Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Publication year range
1.
J Allergy Clin Immunol ; 147(3): 955-966.e16, 2021 03.
Article in English | MEDLINE | ID: mdl-32634452

ABSTRACT

BACKGROUND: Staphylococcus aureus and Staphylococcus epidermidis are the most abundant bacteria found on the skin of patients with atopic dermatitis (AD). S aureus is known to exacerbate AD, whereas S epidermidis has been considered a beneficial commensal organism. OBJECTIVE: In this study, we hypothesized that S epidermidis could promote skin damage in AD by the production of a protease that damages the epidermal barrier. METHODS: The protease activity of S epidermidis isolates was compared with that of other staphylococcal species. The capacity of S epidermidis to degrade the barrier and induce inflammation was examined by using human keratinocyte tissue culture and mouse models. Skin swabs from atopic and healthy adult subjects were analyzed for the presence of S epidermidis genomic DNA and mRNA. RESULTS: S epidermidis strains were observed to produce strong cysteine protease activity when grown at high density. The enzyme responsible for this activity was identified as EcpA, a cysteine protease under quorum sensing control. EcpA was shown to degrade desmoglein-1 and LL-37 in vitro, disrupt the physical barrier, and induce skin inflammation in mice. The abundance of S epidermidis and expression of ecpA mRNA were increased on the skin of some patients with AD, and this correlated with disease severity. Another commensal skin bacterial species, Staphylococcus hominis, can inhibit EcpA production by S epidermidis. CONCLUSION: S epidermidis has commonly been regarded as a beneficial skin microbe, whereas S aureus has been considered deleterious. This study suggests that the overabundance of S epidermidis found on some atopic patients can act similarly to S aureus and damage the skin by expression of a cysteine protease.


Subject(s)
Bacterial Proteins/metabolism , Cysteine Proteases/metabolism , Dermatitis, Atopic/microbiology , Microbiota , Skin/microbiology , Staphylococcal Skin Infections/microbiology , Staphylococcus epidermidis/enzymology , Animals , Antimicrobial Cationic Peptides/metabolism , Cells, Cultured , DNA, Bacterial/genetics , Dermatitis, Atopic/pathology , Desmoglein 1/metabolism , Humans , Keratinocytes/microbiology , Keratinocytes/pathology , Mice , Mice, Inbred C57BL , Severity of Illness Index , Skin/pathology , Staphylococcal Skin Infections/pathology , Cathelicidins
2.
Proc Natl Acad Sci U S A ; 112(17): E2120-9, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25825778

ABSTRACT

The human skin is an organ with a surface area of 1.5-2 m(2) that provides our interface with the environment. The molecular composition of this organ is derived from host cells, microbiota, and external molecules. The chemical makeup of the skin surface is largely undefined. Here we advance the technologies needed to explore the topographical distribution of skin molecules, using 3D mapping of mass spectrometry data and microbial 16S rRNA amplicon sequences. Our 3D maps reveal that the molecular composition of skin has diverse distributions and that the composition is defined not only by skin cells and microbes but also by our daily routines, including the application of hygiene products. The technological development of these maps lays a foundation for studying the spatial relationships of human skin with hygiene, the microbiota, and environment, with potential for developing predictive models of skin phenotypes tailored to individual health.


Subject(s)
Imaging, Three-Dimensional , Microbiota/physiology , Models, Biological , RNA, Bacterial , RNA, Ribosomal, 16S , Skin/microbiology , Adult , Female , Humans , Male , Mass Spectrometry , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism
3.
Exp Dermatol ; 25(1): 26-31, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26307945

ABSTRACT

Ixodes hard tick induces skin injury by its sophisticated biting process. Its saliva plays a key role to enable an efficient blood meal that lasts for several days. We hypothesized that this feeding process may also be exploited by pathogens to facilitate their transmission, especially in the context of arthropod-borne diseases. To test this, we used Lyme borreliosis as a model. This bacterial infection is caused by Borrelia burgdorferi sensu lato transmitted by Ixodes. We co-incubated Borrelia with human keratinocytes in the presence of poly (I: C), a dsRNA TLR3 agonist generated by skin injury. This induced a strong cytokine response from human primary keratinocytes that was much greater than that induced by Borrelia alone. OspC, a TLR2/1 agonist and a major surface lipoprotein of Borrelia also amplified the process. Interestingly, tick saliva inhibited cytokine responses by keratinocytes to these TLR agonists. We propose that Borrelia uses the immunoprivileged site produced by tick saliva to facilitate its transmission.


