (CCL4), nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), was observed with cells isolated from the degenerative discs of eight patients versus non-infected controls. Expression levels did, however, depend on gene target, multiplicity and period of infection and, notably, donor response. Pre-treatment of cells with clindamycin prior to infection significantly reduced the production of pro-inflammatory mediators. This study confirms that C. acnes can stimulate the expression of IL-1ß and other host molecules previously associated with pathological changes in disc tissue, including neo-innervation. While still controversial, the role of C. acnes in DDD remains biologically credible, and its ability to cause disease likely reflects a combination of factors, particularly individualised response to infection.


Inflammation/microbiology , Intervertebral Disc Degeneration/microbiology , Nerve Growth Factors/genetics , Propionibacterium acnes/physiology , Adult , Cells, Cultured , Female , Host-Pathogen Interactions , Humans , Inflammation/genetics , Interleukin-1beta/genetics , Intervertebral Disc/metabolism , Intervertebral Disc/microbiology , Intervertebral Disc Degeneration/genetics , Male , Middle Aged , Up-Regulation
14.
FEMS Microbiol Lett ; 368(4)2021 03 19.
Article En | MEDLINE | ID: mdl-33580963

Staphylococcus aureus and Cutibacterium acnes are involved in several tissue infections and can encounter mesenchymal stem cells (MSCs) during their role in tissue regenerative process. C. acnes and S. aureus internalization by three types of MSCs derived from bone marrow, dental pulp and Wharton's jelly; and bacterial biofilm production were compared. Internalization rates ranged between 1.7-6.3% and 0.8-2.7% for C. acnes and S. aureus, respectively. While C. acnes strains exhibited limited cytotoxic effect on MSCs, S. aureus were more virulent with marked effect starting after only 3 h of interaction. Both bacteria were able to produce biofilms with respectively aggregated and monolayered structures for C. acnes and S. aureus. The increase in C. acnes capacity to develop biofilm following MSCs' internalization was not linked to the significant increase in number of live bacteria, except for bone marrow-MSCs/C. acnes CIP 53.117 with 79% live bacteria compared to the 36% before internalization. On the other hand, internalization of S. aureus had no impact on its ability to form biofilms composed mainly of living bacteria. The present study underlined the complexity of MSCs-bacteria cross-interaction and brought insights into understanding the MSCs behavior in response to bacterial infection in tissue regeneration context.


Mesenchymal Stem Cells/microbiology , Propionibacterium acnes/physiology , Staphylococcus aureus/physiology , Biofilms/growth & development , Cell Survival , Cytoplasm/microbiology , Host-Pathogen Interactions , Humans , Prosthesis-Related Infections/microbiology
15.
Oxid Med Cell Longev ; 2021: 4657014, 2021.
Article En | MEDLINE | ID: mdl-33603947

Our previous study verified the occurrence of Propionibacterium acnes (P. acnes), a low-virulence anaerobic bacterium, latently residing in degenerated intervertebral discs (IVDs), and the infection had a strong association with IVD degeneration. We explored whether P. acnes induces nucleus pulposus cell (NPC) pyroptosis, a more dangerous cell death process than apoptosis, and accelerates IVD degeneration via the pyroptotic products interleukin- (IL-) 1ß and IL-18. After coculturing with P. acnes, human NPCs showed significant upregulation of NOD-like receptor 3 (NLRP3), cleaved IL-1ß, cleaved caspase-1, and cleaved gasdermin D in response to the overexpression of IL-1ß and IL-18 in a time- and dose-dependent manner. In addition, the gene expression of inflammatory factors and catabolic enzymes significantly increased in normal NPCs when cocultured with pyroptotic NPCs in a transwell system, and the adverse effects were inhibited when NPC pyroptosis was suppressed. Furthermore, inoculation of P. acnes into the IVDs of rats caused significant pyroptosis of NPCs and remarkable IVD degeneration. Finally, coculture of NPCs with P. acnes induced the overexpression of reactive oxygen species (ROS) and NLRP3, while inhibition of both factors reduced NPC pyroptosis. Therefore, P. acnes induces NPC pyroptosis via the ROS-NLRP3 signaling pathway, and the pyroptotic NPCs cause an IVD degeneration cascade. Targeting the P. acnes-induced pyroptosis of NPCs may become an alternative treatment strategy for IVD degeneration in the future.


