Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 544
Filter
1.
Elife ; 132024 May 20.
Article in English | MEDLINE | ID: mdl-38767331

ABSTRACT

Wound infections are highly prevalent and can lead to delayed or failed healing, causing significant morbidity and adverse economic impacts. These infections occur in various contexts, including diabetic foot ulcers, burns, and surgical sites. Enterococcus faecalis is often found in persistent non-healing wounds, but its contribution to chronic wounds remains understudied. To address this, we employed single-cell RNA sequencing (scRNA-seq) on infected wounds in comparison to uninfected wounds in a mouse model. Examining over 23,000 cells, we created a comprehensive single-cell atlas that captures the cellular and transcriptomic landscape of these wounds. Our analysis revealed unique transcriptional and metabolic alterations in infected wounds, elucidating the distinct molecular changes associated with bacterial infection compared to the normal wound healing process. We identified dysregulated keratinocyte and fibroblast transcriptomes in response to infection, jointly contributing to an anti-inflammatory environment. Notably, E. faecalis infection prompted a premature, incomplete epithelial-mesenchymal transition in keratinocytes. Additionally, E. faecalis infection modulated M2-like macrophage polarization by inhibiting pro-inflammatory resolution in vitro, in vivo, and in our scRNA-seq atlas. Furthermore, we discovered macrophage crosstalk with neutrophils, which regulates chemokine signaling pathways, while promoting anti-inflammatory interactions with endothelial cells. Overall, our findings offer new insights into the immunosuppressive role of E. faecalis in wound infections.


If wounds get infected, they heal much more slowly, sometimes leading to skin damage and other complications, including disseminated infections or even amputation. Infections can happen in many types of wounds, ranging from ulcers in patients with diabetes to severe burns. If infections are not cleared quickly, the wounds can become 'chronic' and are unable to heal without intervention. Enterococcus faecalis is a type of bacteria that normally lives in the gut. Within that environment, in healthy people, it is not harmful. However, if it comes into contact with wounds ­ particularly diabetic ulcers or the site of a surgery ­ it can cause persistent infections and prevent healing. Although researchers are beginning to understand how E. faecalis initially colonises wounds, the biological mechanisms that transform these infections into chronic wounds are still largely unknown. Celik et al. therefore set out to investigate exactly how E. faecalis interferes with wound healing. To do this, Celik et al. looked at E. faecalis-infected wounds in mice and compared them to uninfected ones. Using a genetic technique called single-cell RNA sequencing, Celik et al. were able to determine which genes were switched on in individual skin and immune cells at the site of the wounds. This in turn allowed the researchers to determine how those cells were behaving in both infected and uninfected conditions. The experiments revealed that when E. faecalis was present in wounds, several important cell types in the wounds did not behave normally. For example, although the infected skin cells still underwent a change in behaviour required for healing (called an epithelial-mesenchymal transition), the change was both premature and incomplete. In other words, the skin cells in infected wounds started changing too early and did not finish the healing process properly. E. faecalis also changed the way macrophages and neutrophils worked within the wounds. These are cells in our immune system that normally promote inflammation, a process involved in both uninfected wounds or during infections and is a key part of wound healing when properly controlled. In the E. faecalis-infected wounds, these cells' inflammatory properties were suppressed, making them less helpful for healing. These results shed new light on how E. faecalis interacts with skin cells and the immune system to disrupt wound healing. Celik et al. hope that this knowledge will allow us to find new ways to target E. faecalis infections, and ultimately develop treatments to help chronic wounds heal better and faster.


Subject(s)
Enterococcus faecalis , Gram-Positive Bacterial Infections , Keratinocytes , Wound Healing , Enterococcus faecalis/physiology , Enterococcus faecalis/genetics , Animals , Mice , Gram-Positive Bacterial Infections/microbiology , Keratinocytes/microbiology , Keratinocytes/metabolism , Macrophages/microbiology , Macrophages/metabolism , Macrophages/immunology , Disease Models, Animal , Wound Infection/microbiology , Transcriptome , Mice, Inbred C57BL , Single-Cell Analysis , Epithelial-Mesenchymal Transition/genetics , Male , Fibroblasts/microbiology , Fibroblasts/metabolism
2.
Mycopathologia ; 189(3): 37, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38704808

ABSTRACT

Trichophyton rubrum is a human fungal pathogen that causes dermatophytosis, an infection that affects keratinized tissues. Integrated molecular signals coordinate mechanisms that control pathogenicity. Transcriptional regulation is a core regulation of relevant fungal processes. Previous RNA sequencing data revealed that the absence of the transcription factor StuA resulted in the differential expression of the MAPK-related high glycerol osmolarity gene (hog1) in T. rubrum. Here we validated the role of StuA in regulating the transcript levels of hog1. We showed through RT-qPCR that transcriptional regulation controls hog1 levels in response to glucose, keratin, and co-culture with human keratinocytes. In addition, we also detected hog1 pre-mRNA transcripts that underwent alternative splicing, presenting intron retention in a StuA-dependent mechanism. Our findings suggest that StuA and alternative splicing simultaneously, but not dependently, coordinate hog1 transcript levels in T. rubrum. As a means of preventing and treating dermatophytosis, our results contribute to the search for new potential drug therapies based on the molecular aspects of signaling pathways in T. rubrum.