Subject(s)
Borrelia burgdorferi , Ixodes , Saliva/microbiology , Skin Diseases/microbiology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 3/metabolism , Animals , Antigens, Bacterial/metabolism , Apoptosis , Bacterial Outer Membrane Proteins/metabolism , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Humans , Inflammation , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/microbiology , Ligands , Lyme Disease/immunology , Mice , Mice, Inbred C57BL , Poly I-C/chemistry , RNA, Double-Stranded/chemistry , Skin/immunology , Skin/injuries , Skin/microbiology , Skin Diseases/immunology
6.
J Invest Dermatol ; 144(1): 84-95.e3, 2024 01.
Article in English | MEDLINE | ID: mdl-37544587

ABSTRACT

Absorption of dietary iron is largely regulated by the liver hormone hepcidin, which is released under conditions of iron overload and inflammation. Although hepcidin-dependent regulation of iron uptake and circulation is well-characterized, recent studies have suggested that the skin may play an important role in iron homeostasis, including transferrin receptor-mediated epidermal iron uptake and direct hepcidin production by keratinocytes. In this study, we characterized direct keratinocyte responses to conditions of high and low iron. We observed potent iron storage capacity by keratinocytes in vitro and in vivo and the effects of iron on epidermal differentiation and gene expression associated with inflammation and barrier function. In mice, systemic iron was observed to be coupled to epidermal iron content. Furthermore, topical inflammation, as opposed to systemic inflammation, resulted in a primary iron-deficiency phenotype associated with low liver hepcidin. These studies suggest a role for keratinocytes and epidermal iron storage as regulators of iron homeostasis with direct contribution by the cutaneous inflammatory state.


Subject(s)
Ferritins , Hepcidins , Animals , Mice , Ferritins/metabolism , Hepcidins/genetics , Hepcidins/metabolism , Iron/metabolism , Homeostasis , Inflammation
7.
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
8.
J Exp Med ; 221(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38393304

ABSTRACT

The skin provides an essential barrier for host defense through rapid action of multiple resident and recruited cell types, but the complex communication network governing these processes is incompletely understood. To define these cell-cell interactions more clearly, we performed an unbiased network analysis of mouse skin during invasive S. aureus infection and revealed a dominant role for CXCL12+ fibroblast subsets in neutrophil communication. These subsets predominantly reside in the reticular dermis, express adipocyte lineage markers, detect IL-17 and TNFα, and promote robust neutrophil recruitment through NFKBIZ-dependent release of CXCR2 ligands and CXCL12. Targeted deletion of Il17ra in mouse fibroblasts resulted in greatly reduced neutrophil recruitment and increased infection by S. aureus. Analogous human CXCL12+ fibroblast subsets abundantly express neutrophil chemotactic factors in psoriatic skin that are subsequently decreased upon therapeutic targeting of IL-17. These findings show that CXCL12+ dermal immune acting fibroblast subsets play a critical role in cutaneous neutrophil recruitment and host defense.


Subject(s)
Interleukin-17 , Staphylococcus aureus , Mice , Animals , Humans , Neutrophil Infiltration , Skin , Fibroblasts , Chemokine CXCL12
9.
Cell Rep ; 42(5): 112494, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37167061

ABSTRACT

During inflammation, the skin deploys antimicrobial peptides (AMPs) yet during allergic inflammation it becomes more susceptible to Staphylococcus aureus. To understand this contradiction, single-cell sequencing of Il4ra-/- mice combined with skin microbiome analysis reveals that lower production of AMPs from interleukin-4 receptor α (IL-4Rα) activation selectively inhibits survival of antibiotic-producing strains of coagulase-negative Staphylococcus (CoNS). Diminished AMPs under conditions of T helper type 2 (Th2) inflammation enable expansion of CoNS strains without antibiotic activity and increase Staphylococcus aureus (S. aureus), recapitulating the microbiome on humans with atopic dermatitis. This response is rescued in Camp-/- mice or after topical steroids, since further inhibition of AMPs enables survival of antibiotic-producing CoNS strains. In conditions of Th17 inflammation, a higher expression of host AMPs is sufficient to directly inhibit S. aureus survival. These results show that antimicrobials produced by the host and commensal bacteria each act to control S. aureus on the skin.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Humans , Animals , Mice , Staphylococcus aureus/metabolism , Antimicrobial Peptides , Skin/microbiology , Inflammation , Bacteria , Staphylococcus , Anti-Bacterial Agents/metabolism
10.
Curr Opin Immunol ; 72: 324-330, 2021 10.
Article in English | MEDLINE | ID: mdl-34537476