Intervertebral Disc Degeneration/microbiology , Intervertebral Disc Degeneration/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nucleus Pulposus/pathology , Propionibacterium acnes/physiology , Pyroptosis , Reactive Oxygen Species/metabolism , Signal Transduction , Adult , Animals , Female , Humans , Inflammation/pathology , Male , Nucleus Pulposus/metabolism , Rats, Sprague-Dawley
16.
Infect Immun ; 89(4)2021 03 17.
Article En | MEDLINE | ID: mdl-33468579

Cutibacterium acnes role is well described during acne but remains a mystery regarding its implication in bone and prosthesis or cerebrospinal fluid shunt infections. The main issue is that these low-grade symptom infections are difficult to diagnose and lead to irreversible and grave sequelae for patients. Consequently, there is an urgent need to find new biomarkers to accelerate the diagnosis of disease, an issue addressed by Beaver et al. thanks to a promising proteomic approach.


Biomarkers , Gram-Positive Bacterial Infections/diagnosis , Gram-Positive Bacterial Infections/microbiology , Propionibacterium acnes/physiology , Acne Vulgaris/diagnosis , Acne Vulgaris/microbiology , Animals , Biofilms/growth & development , Humans , Opportunistic Infections/diagnosis , Opportunistic Infections/microbiology
17.
J Dermatolog Treat ; 32(3): 277-285, 2021 May.
Article En | MEDLINE | ID: mdl-31393195

Acne vulgaris, a common and chronic disorder of the pilosebaceous unit, affects up to 85% of adolescent and young adults. While a lot is already known about acne and its treatment, still the gaps in our understanding of acne remains. This article will review the emerging evidence in the complex pathogenesis of acne and provide an overview of the potential future therapy in management of acne vulgaris.Key pointsWhat is known? Propionibacterium acnes targeted therapy has been the mainstay in the management of acne till now.What is new? Sebocyte activity is controlled via a range of cellular pathways and hormones in addition to androgens. This has opened an array of therapeutic options to be available for treating acne in the near future.


Acne Vulgaris/pathology , Acne Vulgaris/drug therapy , Acne Vulgaris/microbiology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antioxidants/chemistry , Antioxidants/therapeutic use , Biofilms/drug effects , Cortodoxone/therapeutic use , Diet , Enzyme Inhibitors/therapeutic use , Humans , Propionibacterium acnes/isolation & purification , Propionibacterium acnes/physiology
18.
J Ethnopharmacol ; 268: 113552, 2021 Mar 25.
Article En | MEDLINE | ID: mdl-33152431

ETHNOPHARMACOLOGICAL RELEVANCE: Sapindus mukorossi Gaertn. (S. mukorossi), known as 'mu huan zi' in Chinese folklore, belongs to the family Sapindaceae and it has been traditionally used for treating coughing and excessive salivation, removing freckle, whitening skin, etc. Evidence-based medicine also verified the antimicrobial, anti-tyrosinase and anti-acne activity of S. mukorossi extract, suggesting that it has the potential to be a pharmaceutical and cosmetic additive. AIM OF THE STUDY: The present study was intended to evaluate the freckle-removing and skin-whitening activities of S. mukorossi extracts, and further analyzing the potential anti-acne mechanism. METHODS: Saponin fractions were purified by using the semi-preparative high-performance liquid chromatography, and their antibacterial activity was detected against Propionibacterium acnes (P. acnes), which was the leading cause of inflamed lesions in acne vulgaris. The anti-lipase and anti-tyrosinase activities were assayed using a commercial kit, while the potential anti-acne mechanism was predicted on the basis of the network pharmacology. Active components of saponin fraction were identified by HPLC-MS analysis. Furthermore, the different toxicity level of compounds was predicted according to the quantitative structure-activity relationship, and the first application of crude extract and saponin fraction to facial masks was analyzed based on the comprehensive evaluation method. RESULTS: The saponin fraction (F4) purified from the fermentation liquid-based water extract (SWF) showed the best antibacterial activity against P. acnes ATCC 6919 with the MIC of 0.06 mg/mL, which was 33-fold of its parent SWF (with the MIC of 2.0 mg/mL). Compared with SWF, the application of F4 caused greater inhibition rates on lipase and tyrosinase. Chemical constituents of F4 were evaluated, from which four oleanane-type triterpenoid saponins were detected to contribute to the above biological activities of F4. The mechanism of the four compounds on anti-acne was predicted, and seven targets such as PTGS2 and F2RL1 were obtained to be important for the treatment of acne. The four compounds were also predicted to have different levels of toxicity to various species, and they were not harmful to rats. Besides, F4 and SWF were applied to facial masks and there was no significant influence on the physicochemical properties including pH, stability, and sensory characteristics. CONCLUSION: This work demonstrated that oleanane-type triterpenoid saponins were speculated to contribute to the skin-whitening, freckle-removing, and anti-acne activities of F4. These findings will facilitate the development of the S. mukorossi extract and the allied products as the new and natural anti-acne agent and cosmetic additives.