Subject(s)
Alternative Splicing , Arthrodermataceae , Gene Expression Regulation, Fungal , Mitogen-Activated Protein Kinases , Tinea , Transcription Factors , Humans , Arthrodermataceae/genetics , Arthrodermataceae/metabolism , Glucose/metabolism , Keratinocytes/microbiology , Keratins/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Tinea/metabolism , Tinea/microbiology
3.
Microbiol Spectr ; 12(6): e0034724, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38700333

ABSTRACT

We have evaluated the inhibitory effects of supernatants and lysates derived from several candidate probiotics, on the growth and biofilm formation of wound pathogens, and their ability to protect human primary epidermal keratinocytes from the toxic effects of pathogens. Supernatants (neutralized and non-neutralized) and lysates (via sonication) from Lactiplantibacillus plantarum, Limosilactobacillus reuteri, Bifidobacterium longum, Lacticaseibacillus rhamnosus GG, and Escherichia coli Nissle 1917 were tested for their inhibitory effects against Staphylococcus aureus, Streptococcus pyogenes, Escherichia coli, Pseudomonas aeruginosa, and Acinetobacter baumanni. The supernatants of L. plantarum, L. rhamnosus, B. longum, and L. rhamnosus GG reduced the growth of S. aureus, E. coli, and A. baumanni. B. longum additionally inhibited P. aeruginosa growth. However, neutralized Lactobacillus supernatants did not inhibit growth and in some cases were stimulatory. Lysates of L. plantarum and L. reuteri inhibited S. pyogenes while B. longum lysates inhibited E. coli and S. aureus growth. E. coli Nissle 1917 lysates enhanced the growth of S. pyogenes and P. aeruginosa. Biofilm formation by E. coli was reduced by lysates of L. reuteri and neutralized supernatants of all candidate probiotics. P. aeruginosa biofilm formation was reduced by E. coli Nissle supernatant but increased by L. plantarum, L. reuteri, and Bifidobacterium longum lysates. L. reuteri decreased the toxic effects of S. aureus on keratinocytes while E. coli Nissle 1917 lysates protected keratinocytes from S. pyogenes toxicity. In conclusion, lactobacilli and E. coli Nissle lysates confer inhibitory effects on pathogenic growth independently of acidification and may beneficially alter the outcome of interactions between host cell-pathogen in a species-specific manner.IMPORTANCEOne of the attributes of probiotics is their ability to inhibit pathogens. For this reason, many lactobacilli have been investigated for their effects as potential topical therapeutics against skin pathogens. However, this field is in its infancy. Even though probiotics are known to be safe when taken orally, the potential safety concerns when applied to potentially compromised skin are unknown. For this reason, we believe that extracts of probiotics will offer advantages over the use of live bacteria. In this study, we have surveyed five candidate probiotics, when used as extracts, in terms of their effects against common wound pathogens. Our data demonstrate that some probiotic extracts promote the growth of pathogens and highlight the need for careful selection of species and strains when probiotics are to be used topically.


Subject(s)
Biofilms , Escherichia coli , Keratinocytes , Probiotics , Pseudomonas aeruginosa , Staphylococcus aureus , Humans , Keratinocytes/microbiology , Escherichia coli/drug effects , Escherichia coli/growth & development , Biofilms/drug effects , Biofilms/growth & development , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/physiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Streptococcus pyogenes/drug effects , Streptococcus pyogenes/growth & development , Streptococcus pyogenes/physiology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/physiology , Acinetobacter baumannii/growth & development , Wound Infection/microbiology
4.
Infect Immun ; 92(6): e0008324, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38712951

ABSTRACT

Streptococcus pyogenes [group A streptococcus (GAS)] is a human pathogen capable of infecting diverse tissues. To successfully infect these sites, GAS must detect available nutrients and adapt accordingly. The phosphoenolpyruvate transferase system (PTS) mediates carbohydrate uptake and metabolic gene regulation to adapt to the nutritional environment. Regulation by the PTS can occur through phosphorylation of transcriptional regulators at conserved PTS-regulatory domains (PRDs). GAS has several PRD-containing stand-alone regulators with regulons encoding both metabolic genes and virulence factors [PRD-containing virulence regulators (PCVRs)]. One is RofA, which regulates the expression of virulence genes in multiple GAS serotypes. It was hypothesized that RofA is phosphorylated by the PTS in response to carbohydrate levels to coordinate virulence gene expression. In this study, the RofA regulon of M1T1 strain 5448 was determined using RNA sequencing. Two operons were consistently differentially expressed across growth in the absence of RofA; the pilus operon was downregulated, and the capsule operon was upregulated. This correlated with increased capsule production and decreased adherence to keratinocytes. Purified RofA-His was phosphorylated in vitro by PTS proteins EI and HPr, and phosphorylated RofA-FLAG was detected in vivo when GAS was grown in low-glucose C medium. Phosphorylated RofA was not observed when C medium was supplemented 10-fold with glucose. Mutations of select histidine residues within the putative PRDs contributed to the in vivo phosphorylation of RofA, although phosphorylation of RofA was still observed, suggesting other phosphorylation sites exist in the protein. Together, these findings support the hypothesis that RofA is a PCVR that may couple sugar metabolism with virulence regulation.


Subject(s)
Bacterial Proteins , Gene Expression Regulation, Bacterial , Streptococcus pyogenes , Virulence Factors , Streptococcus pyogenes/pathogenicity , Streptococcus pyogenes/genetics , Streptococcus pyogenes/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Virulence Factors/genetics , Virulence Factors/metabolism , Virulence , Phosphorylation , Humans , Regulon , Operon , Streptococcal Infections/microbiology , Phosphoenolpyruvate Sugar Phosphotransferase System/metabolism , Phosphoenolpyruvate Sugar Phosphotransferase System/genetics , Keratinocytes/microbiology
5.
mSystems ; 9(5): e0017924, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38656122