ABSTRACT

The skin represents the largest area for direct contact between microbes and host immunocytes and is a site for constant communication between the host and this diverse and essential microbial community. Coagulase-negative staphylococci are an abundant bacterial genus on the human skin and are regulated through various mechanisms that include the epidermal barrier environment and innate and adaptive immune systems within the epidermis and dermis. In turn, some species and strains of these bacteria produce beneficial products that augment host immunity by exerting specifically targeted antimicrobial, anti-inflammatory, or anti-neoplastic activity while also promoting broad innate and adaptive immune responses. The use of selected skin commensals as a therapeutic has shown promise in recent human clinical trials. This emerging concept of bacteriotherapy is defining mechanisms of action and validating the dependence on the microbiome for maintenance of immune homeostasis.


Subject(s)
Host Microbial Interactions/immunology , Microbiota/immunology , Skin Physiological Phenomena , Skin/immunology , Skin/microbiology , Animals , Biomarkers , Disease Management , Disease Susceptibility , Dysbiosis , Humans , Immunomodulation , Microbial Interactions/immunology
11.
J Invest Dermatol ; 141(5): 1188-1197.e5, 2021 05.
Article in English | MEDLINE | ID: mdl-33197483

ABSTRACT

The outer layer of the epidermis composes the skin barrier, a sophisticated filter constituted by layers of corneocytes in a lipid matrix. The matrix lipids, especially the ceramide-generated sphingosine 1-phosphate, are the messengers that the skin barrier uses to communicate with the basal layer of the epidermis where replicating keratinocytes are located. Sphingosine 1-phosphate is a bioactive sphingolipid mediator involved in various cellular functions through S1PR1‒5, expressed by keratinocytes. We discovered that the S1pr2 absence is linked to an impairment in the skin barrier function. Although S1pr2-/- mouse skin has no difference in its phenotype and barrier function compared with that of wild-type mouse, after tape stripping, S1pr2-/- mouse showed significantly higher transepidermal water loss and required another 24 hours to normalize their transepidermal water loss levels. Moreover, after epicutaneous Staphylococcus aureus application, impaired S1pr2-/- mouse epidermal barrier function allowed deeper bacterial penetration and denser neutrophil infiltration in the dermis. Microarray and RNA sequence of S1pr2-/- mouse epidermis linked the barrier dysfunction with a decrease in FLG2 and tight junction components. In conclusion, S1pr2-/- mice have compromised skin barrier function and increased bacteria permeability, making them a suitable model for diseases that present similar characteristics, such as atopic dermatitis.


Subject(s)
Epidermis/metabolism , Homeostasis/physiology , Sphingosine-1-Phosphate Receptors/physiology , Animals , Cells, Cultured , Filaggrin Proteins , Humans , Lysophospholipids/pharmacology , Mice , Mice, Inbred BALB C , Permeability , S100 Proteins/analysis , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Stress, Mechanical
12.
JAMA Dermatol ; 2021 Jun 16.
Article in English | MEDLINE | ID: mdl-34132739