Acne Vulgaris/drug therapy , Cosmetics/administration & dosage , Plant Extracts/administration & dosage , Propionibacterium acnes/drug effects , Sapindus , Saponins/administration & dosage , Acne Vulgaris/diagnosis , Acne Vulgaris/microbiology , Adult , Cosmetics/isolation & purification , Cosmetics/toxicity , Drug Evaluation, Preclinical/methods , Female , Forecasting , Humans , Male , Microbial Sensitivity Tests/methods , Plant Extracts/isolation & purification , Plant Extracts/toxicity , Propionibacterium acnes/physiology , Saponins/isolation & purification , Saponins/toxicity , Young Adult
19.
Int J Mol Sci ; 21(21)2020 Oct 24.
Article En | MEDLINE | ID: mdl-33114460

Understanding the importance of oral microbiota in human health and disease also leads to an expansion of the knowledge on functional, metabolic, and molecular alterations directly contributing to oral and systemic pathologies. To date, a compelling number of studies have documented the crucial role of some oral cavity-occurring microbes in the initiation and progression of cancers. Although this effect was noted primarily for Fusobacterium spp., the potential impact of other oral microbes is also worthy of investigation. In this study, we aimed to assess the effect of Enterococcus faecalis, Actinomyces odontolyticus, and Propionibacterium acnes on the proliferation capability and mechanical features of gingival cells and cell lines derived from lung, breast, and ovarian cancers. For this purpose, we incubated selected cell lines with heat-inactivated bacteria and supernatants collected from biofilms, cultured in both anaerobic and aerobic conditions, in the presence of surgically removed teeth and human saliva. The effect of oral bacteria on cell population growth is variable, with the highest growth-promoting abilities observed for E. faecalis in relation to human primary gingival fibroblasts (HGF) and lung cancer A549 cells, and P. acnes in relation to breast cancer MCF-7 and ovarian cancer SKOV-3 cells. Notably, this effect seems to depend on a delicate balance between the pro-stimulatory and toxic effects of bacterial-derived products. Regardless of the diverse effect of bacterial products on cellular proliferation capability, we observed significant alterations in stiffness of gingival and lung cancer cells stimulated with E. faecalis bacteria and corresponding biofilm supernatants, suggesting a novel molecular mechanism involved in the pathogenesis of diseases in oral cavities and tooth tissues. Accordingly, it is proposed that analysis of cancerogenic features of oral cavity bacteria should be multivariable and should include investigation of potential alterations in cell mechanical properties. These findings corroborate the important role of oral hygiene and root canal treatment to assure the healthy stage of oral microbiota.


Actinomyces/physiology , Dental Pulp Cavity/microbiology , Enterococcus faecalis/physiology , Gingiva/cytology , Neoplasms/microbiology , Propionibacterium acnes/physiology , A549 Cells , Biofilms , Biomechanical Phenomena , Cell Line , Cell Line, Tumor , Cell Proliferation , Gingiva/microbiology , Hot Temperature , Humans , Ki-67 Antigen/metabolism , MCF-7 Cells , Neoplasms/metabolism
20.
Exp Dermatol ; 29(9): 840-859, 2020 09.
Article En | MEDLINE | ID: mdl-32779248

Current experimental research on acne pathophysiology has revealed a more complicated background than the classically reported four-factor aetiology. Cells of the pilosebaceous unit, which represent the template for the development of acne lesions, seem to be parallelly affected by endocrinological/metabolic factors as well as inflammatory/immunological ones that cooperate in sebocyte differentiation and lipogenesis. Indeed, the unique programme of sebocyte terminal differentiation and death, the so called holocrine secretion, is influenced by inflammatory and metabolic (lipid) signalling with common denominator the selective regulation of peroxisome proliferator-activated receptors. Autophagy provides substrates for energy generation and biosynthesis of new cell structure proteins contributing to the normally increased sebaceous gland metabolic functions, which are also regulated by extracellular calcium signalling, essential lipids and hormones. The ultimate differentiation product of human sebocytes, sebum, co-regulates the inflammatory sebocyte status. Sebum composition is controlled among others by Propionibacterium acnes and other bacteria, sexual hormones, neuropeptides, endogenous opioids and environmental agents, which may function as endocrine disruptors. Diet may also be an important source of substrates for the synthesis of pro-inflammatory and anti-inflammatory sebaceous lipids. Sebum changes might induce inflammation and initiate underlying immune mechanisms leading to acne lesions. Current new therapeutic efforts on acne concentrate on anti-inflammatory/immunologically active concepts, which are able to regulate sebaceous lipogenesis. At last, current molecular studies based on published molecular data sets confirmed the major role of inflammation in acne development.


Acne Vulgaris/immunology , Hormones/physiology , Sebaceous Glands/physiology , Acne Vulgaris/drug therapy , Calcium Signaling , Cell Differentiation , Host-Pathogen Interactions , Humans , Lipogenesis , Propionibacterium acnes/physiology , Sebaceous Glands/cytology , Sebum/physiology
...