ABSTRACT

The utilization of ATP within cells plays a fundamental role in cellular processes that are essential for the regulation of host-pathogen dynamics and the subsequent immune response. This study focuses on ATP-binding proteins to dissect the complex interplay between Staphylococcus aureus and human cells, particularly macrophages (THP-1) and keratinocytes (HaCaT), during an intracellular infection. A snapshot of the various protein activity and function is provided using a desthiobiotin-ATP probe, which targets ATP-interacting proteins. In S. aureus, we observe enrichment in pathways required for nutrient acquisition, biosynthesis and metabolism of amino acids, and energy metabolism when located inside human cells. Additionally, the direct profiling of the protein activity revealed specific adaptations of S. aureus to the keratinocytes and macrophages. Mapping the differentially activated proteins to biochemical pathways in the human cells with intracellular bacteria revealed cell-type-specific adaptations to bacterial challenges where THP-1 cells prioritized immune defenses, autophagic cell death, and inflammation. In contrast, HaCaT cells emphasized barrier integrity and immune activation. We also observe bacterial modulation of host processes and metabolic shifts. These findings offer valuable insights into the dynamics of S. aureus-host cell interactions, shedding light on modulating host immune responses to S. aureus, which could involve developing immunomodulatory therapies. IMPORTANCE: This study uses a chemoproteomic approach to target active ATP-interacting proteins and examines the dynamic proteomic interactions between Staphylococcus aureus and human cell lines THP-1 and HaCaT. It uncovers the distinct responses of macrophages and keratinocytes during bacterial infection. S. aureus demonstrated a tailored response to the intracellular environment of each cell type and adaptation during exposure to professional and non-professional phagocytes. It also highlights strategies employed by S. aureus to persist within host cells. This study offers significant insights into the human cell response to S. aureus infection, illuminating the complex proteomic shifts that underlie the defense mechanisms of macrophages and keratinocytes. Notably, the study underscores the nuanced interplay between the host's metabolic reprogramming and immune strategy, suggesting potential therapeutic targets for enhancing host defense and inhibiting bacterial survival. The findings enhance our understanding of host-pathogen interactions and can inform the development of targeted therapies against S. aureus infections.


Subject(s)
Adenosine Triphosphate , Host-Pathogen Interactions , Keratinocytes , Macrophages , Staphylococcus aureus , Humans , Staphylococcus aureus/metabolism , Adenosine Triphosphate/metabolism , Host-Pathogen Interactions/immunology , Macrophages/microbiology , Macrophages/metabolism , Macrophages/immunology , Keratinocytes/microbiology , Keratinocytes/metabolism , Keratinocytes/immunology , THP-1 Cells , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Proteomics/methods , Bacterial Proteins/metabolism , HaCaT Cells
6.
Cell Rep ; 43(4): 114022, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38568806

ABSTRACT

Staphylococcus aureus causes the majority of skin and soft tissue infections, but this pathogen only transiently colonizes healthy skin. However, this transient skin exposure enables S. aureus to transition to infection. The initial adhesion of S. aureus to skin corneocytes is mediated by surface protein G (SasG). Here, phylogenetic analyses reveal the presence of two major divergent SasG alleles in S. aureus: SasG-I and SasG-II. Structural analyses of SasG-II identify a nonaromatic arginine in the binding pocket of the lectin subdomain that mediates adhesion to corneocytes. Atomic force microscopy and corneocyte adhesion assays indicate that SasG-II can bind to a broader variety of ligands than SasG-I. Glycosidase treatment results in different binding profiles between SasG-I and SasG-II on skin cells. In addition, SasG-mediated adhesion is recapitulated using differentiated N/TERT keratinocytes. Our findings indicate that SasG-II has evolved to adhere to multiple ligands, conferring a distinct advantage to S. aureus during skin colonization.


Subject(s)
Bacterial Adhesion , Keratinocytes , Skin , Staphylococcus aureus , Staphylococcus aureus/metabolism , Humans , Skin/microbiology , Skin/metabolism , Keratinocytes/microbiology , Keratinocytes/metabolism , Lectins/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Phylogeny , Protein Binding
7.
Exp Dermatol ; 33(5): e15087, 2024 May.
Article in English | MEDLINE | ID: mdl-38685821

ABSTRACT

Hidradenitis Suppurativa (HS) is a chronic autoinflammatory skin disease with activated keratinocytes, tunnel formation and a complex immune infiltrate in tissue. The HS microbiome is polymicrobial with an abundance of commensal gram-positive facultative (GPs) Staphylococcus species and gram-negative anaerobic (GNA) bacteria like Prevotella, Fusobacterium and Porphyromonas with increasing predominance of GNAs with disease severity. We sought to define the keratinocyte response to bacteria commonly isolated from HS lesions to probe pathogenic relationships between HS and the microbiome. Type strains of Prevotella nigrescens, Prevotella melaninogenica, Prevotella intermedia, Prevotella asaccharolytica, Fusobacterium nucleatum, as well as Staphylococcus aureus and the normal skin commensal Staphylococcus epidermidis were heat-killed and co-incubated with normal human keratinocytes. RNA was collected and analysed using RNAseq and RT-qPCR. The supernatant was collected from cell culture for protein quantification. Transcriptomic profiles between HS clinical samples and stimulated keratinocytes were compared. Co-staining of patient HS frozen sections was used to localize bacteria in lesions. A mouse intradermal injection model was used to investigate early immune recruitment. TLR4 and JAK inhibitors were used to investigate mechanistic avenues of bacterial response inhibition. GNAs, especially F. nucleatum, stimulated vastly higher CXCL8, IL17C, CCL20, IL6, TNF and IL36γ transcription in normal skin keratinocytes than the GPs S. epidermidis and S. aureus. Using RNAseq, we found that F. nucleatum (and Prevotella) strongly induced the IL-17 pathway in keratinocytes and overlapped with transcriptome profiles of HS patient clinical samples. Bacteria were juxtaposed to activated keratinocytes in vivo, and F. nucleatum strongly recruited murine neutrophil and macrophage migration. Both the TLR4 and pan-JAK inhibitors reduced cytokine production. Detailed transcriptomic profiling of healthy skin keratinocytes exposed to GNAs prevalent in HS revealed a potent, extensive inflammatory response vastly stronger than GPs. GNAs stimulated HS-relevant genes, including many genes in the IL-17 response pathway, and were significantly associated with HS tissue transcriptomes. The close association of activated keratinocytes with bacteria in HS lesions and innate infiltration in murine skin cemented GNA pathogenic potential. These novel mechanistic insights could drive future targeted therapies.