ABSTRACT

IMPORTANCE: Atopic dermatitis (AD) can be negatively affected by Staphylococcus aureus. The skin microbiome of AD is deficient in coagulase-negative Staphylococcus (CoNS) that can kill S aureus. OBJECTIVE: To evaluate if the antimicrobial-producing CoNS (CoNS-AM+) of a patient with AD can be autologously reintroduced to the same patient to inhibit survival of S aureus and improve clinical outcomes. DESIGN, SETTING, AND PARTICIPANTS: This double-blind, vehicle-controlled, single-center randomized clinical trial of 11 adult patients with moderate to severe AD who were randomized to receive either an autologous CoNS-AM+ (n = 5) or the vehicle (n = 6) was conducted between April 2016 and May 2018. The data were analyzed from May 2018 to July 2019. INTERVENTIONS: Autologous CoNS-AM+ was isolated from swabs that were obtained from the nonlesional skin of each patient with AD, expanded by culture, and then reapplied topically to the forearms at a concentration of 107 colony-forming units/g. MAIN OUTCOMES AND MEASURES: The primary end point of this study was to assess S aureus abundance after 1 week of application of autologous CoNS-AM+ on patients with AD by culture-based and DNA-based methods. The secondary end points were to assess the safety and clinical outcomes. RESULTS: Eleven patients (4 men [36.4%] and 7 women [63/6%]) were recruited based on the inclusion criteria. There were no serious adverse events in groups treated with autologous CoNS-AM+ or the vehicle. Staphylococcus aureus colonization on lesional skin at the end of treatment on patients who were treated with autologous CoNS-AM+ (mean of log10 ratio to baseline, -1.702; 95% CI, -2.882 to -0.523) was reduced by 99.2% compared with vehicle treatment (mean of log10 ratio to baseline, 0.671; 95% CI, -0.289 to 1.613; P = .01) and persisted for 4 days after treatment (CoNS-AM+: mean of log10 ratio to baseline, -1.752; 95% CI, -3.051 to -0.453; vehicle: mean of log10 ratio to baseline, -0.003; 95% CI, -1.083 to 1.076; P = .03). Importantly, local Eczema Area And Severity Index scores that were assessed at day 11 on patients who received CoNS-AM+ (mean of percentage change, -48.45; 95% CI, -84.34 to -12.55) were significantly improved compared with vehicle treatment (mean of percentage change, -4.52; 95% CI, -36.25 to 27.22; P = .04). CONCLUSIONS AND RELEVANCE: The data from this randomized clinical trial suggest that bacteriotherapy with an autologous strain of skin commensal bacteria can safely decrease S aureus colonization and improve disease severity. Although larger studies will be needed, this personalized approach for S aureus reduction may provide an alternative treatment for patients with AD beyond antibiotics, immunosuppression, and immunomodulation. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT03158012.

13.
Sci Immunol ; 6(59)2021 05 21.
Article in English | MEDLINE | ID: mdl-34021025

ABSTRACT

The skin typically tolerates exposure to various microbes and chemicals in the environment. Here, we investigated how the epidermis maintains this innate immune tolerance to stimuli that are recognized by Toll-like receptors (TLRs). Loss of tolerance to TLR ligands occurred after silencing of the histone deacetylases (HDACs) HDAC8 and HDAC9 in keratinocytes. Transcriptional analysis identified MAP2K3 as suppressed by HDAC8/9 activity and a potential key intermediary for establishing this tolerance. HDAC8/9 influenced acetylation at H3K9 and H3K27 marks in the MAP2K3 promoter. Proteomic analysis further identified SSRP1 and SUPT16H as associated with HDAC8/9 and responsible for transcriptional elongation of MAP2K3. Silencing of MAP2K3 blocked the capacity of HDAC8/9 to influence cytokine responses. Relevance in vivo was supported by observations of increased MAP2K3 in human inflammatory skin conditions and the capacity of keratinocyte HDAC8/9 to influence dendritic cell maturation and T cell proliferation. Keratinocyte-specific deletion of HDAC8/9 also increased inflammation in mice after exposure to ultraviolet radiation, imiquimod, or Staphylococcus aureus These findings define a mechanism for the epidermis to regulate inflammation in the presence of ubiquitous TLR ligands.


Subject(s)
Histone Deacetylases/immunology , MAP Kinase Kinase 3/immunology , Repressor Proteins/immunology , Skin/immunology , Animals , Cells, Cultured , Dendritic Cells/immunology , Epigenesis, Genetic , Histone Deacetylases/genetics , Humans , Imiquimod/pharmacology , Immune Tolerance , Immunity, Innate , Keratinocytes/immunology , MAP Kinase Kinase 3/genetics , Mice, Transgenic , Repressor Proteins/genetics , Staphylococcus aureus , T-Lymphocytes/immunology , Toll-Like Receptors/immunology , Ultraviolet Rays
14.
Elife ; 102021 10 19.
Article in English | MEDLINE | ID: mdl-34664551