Subject(s)
Hidradenitis Suppurativa , Keratinocytes , Keratinocytes/immunology , Keratinocytes/microbiology , Keratinocytes/metabolism , Humans , Animals , Mice , Hidradenitis Suppurativa/microbiology , Hidradenitis Suppurativa/immunology , Staphylococcus aureus/immunology , Staphylococcus epidermidis/immunology , Fusobacterium nucleatum/immunology , Transcriptome , Cytokines/metabolism , Bacteria, Anaerobic , Interleukin-17/metabolism , Microbiota , Prevotella/immunology
8.
Int J Cosmet Sci ; 46(3): 414-423, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38229273

ABSTRACT

OBJECTIVE: This study aimed to expound on the correlation between facial skin microbiome and sensitive skin (SS) using a novel sequencing technique. METHODS: We applied the 2bRAD sequencing for the microbiome, which enables accurate characterization of the low-biomass microbiome at species resolution to profile facial skin microbes in SS and non-SS groups. Further, the bacterial colonies were isolated and cultured from skin surfaces to study the pro-inflammatory effect on human keratinocytes by ELISA. RESULTS: We accordingly identified 1142 genera and 4436 strains. In the SS group, the proportions of Actinomyces and Microbotryomycetes were significantly increased, whereas that of Acidimicrobiia was decreased. Kruskal-Wallis analysis revealed significant differences in 11 genera and 35 species, among which the proportions of Dermabacter, Chryseobacterium, Rhodotorula and Peptoniphilus A were increased in the SS group. Analysis of the top 10 genera revealed increased proportions of Cutibacterium, Corynebacterium and Staphylococcus. Moreover, the proportion of Dermabacter hominis was significantly increased by 18.9-fold in the SS group, whereas those of many Streptococcus strains were significantly decreased. Focus on the isolated bacterial colonies from skin surfaces, more yellow colonies were found in SS group when cultured in Tryptic Soy Broth medium for 48 h, and more interleukin-8 was detected on keratinocytes after yellow colonies stimulation, such as S.capitis, M.luteus. CONCLUSIONS: This study suggests that more SS-associated microorganisms can be identified using the 2bRAD technique even with a small sample size. Dermabacter hominis and Chryseobacterium was firstly reported with a significantly increase in SS, and the S.capitis, as well as M.luteus, but not S.aureus, may be associated with skin inflammation.


OBJECTIF: Cette étude visait à expliquer la corrélation entre le microbiome de la peau du visage et la peau sensible (PS) à l'aide d'une nouvelle technique de séquençage. MÉTHODES: Nous avons appliqué le séquençage 2bRAD pour le microbiome, ce qui nous a permis de caractériser précisément le microbiome à faible biomasse à la résolution des espèces pour profiler les microbes de la peau du visage dans les groupes PS et non­PS. En outre, les colonies bactériennes ont été isolées et cultivées à partir de surfaces cutanées pour étudier l'effet pro­inflammatoire sur les kératinocytes humains par ELISA. RÉSULTATS: Nous avons donc identifié 1 142 genres et 4 436 souches. Dans le groupe PS, on a pu constater des proportions d'Actinomyces et de microbotryomycètes significativement accrues, pour de moindres proportions d'Acidimicrobiia. L'analyse de Kruskal­Wallis a révélé des différences significatives dans 11 genres et 35 espèces, parmi lesquelles des proportions de Dermabacter, Chryseobacterium, Rhodotorula et Peptoniphilus A accrues dans le groupe PS. L'analyse des 10 principaux genres a montré une augmentation des proportions de Cutibacterium, Corynebacterium et Staphylococcus. En outre, la proportion de Dermabacter hominis a été multipliée par 18,9 dans le groupe PS, soit une augmentation significative, tandis que celle de nombreuses souches de Streptococcus s'est avérée significativement plus basse. En se concentrant sur les colonies bactériennes isolées des surfaces cutanées, plus de colonies jaunes ont été trouvées dans le groupe PS lorsqu'elles étaient cultivées dans du milieu de bouillon trypticase soja pendant 48 h, et davantage d'interleukine­8 a été détectée sur les kératinocytes après la stimulation des colonies jaunes comme S. capitis, M. luteus. CONCLUSIONS: Cette étude suggère que davantage de micro­organismes associés au PS peuvent être identifiés à l'aide de la technique 2bRAD, même avec un échantillon de petite taille. Dermabacter hominis et Chryseobacterium ont été rapportés avec une augmentation significative pour les PS, et S. capitis, ainsi que M. luteus, mais pas S. aureus, pouvant être associés à une inflammation cutanée.


Subject(s)
Face , Microbiota , Skin , Humans , Skin/microbiology , Face/microbiology , Adult , Female , Keratinocytes/microbiology , Middle Aged
9.
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
10.
Indian J Med Microbiol ; 40(2): 239-244, 2022.
Article in English | MEDLINE | ID: mdl-35131138

ABSTRACT

PURPOSE: Staphylococcus aureus infections are a major cause of concern in nosocomial infections and especially so, in the hospitalized immunocompromised patients. Staphylococcus epidermidis is a skin commensal that could have a role in preventing colonization on human skin by potential pathogen. METHODS: The probable protective role of S. epidermidis, its lysate (S.epi lysate) and spent culture fluid (SCF) has been explored against S. aureus using human epidermal keratinocytes as a model system. The viability of keratinocytes and bacterial adhesion was investigated pre- and post-exposure to S. epi lysate and SCF. RESULTS: The viability of keratinocytes was significantly reduced when incubated with S. aureus for 24 â€‹h while S. epidermidis and its extracts exhibited no significant effect. S. aureus infected keratinocytes showed increased viability when incubated with viable S. epidermidis which was even greater with its lysate and SCF. The timing of the application of lysate and SCF affected the degree of protection conferred to the keratinocytes against S. aureus induced toxicity. Co-exposed and post-exposed keratinocytes were afforded equal protection. However, a pre-exposure of 2 â€‹h was not efficient enough to provide significant protection. S. epi lysate and SCF reduced the number of adherent cells considerably even after 8 â€‹h of pathogen exposure. CONCLUSIONS: S. epidermidis and its extracts protect human epidermal keratinocytes from the toxic effects of S. aureus by competitive displacement of pathogen and reduction in adhesion. S. epi lysate and SCF are safer options for the treatment of pathogen induced skin damage.


Subject(s)
Staphylococcal Infections , Staphylococcus epidermidis , Humans , Keratinocytes/microbiology , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcus aureus
11.
Emerg Microbes Infect ; 10(1): 2326-2339, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34821529

ABSTRACT

ABSTRACTMelioidosis is a serious infectious disease endemic in Southeast Asia, Northern Australia and has been increasingly reported in other tropical and subtropical regions in the world. Percutaneous inoculation through cuts and wounds on the skin is one of the major modes of natural transmission. Despite cuts in skin being a major route of entry, very little is known about how the causative bacterium Burkholderia pseudomallei initiates an infection at the skin and the disease manifestation at the skin known as cutaneous melioidosis. One key issue is the lack of suitable and relevant infection models. Employing an in vitro 2D keratinocyte cell culture, a 3D skin equivalent fibroblast-keratinocyte co-culture and ex vivo organ culture from human skin, we developed infection models utilizing surrogate model organism Burkholderia thailandensis to investigate Burkholderia-skin interactions. Collectively, these models show that the bacterial infection was largely limited at the wound's edge. Infection impedes wound closure, triggers inflammasome activation and cellular extrusion in the keratinocytes as a potential way to control bacterial infectious load at the skin. However, extensive infection over time could result in the epidermal layer being sloughed off, potentially contributing to formation of skin lesions.


Subject(s)
Burkholderia pseudomallei/physiology , Burkholderia/physiology , Epidermis/microbiology , Inflammasomes/metabolism , Keratinocytes/microbiology , Melioidosis/microbiology , Skin/microbiology , Wounds and Injuries/microbiology , Cells, Cultured , Epidermis/metabolism , Humans , Keratinocytes/metabolism , Melioidosis/metabolism , Melioidosis/pathology , Models, Biological , Skin/metabolism , Skin/pathology , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
12.
Int J Mol Sci ; 22(22)2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34830121

ABSTRACT

Mesenchymal stem cells (MSCs) can improve chronic wound healing; however, recent studies suggest that the therapeutic effect of MSCs is mediated mainly through the growth factors and cytokines secreted by these cells, referred to as the MSC secretome. To overcome difficulties related to the translation of cell therapy into clinical use such as efficacy, safety and cost, we propose a hydrogel loaded with a secretome from the recently established human adipose tissue mesenchymal stem cell line (HATMSC2) as a potential treatment for chronic wounds. Biocompatibility and biological activity of hydrogel-released HATMSC2 supernatant were investigated in vitro by assessing the proliferation and metabolic activity of human fibroblast, endothelial cells and keratinocytes. Hydrogel degradation was measured using hydroxyproline assay while protein released from the hydrogel was assessed by interleukin-8 (IL-8) and macrophage chemoattractant protein-1 (MCP-1) ELISAs. Pro-angiogenic activity of the developed treatment was assessed by tube formation assay while the presence of pro-angiogenic miRNAs in the HATMSC2 supernatant was investigated using real-time RT-PCR. The results demonstrated that the therapeutic effect of the HATMSC2-produced factors is maintained following incorporation into collagen hydrogel as confirmed by increased proliferation of skin-origin cells and improved angiogenic properties of endothelial cells. In addition, HATMSC2 supernatant revealed antimicrobial activity, and which therefore, in combination with the hydrogel has a potential to be used as advanced wound-healing dressing.


Subject(s)
Adipose Tissue/cytology , Culture Media, Conditioned/pharmacology , Hydrogels/pharmacology , Mesenchymal Stem Cells/metabolism , Secretome/metabolism , Skin/metabolism , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Bacteria/classification , Bacteria/drug effects , Bacteria/growth & development , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Culture Media, Conditioned/chemistry , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/microbiology , Humans , Hydrogels/chemistry , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/microbiology , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Skin/cytology , Skin/microbiology
13.
Microbiologyopen ; 10(4): e1203, 2021 08.
Article in English | MEDLINE | ID: mdl-34459556

ABSTRACT

Streptococcus mitis is a commensal bacterial species of the oral cavity, with the potential for opportunistic pathogenesis. For successful colonization, S. mitis must be able to adhere to surfaces of the oral cavity and survive and adapt to frequently changing environmental conditions. Cyclic-di-AMP (c-di-AMP) is a nucleotide second messenger, involved in the regulation of stress responses and biofilm formation in several bacterial species. Cyclic-di-AMP is produced by diadenylate cyclases and degraded by phosphodiesterases. We have previously shown that in S. mitis, one diadenylate cyclase (CdaA) and at least two phosphodiesterases (Pde1 and Pde2) regulate the intracellular concentration of c-di-AMP. In this study, we utilized S. mitis deletion mutants of cdaA, pde1, and pde2 to analyze the role of c-di-AMP signaling in various stress responses, biofilm formation, and adhesion to eukaryotic cells. Here, we demonstrate that the Δpde1 mutant displayed a tendency toward increased susceptibility to acetic acid at pH 4.0. Deletion of cdaA increases auto-aggregation of S. mitis but reduces biofilm formation on an abiotic surface. These phenotypes are more pronounced under acidic extracellular conditions. Inactivation of pde1 or pde2 reduced the tolerance to ciprofloxacin, and UV radiation and the Δpde1 mutant was more susceptible to Triton X-100, indicating a role for c-di-AMP signaling in responses to DNA damage and cell membrane perturbation. Finally, the Δpde2 mutant displayed a tendency toward a reduced ability to adhere to oral keratinocytes. Taken together, our results indicate an important role for c-di-AMP signaling in cellular processes important for colonization of the mouth.