ABSTRACT

Methicillin-resistant Staphylococcus pseudintermedius (MRSP) is an important emerging zoonotic pathogen that causes severe skin infections. To combat infections from drug-resistant bacteria, the transplantation of commensal antimicrobial bacteria as a therapeutic has shown clinical promise. We screened a collection of diverse staphylococcus species from domestic dogs and cats for antimicrobial activity against MRSP. A unique strain (S. felis C4) was isolated from feline skin that inhibited MRSP and multiple gram-positive pathogens. Whole genome sequencing and mass spectrometry revealed several secreted antimicrobials including a thiopeptide bacteriocin micrococcin P1 and phenol-soluble modulin beta (PSMß) peptides that exhibited antimicrobial and anti-inflammatory activity. Fluorescence and electron microscopy revealed that S. felis antimicrobials inhibited translation and disrupted bacterial but not eukaryotic cell membranes. Competition experiments in mice showed that S. felis significantly reduced MRSP skin colonization and an antimicrobial extract from S. felis significantly reduced necrotic skin injury from MRSP infection. These findings indicate a feline commensal bacterium that could be utilized in bacteriotherapy against difficult-to-treat animal and human skin infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/pharmacology , Bacteriocins/pharmacology , Drug Resistance, Bacterial , Staphylococcal Infections/veterinary , Staphylococcus/chemistry , Staphylococcus/drug effects , Animals , Anti-Bacterial Agents/chemistry , Anti-Infective Agents/chemistry , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Bacteriocins/chemistry , Cats/microbiology , Mass Spectrometry , Pore Forming Cytotoxic Proteins/chemistry , Pore Forming Cytotoxic Proteins/pharmacology , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Whole Genome Sequencing
15.
Nat Med ; 27(4): 700-709, 2021 04.
Article in English | MEDLINE | ID: mdl-33619370

ABSTRACT

Staphylococcus aureus colonizes patients with atopic dermatitis (AD) and exacerbates disease by promoting inflammation. The present study investigated the safety and mechanisms of action of Staphylococcus hominis A9 (ShA9), a bacterium isolated from healthy human skin, as a topical therapy for AD. ShA9 killed S. aureus on the skin of mice and inhibited expression of a toxin from S. aureus (psmα) that promotes inflammation. A first-in-human, phase 1, double-blinded, randomized 1-week trial of topical ShA9 or vehicle on the forearm skin of 54 adults with S. aureus-positive AD (NCT03151148) met its primary endpoint of safety, and participants receiving ShA9 had fewer adverse events associated with AD. Eczema severity was not significantly different when evaluated in all participants treated with ShA9 but a significant decrease in S. aureus and increased ShA9 DNA were seen and met secondary endpoints. Some S. aureus strains on participants were not directly killed by ShA9, but expression of mRNA for psmα was inhibited in all strains. Improvement in local eczema severity was suggested by post-hoc analysis of participants with S. aureus directly killed by ShA9. These observations demonstrate the safety and potential benefits of bacteriotherapy for AD.


Subject(s)
Dermatitis, Atopic/microbiology , Dermatitis, Atopic/therapy , Skin/microbiology , Staphylococcus hominis/physiology , Administration, Topical , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Bacterial Proteins/metabolism , Bacteriocins/pharmacology , Colony Count, Microbial , Humans , Inflammation/complications , Inflammation/pathology , Mice, Inbred BALB C , Microbial Sensitivity Tests , Microbial Viability/drug effects , Middle Aged , Peptides, Cyclic/metabolism , Reproducibility of Results , Skin/drug effects , Skin/pathology , Staphylococcal Infections/microbiology , Staphylococcal Infections/therapy , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus aureus/physiology , Transcription, Genetic/drug effects , Treatment Outcome , Virulence Factors/metabolism , Young Adult
16.
Extremophiles ; 14(2): 171-83, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20091073

ABSTRACT

The analysis of the cold-shock domain (CSD)-encoding genes, capB and cspA, by PCR amplification showed presence of capB in all 18 Antarctic Pseudomonas isolates, but the absence of cspA. Nucleotide sequence analysis of capB ORF from a biodegradative Pseudomonas 30/3 and its regulatory sequences including the promoter and 5'-UTR was determined and compared with the other CSD-encoding genes. Expression analysis using translational gene fusion of the putative capB promoter and its flanking sequence from Pseudomonas sp. 30/3 with lacZ' exhibited a significant increase in beta-galactosidase activity at 15 and 6 degrees C. Unlike the expression of E. coli CspA, Pseudomonas sp. 30/3 showed a slow but steady increase of the CapB expression at 6 degrees C. Subcellular localization of CapB at 6 degrees C showed accumulation in and around the nucleoid whereas at 22 or 30 degrees C, it was identified around the nucleoid as well as in the cytosol. Our study attempts to elucidate the detailed structure of capB from Pseudomonas 30/3 and the role of 5'UTR in the transcriptional regulation along with the possible role of CapB in transcription and translation suited for the cold adaptation of this bacterium in Antarctic environment.


Subject(s)
Genes, Bacterial , Pseudomonas/genetics , 5' Untranslated Regions , Acclimatization , Amino Acid Sequence , Antarctic Regions , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Carrier Proteins/genetics , Cold Climate , DNA Primers/genetics , DNA, Bacterial/genetics , Gene Expression Regulation, Bacterial , Heat-Shock Proteins/genetics , Immunohistochemistry , Molecular Sequence Data , Open Reading Frames , Polymerase Chain Reaction , Promoter Regions, Genetic , Protein Structure, Tertiary , Pseudomonas/isolation & purification , Pseudomonas/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
17.
Sci Rep ; 10(1): 21237, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33277548

ABSTRACT

Biofilm formation by bacterial pathogens is associated with numerous human diseases and can confer resistance to both antibiotics and host defenses. Many strains of Staphylococcus epidermidis are capable of forming biofilms and are important human pathogens. Since S. epidermidis coexists with abundant Cutibacteria acnes on healthy human skin and does not typically form a biofilm in this environment, we hypothesized that C. acnes may influence biofilm formation of S. epidermidis. Culture supernatants from C. acnes and other species of Cutibacteria inhibited S. epidermidis but did not inhibit biofilms by Pseudomonas aeruginosa or Bacillus subtilis, and inhibited biofilms by S. aureus to a lesser extent. Biofilm inhibitory activity exhibited chemical properties of short chain fatty acids known to be produced from C. acnes. The addition of the pure short chain fatty acids propionic, isobutyric or isovaleric acid to S. epidermidis inhibited biofilm formation and, similarly to C. acnes supernatant, reduced polysaccharide synthesis by S. epidermidis. Both short chain fatty acids and C. acnes culture supernatant also increased sensitivity of S. epidermidis to antibiotic killing under biofilm-forming conditions. These observations suggest the presence of C. acnes in a diverse microbial community with S. epidermidis can be beneficial to the host and demonstrates that short chain fatty acids may be useful to limit formation of a biofilm by S. epidermidis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Fatty Acids, Volatile/pharmacology , Propionibacteriaceae/metabolism , Staphylococcus epidermidis/drug effects , Bacillus subtilis/drug effects , Bacillus subtilis/physiology , Culture Media, Conditioned/analysis , Culture Media, Conditioned/pharmacology , Drug Synergism , Hemiterpenes/pharmacology , Isobutyrates/pharmacology , Pentanoic Acids/pharmacology , Polysaccharides/biosynthesis , Propionates/pharmacology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/metabolism , Staphylococcus epidermidis/physiology
18.
J Invest Dermatol ; 140(8): 1619-1628.e2, 2020 08.
Article in English | MEDLINE | ID: mdl-31981578

ABSTRACT

The microbiome represents a vast resource for drug discovery, as its members engage in constant conflict to outcompete one another by deploying diverse strategies for survival. Cutibacterium acnes is one of the most common bacterial species on human skin and can promote the common disease acne vulgaris. By employing a combined strategy of functional screening, genetics, and proteomics we discovered a strain of Staphylococcus capitis (S. capitis E12) that selectively inhibited growth of C. acnes with potency greater than antibiotics commonly used in the treatment of acne. Antimicrobial peptides secreted from S. capitis E12 were identified as four distinct phenol-soluble modulins acting synergistically. These peptides were not toxic to human keratinocytes and the S. capitis extract did not kill other commensal skin bacteria but was effective against C. acnes on pig skin and on mice. Overall, these data show how a member of the human skin microbiome can be useful as a biotherapy for acne vulgaris.