Subject(s)
Adaptation, Physiological/physiology , Bacterial Adhesion/physiology , Biofilms/growth & development , Cyclic AMP/metabolism , Second Messenger Systems/physiology , Streptococcus mitis/metabolism , Acetic Acid/pharmacology , Cell Line, Tumor , Ciprofloxacin/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Gene Deletion , Gene Expression Regulation, Bacterial/genetics , Humans , Keratinocytes/microbiology , Mouth/microbiology , Octoxynol/pharmacology , Phosphorus-Oxygen Lyases/genetics , Phosphorus-Oxygen Lyases/metabolism , Streptococcus mitis/growth & development , Stress, Physiological/physiology
14.
Biomolecules ; 11(8)2021 08 06.
Article in English | MEDLINE | ID: mdl-34439831

ABSTRACT

Wound healing is an essential process to restore tissue integrity after trauma. Large skin wounds such as burns often heal with hypertrophic scarring and contractures, resulting in disfigurements and reduced joint mobility. Such adverse healing outcomes are less common in the oral mucosa, which generally heals faster compared to skin. Several studies have identified differences between oral and skin wound healing. Most of these studies however focus only on a single stage of wound healing or a single cell type. The aim of this review is to provide an extensive overview of wound healing in skin versus oral mucosa during all stages of wound healing and including all cell types and molecules involved in the process and also taking into account environmental specific factors such as exposure to saliva and the microbiome. Next to intrinsic properties of resident cells and differential expression of cytokines and growth factors, multiple external factors have been identified that contribute to oral wound healing. It can be concluded that faster wound closure, the presence of saliva, a more rapid immune response, and increased extracellular matrix remodeling all contribute to the superior wound healing and reduced scar formation in oral mucosa, compared to skin.


Subject(s)
Extracellular Matrix/immunology , Microbiota/immunology , Mouth Mucosa/injuries , Skin/injuries , Wound Healing/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Extracellular Matrix/chemistry , Fibroblasts/immunology , Fibroblasts/microbiology , Gene Expression Regulation , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Keratinocytes/immunology , Keratinocytes/microbiology , Macrophages/immunology , Macrophages/microbiology , Mouth Mucosa/immunology , Mouth Mucosa/microbiology , Mouth Mucosa/pathology , Neutrophils/immunology , Neutrophils/microbiology , Organ Specificity , Saliva/immunology , Saliva/microbiology , Signal Transduction , Skin/immunology , Skin/microbiology , Skin/pathology
15.
Actas dermo-sifiliogr. (Ed. impr.) ; 112(7): 619-624, jul.-ago. 2021. graf
Article in Spanish | IBECS | ID: ibc-213435

ABSTRACT

Antecedentes y objetivo Las micosis superficiales son algunas de las enfermedades más comunes en todo el mundo, siendo los agentes causales más frecuentes las levaduras de los géneros Malassezia y Candida, comensales habituales de la piel que pueden actuar como patógenos oportunistas. El objetivo de este trabajo es investigar si los glicosaminoglicanos (GAG) de las células epiteliales son utilizados por estos microrganismos como receptores de adhesión a las mismas. Materiales y métodos Se utilizaron cultivos de queratinocitos y fibroblastos dérmicos. La participación de los GAG en la adhesión de Candida albicans (C. albicans) y Malassezia spp. se estudió mediante inhibición específica de la síntesis de estas moléculas empleando rodamina B o genisteína. También se analizó mediante digestión enzimática in situ empleando liasas específicas. Resultados El tratamiento con rodamina B produjo una inhibición parcial de la adherencia de ambas especies fúngicas a queratinocitos, pero no a fibroblastos. La digestión selectiva del heparán sulfato produjo un aumento de la unión de Malassezia a los queratinocitos y de ambas especies a los fibroblastos. La digestión del condroitín sulfato redujo la unión de C. albicans en los queratinocitos, pero favoreció la unión de la forma filamentada de esta levadura en los fibroblastos. Conclusiones Los GAG de superficie celular de queratinocitos parecen estar implicados en la adherencia de Candida y Malasezzia a la superficie celular. En los fibroblastos, por el contrario, su eliminación favorece la adherencia, sugiriendo la implicación de otro tipo de receptores (AU)


Background and objective Superficial mycoses are some of the most common diseases worldwide. The usual culprits — yeasts belonging to the genera Malassezia and Candida — are commensal species in the skin that can cause opportunistic infections. We aimed to determine whether these yeasts use glycosaminoglycans (GAGs) as adhesion receptors to mediate binding to epithelial cells. Material and methods In keratinocyte and dermal fibroblast cultures, we used rhodamine B and genistein to inhibit GAG synthesis to study the role these molecules play in the adhesion of Candida albicans (C. albicans) and Malassezia species to cells. We also analyzed GAG involvement by means of enzyme digestion, using specific lyases. Results Rhodamine B partially inhibited the adhesion of both fungi to keratinocytes but not to fibroblasts. Selective digestion of heparan sulfate enhanced the binding of Malassezia species to keratinocytes and of both fungi to fibroblasts. Chondroitin sulfate digestion decreased C. albicans adhesion to keratinocytes, but increased the adhesion of the filamentous forms of this species to fibroblasts. Conclusions Cell surface GAGs appear to play a role in the adhesion of C albicans and Malasezzia species to keratinocytes. In contrast, their adhesion to fibroblasts appears to be enhanced by GAG inhibition, suggesting that some other type of receptor is the mediator (AU)


Subject(s)
Humans , Glycosaminoglycans/metabolism , Candida albicans/physiology , Malassezia/physiology , Keratinocytes/microbiology , Fibroblasts/microbiology , Rhodamines/pharmacology , Candida albicans/drug effects , Malassezia/drug effects
16.
mSphere ; 6(4): e0060821, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34319127

ABSTRACT

Atopic dermatitis (AD) is a condition affecting 30 million persons in the United States. AD patients are heavily infected with Staphylococcus aureus on the skin. A particularly severe form of AD is eczema herpeticum (ADEH), where the patients' AD is complicated by S. aureus and herpes simplex virus (HSV) infection. This study examined the S. aureus strains from 15 ADEH patients, provided blinded, and showed a high association of ADEH with strains that produce toxic shock syndrome toxin-1 (TSST-1; 73%) compared to 10% production by typical AD isolates from patients without EH and those from another unrelated condition, cystic fibrosis. The ADEH isolates produced the superantigens associated with TSS (TSST-1 and staphylococcal enterotoxins A, B, and C). This association may in part explain the potential severity of ADEH. We also examined the effect of TSST-1 and HSV-1 on human epithelial cells and keratinocytes. TSST-1 used CD40 as its receptor on epithelial cells, and HSV-1 either directly or indirectly interacted with CD40. The consequence of these interactions was chemokine production, which is capable of causing harmful inflammation, with epidermal/keratinocyte barrier disruption. Human epithelial cells treated first with TSST-1 and then HSV-1 resulted in enhanced chemokine production. Finally, we showed that TSST-1 modestly increased HSV-1 replication but did not increase viral plaque size. Our data suggest that ADEH is associated with production of the major TSS-associated superantigens, together with HSV reactivation. The superantigens plus HSV may damage the skin barrier by causing harmful inflammation, thereby leading to increased symptoms. IMPORTANCE Atopic dermatitis (eczema, AD) with concurrent herpes simplex virus infection (eczema herpeticum, ADEH) is a severe form of AD. We show that ADEH patients are colonized with Staphylococcus aureus that primarily produces the superantigen toxic shock syndrome toxin-1 (TSST-1); however, significantly but to a lesser extent the superantigens staphylococcal enterotoxins A, B, and C are also represented in ADEH. Our studies showed that TSST-1 uses the immune costimulatory molecule CD40 as its epithelial cell receptor. Herpes simplex virus (HSV) also interacted directly or indirectly with CD40 on epithelial cells. Treatment of epithelial cells with TSST-1 and then HSV-1 resulted in enhanced chemokine production. We propose that this combination of exposures (TSST-1 and then HSV) leads to opening of epithelial and skin barriers to facilitate potentially serious ADEH.


Subject(s)
Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Enterotoxins/genetics , Enterotoxins/metabolism , Herpesvirus 1, Human/metabolism , Kaposi Varicelliform Eruption/microbiology , Staphylococcus aureus/pathogenicity , Superantigens/genetics , Superantigens/metabolism , Bacterial Toxins/immunology , Bacterial Toxins/pharmacology , CD40 Antigens/immunology , Chemokines/immunology , Enterotoxins/immunology , Enterotoxins/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/virology , HaCaT Cells , Herpesvirus 1, Human/immunology , Humans , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/microbiology , Keratinocytes/virology , Staphylococcus aureus/metabolism , Superantigens/immunology , Superantigens/pharmacology
17.
Nat Immunol ; 22(7): 839-850, 2021 07.
Article in English | MEDLINE | ID: mdl-34168371

ABSTRACT

Granulomas are complex cellular structures composed predominantly of macrophages and lymphocytes that function to contain and kill invading pathogens. Here, we investigated the single-cell phenotypes associated with antimicrobial responses in human leprosy granulomas by applying single-cell and spatial sequencing to leprosy biopsy specimens. We focused on reversal reactions (RRs), a dynamic process whereby some patients with disseminated lepromatous leprosy (L-lep) transition toward self-limiting tuberculoid leprosy (T-lep), mounting effective antimicrobial responses. We identified a set of genes encoding proteins involved in antimicrobial responses that are differentially expressed in RR versus L-lep lesions and regulated by interferon-γ and interleukin-1ß. By integrating the spatial coordinates of the key cell types and antimicrobial gene expression in RR and T-lep lesions, we constructed a map revealing the organized architecture of granulomas depicting compositional and functional layers by which macrophages, T cells, keratinocytes and fibroblasts can each contribute to the antimicrobial response.


Subject(s)
Leprosy, Lepromatous/immunology , Leprosy, Tuberculoid/immunology , Mycobacterium leprae/immunology , Skin/immunology , Adolescent , Adult , Aged , Female , Fibroblasts/immunology , Fibroblasts/microbiology , Fibroblasts/pathology , Gene Expression Profiling , Host-Pathogen Interactions , Humans , Keratinocytes/immunology , Keratinocytes/microbiology , Keratinocytes/pathology , Leprosy, Lepromatous/genetics , Leprosy, Lepromatous/microbiology , Leprosy, Lepromatous/pathology , Leprosy, Tuberculoid/genetics , Leprosy, Tuberculoid/microbiology , Leprosy, Tuberculoid/pathology , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Male , Middle Aged , Mycobacterium leprae/pathogenicity , RNA-Seq , Single-Cell Analysis , Skin/microbiology , Skin/pathology , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , T-Lymphocytes/pathology , Transcriptome
18.
mBio ; 12(3): e0050221, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34182783

ABSTRACT

Periodontal disease (PD) is an inflammatory disease of the supporting tissues of the teeth that develops in response to formation of a dysbiotic biofilm on the subgingival tooth surface. Although exacerbated inflammation leads to alveolar bone destruction and may cause tooth loss, the molecular basis of PD initiation and progression remains elusive. Control over the inflammatory reaction and return to homeostasis can be efficiently restored by negative regulators of Toll-like receptor (TLR) signaling pathways such as monocyte chemoattractant protein-induced protein 1 (MCPIP-1), which is constitutively expressed in gingival keratinocytes and prevents hyperresponsiveness in the gingiva. Here, we found that inflammophilic periodontal species influence the stability of MCPIP-1, leading to an aggravated response of the epithelium to proinflammatory stimulation. Among enzymes secreted by periodontal species, gingipains-cysteine proteases from Porphyromonas gingivalis-are considered major contributors to the pathogenic potential of bacteria, strongly influencing the components of the innate and adaptive immune system. Gingipain proteolytic activity leads to a rapid degradation of MCPIP-1, exacerbating the inflammatory response induced by endotoxin. Collectively, these results establish a novel mechanism of corruption of inflammatory signaling by periodontal pathogens, indicating new possibilities for treatment of this chronic disease. IMPORTANCE Periodontitis is a highly prevalent disease caused by accumulation of a bacterial biofilm. Periodontal pathogens use a number of virulence strategies that are under intensive study to find optimal therapeutic approaches against bone loss. In our work, we present a novel mechanism utilized by the key periodontal pathogen Porphyromonas gingivalis, based on the selective degradation of the negative regulator of inflammation, MCPIP-1. We found that the diminished levels of MCPIP-1 in gingival keratinocytes-cells at the forefront of the fight against bacteria-cause sensitization to endotoxins produced by other oral species. This results in an enhanced inflammatory response, which promotes the growth of inflammophilic pathobionts and damage of tooth-supporting tissues. Our observation is relevant to understanding the molecular basis of periodontitis and the development of new methods for treatment.


Subject(s)
Gingiva/cytology , Inflammation , Keratinocytes/immunology , Lipopolysaccharides/metabolism , Porphyromonas gingivalis/immunology , Porphyromonas gingivalis/metabolism , Ribonucleases/metabolism , Signal Transduction , Animals , Biofilms/growth & development , Cells, Cultured , Female , Gingipain Cysteine Endopeptidases , Keratinocytes/metabolism , Keratinocytes/microbiology , Mice , Mice, Inbred C57BL , Periodontitis/microbiology , Porphyromonas gingivalis/physiology , Ribonucleases/genetics , Ribonucleases/immunology , Specific Pathogen-Free Organisms
19.
mBio ; 12(3)2021 05 18.
Article in English | MEDLINE | ID: mdl-34006656

ABSTRACT

Early childhood caries is a severe oral disease that results in aggressive tooth decay. Particularly, a synergistic association between a fungus, Candida albicans, and a cariogenic bacterium, Streptococcus mutans, promotes the development of hard-to-remove and highly acidic biofilms, exacerbating the virulent damage. These interactions are largely mediated via glucosyltransferases (GtfB) binding to mannans on the cell wall of C. albicans Here, we present an enzymatic approach to target GtfB-mannan interactions in this cross-kingdom consortium using mannan-degrading exo- and endo-enzymes. These exo- and endo-enzymes are highly effective in reducing biofilm biomass without killing microorganisms, as well as alleviating the production of an acidic pH environment conducive to tooth decay. To corroborate these results, we present biophysical evidence using single-molecule atomic force microscopy, biofilm shearing, and enamel surface topography analyses. Data show a drastic decrease in binding forces of GtfB to C. albicans (∼15-fold reduction) following enzyme treatment. Furthermore, enzymatic activity disrupted biofilm mechanical stability and significantly reduced human tooth enamel demineralization without cytotoxic effects on gingival keratinocytes. Our results represent significant progress toward a novel nonbiocidal therapeutic intervention against pathogenic bacterial-fungal biofilms by targeting the interkingdom receptor-ligand binding interactions.IMPORTANCE Biofilm formation is a key virulence factor responsible for various infectious diseases. Particularly, interactions between a fungus, Candida albicans, and a bacterium, Streptococcus mutans, have been known to play important roles in the pathogenesis of dental caries. Although some antimicrobials have been applied to treat fungal-involved biofilm-associated diseases, these often lack targeting polymicrobial interactions. Furthermore, these may not be appropriate for preventive measures because these antimicrobials may disrupt ecological microbiota and/or induce the prevalence of drug resistance over time. By specifically targeting the interaction mechanism whereby mannoproteins on the C. albicans surface mediate the cross-kingdom interaction, we demonstrated that mannoprotein-degrading enzymes can effectively disrupt biofilm interactions without microbiocidal effects or causing cytotoxicity to human cells. This suggests a potential application as a targeted approach for intervening a pathogenic cross-kingdom biofilm associated with a costly and unresolved oral disease.


Subject(s)
Biofilms/growth & development , Candida albicans/metabolism , Streptococcus mutans/metabolism , Symbiosis , Dental Caries/microbiology , Gingiva/cytology , Humans , Keratinocytes/microbiology , Mannans/metabolism , Microscopy, Atomic Force
20.
Int J Mol Sci ; 22(6)2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33804147

ABSTRACT

Psoriasis is a chronic inflammatory skin disease characterized by IL-17-dominant abnormal innate and acquired immunity, and the hyperproliferation and aberrant differentiation of epidermal keratinocytes, and comorbid arthritis or cardiometabolic diseases. This Special Issue presented updated information on pathogenesis, comorbidities, and therapy of psoriasis. The pathogenesis of psoriasis may involve the dysfunction of indoleamine 2,3-dioxygenase 2 or of UBA domain containing 1-mediated regulation of CARD14/CARMA2sh. The blood cells of psoriasis patients showed the enhanced oxidative stress/autophagy flux and decreased 20S proteasome activity. Elafin, clusterin, or selenoprotein P may act as biomarkers for psoriasis and comorbid metabolic diseases. The proteomic profile of psoriasis lesions showed the dysfunction of dermal fibroblasts; up-regulation of proinflammatory factors and signal transduction or down-regulation of structural molecules. The skin inflammation in psoriasis may populate certain gut bacteria, such as Staphylococcus aureus and Streptococcus danieliae, which worsen the skin inflammation in turn. The psoriasis-associated pruritus may be caused by immune, nervous, or vascular mechanisms. In addition to current oral treatments and biologics, a new treatment option for psoriasis is now being developed, such as retinoic-acid-receptor-related orphan nuclear receptor γt inhibitors, IL-36 receptor antagonist, or aryl hydrocarbon receptor agonist. Antimicrobial peptides and innate immune cells, involved in the pathogenesis of psoriasis, may be novel therapeutic targets. The pathomechanisms and responses to drugs in collagen diseases are partially shared with and partially different from those in psoriasis. Certain nutrients can exacerbate or regulate the progress of psoriasis. The articles in this Special Issue will encourage attractive approaches to psoriasis by future researchers.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , Guanylate Cyclase/genetics , Immunity, Innate/genetics , Inflammation/genetics , Membrane Proteins/genetics , Psoriasis/genetics , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Inflammation/pathology , Inflammation/therapy , Interleukin-17/genetics , Keratinocytes/microbiology , Keratinocytes/pathology , Proteomics , Psoriasis/microbiology , Psoriasis/therapy , Skin/microbiology , Skin/pathology , Staphylococcus aureus/pathogenicity , Streptococcus/pathogenicity , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...