Subject(s)
Acne Vulgaris/therapy , Biological Therapy/methods , Skin/microbiology , Staphylococcus capitis/immunology , Symbiosis/immunology , Acne Vulgaris/immunology , Acne Vulgaris/microbiology , Adult , Animals , Female , Humans , Keratinocytes/immunology , Male , Mice , Microbial Sensitivity Tests , Pore Forming Cytotoxic Proteins/isolation & purification , Pore Forming Cytotoxic Proteins/metabolism , Pore Forming Cytotoxic Proteins/toxicity , Primary Cell Culture , Propionibacterium acnes/immunology , Propionibacterium acnes/pathogenicity , Skin/immunology , Staphylococcus capitis/isolation & purification , Staphylococcus capitis/metabolism , Swine , Toxicity Tests , Young Adult
19.
Cell Rep ; 30(1): 61-68.e4, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31914398

ABSTRACT

Staphylococcus aureus is a major human bacterial pathogen responsible for deep tissue skin infections. Recent observations have suggested that rapid, localized digestion of hyaluronic acid in the extracellular matrix (ECM) of the dermis may influence bacterial invasion and tissue inflammation. In this study we find that cell migration-inducing protein (Cemip) is the major inducible gene responsible for hyaluronan catabolism in mice. Cemip-/- mice failed to digest hyaluronan and had significantly less evidence of infection after intradermal bacterial challenge by S. aureus. Stabilization of large-molecular-weight hyaluronan enabled increased expression of cathelicidin antimicrobial peptide (Camp) that was due in part to enhanced differentiation of preadipocytes to adipocytes, as seen histologically and by increased expression of Pref1, PPARg, and Adipoq. Cemip-/- mice challenged with S. aureus also had greater IL-6 expression and neutrophil infiltration. These observations describe a mechanism for hyaluronan in the dermal ECM to regulate tissue inflammation and host antimicrobial defense.


Subject(s)
Host-Pathogen Interactions , Hyaluronic Acid/metabolism , Hyaluronoglucosaminidase/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Adipogenesis , Animals , Dermis/microbiology , Dermis/pathology , Hyaluronoglucosaminidase/deficiency , Inflammation/pathology , Mice, Inbred C57BL , Mice, Knockout
20.
J Invest Dermatol ; 140(1): 191-202.e7, 2020 01.
Article in English | MEDLINE | ID: mdl-31252032

ABSTRACT

Dupilumab is a fully human antibody to interleukin-4 receptor α that improves the signs and symptoms of moderate to severe atopic dermatitis (AD). To determine the effects of dupilumab on Staphylococcus aureus colonization and microbial diversity on the skin, bacterial DNA was analyzed from swabs collected from lesional and nonlesional skin in a double-blind, placebo-controlled study of 54 patients with moderate to severe AD randomized (1:1) and treated with either dupilumab (200 mg weekly) or placebo for 16 weeks. Microbial diversity and relative abundance of Staphylococcus were assessed by DNA sequencing of 16S ribosomal RNA, and absolute S. aureus abundance was measured by quantitative PCR. Before treatment, lesional skin had lower microbial diversity and higher overall abundance of S. aureus than nonlesional skin. During dupilumab treatment, microbial diversity increased and the abundance of S. aureus decreased. Pronounced changes were seen in nonlesional and lesional skin. Decreased S. aureus abundance during dupilumab treatment correlated with clinical improvement of AD and biomarkers of type 2 immunity. We conclude that clinical improvement of AD that is mediated by interleukin-4 receptor α inhibition and the subsequent suppression of type 2 inflammation is correlated with increased microbial diversity and reduced abundance of S. aureus.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Dermatitis, Atopic/drug therapy , Immunotherapy/methods , Skin/microbiology , Staphylococcal Infections/drug therapy , Staphylococcus aureus/physiology , Th2 Cells/immunology , Cytokines/metabolism , Disease Progression , Double-Blind Method , Female , Follow-Up Studies , Humans , Male , Placebos , RNA, Ribosomal, 16S/genetics , Receptors, Interleukin-4/antagonists & inhibitors , Skin/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL