Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 549
Filtrar
1.
Exp Dermatol ; 33(8): e15150, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39113601

RESUMO

Acne is a chronic inflammatory skin condition that involves Cutibacterium acnes (C. acnes), which is classified into six main phylotypes (IA1, IA2, IB, IC, II and III). Acne development is associated with loss of C. acnes phylotype diversity, characterised by overgrowth of phylotype IA1 relative to other phylotypes. It was also shown that purified extracellular vesicles (EVs) secreted by C. acnes can induce an acne-like inflammatory response in skin models. We aimed to determine if the inflammatory profile of EVs secreted by C. acnes phylotype IA1 from an inflammatory acne lesion was different from C. acnes phylotype IA1 from normal skin, thus playing a direct role in the severity of inflammation. EVs were produced in vitro after culture of two clinical strains of C. acnes phylotype IA1, T5 from normal human skin and A47 from an inflammatory acne lesion, and then incubated with either human immortalised keratinocytes, HaCaT cells, or skin explants obtained from abdominoplasty. Subsequently, quantitative PCR (qPCR) was performed for human ß-defensin 2 (hBD2), cathelicidin (LL-37), interleukin (IL)-1ß, IL-6, IL-8, IL-17α and IL-36γ, and ELISA for IL-6, IL-8 and IL-17α. We found that EVs produced in vitro by C. acnes derived from inflammatory acne lesions significantly increased the pro-inflammatory cytokines and anti-microbial peptides at both transcriptional and protein levels compared with EVs derived from normal human skin. We show for the first time that C. acnes EVs from inflammatory acne play a crucial role in acne-associated inflammation in vitro and that C. acnes phylotype IA1 collected from inflammatory acne lesion and normal skin produce different EVs and inflammatory profiles in vitro.


Assuntos
Acne Vulgar , Vesículas Extracelulares , Queratinócitos , Propionibacterium acnes , Humanos , Vesículas Extracelulares/metabolismo , Acne Vulgar/microbiologia , Queratinócitos/microbiologia , Pele/microbiologia , Inflamação/microbiologia , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Células HaCaT , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Citocinas/metabolismo , Interleucina-17/metabolismo , Propionibacteriaceae
2.
Int J Mol Sci ; 25(15)2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39125871

RESUMO

Caffeine affords several beneficial effects on human health, acting as an antioxidant, anti-inflammatory agent, and analgesic. Caffeine is widely used in cosmetics, but its antimicrobial activity has been scarcely explored, namely against skin infection agents. Dermatophytes are the most common fungal agents of human infection, mainly of skin infections. This work describes the in vitro effect of caffeine during keratinocyte infection by Trichophyton mentagrophytes, one of the most common dermatophytes. The results show that caffeine was endowed with antidermatophytic activity with a MIC, determined following the EUCAST standards, of 8 mM. Caffeine triggered a modification of the levels of two major components of the fungal cell wall, ß-(1,3)-glucan and chitin. Caffeine also disturbed the ultrastructure of the fungal cells, particularly the cell wall surface and mitochondria, and autophagic-like structures were observed. During dermatophyte-human keratinocyte interactions, caffeine prevented the loss of viability of keratinocytes and delayed spore germination. Overall, this indicates that caffeine can act as a therapeutic and prophylactic agent for dermatophytosis.


Assuntos
Antifúngicos , Arthrodermataceae , Cafeína , Queratinócitos , Cafeína/farmacologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/microbiologia , Humanos , Antifúngicos/farmacologia , Arthrodermataceae/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Parede Celular/efeitos dos fármacos , Tinha/tratamento farmacológico , Tinha/microbiologia , Quitina/farmacologia , Quitina/química
3.
Science ; 385(6705): 194-200, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-38991070

RESUMO

Millions of hibernating bats across North America have died from white-nose syndrome (WNS), an emerging disease caused by a psychrophilic (cold-loving) fungus, Pseudogymnoascus destructans, that invades their skin. Mechanisms of P. destructans invasion of bat epidermis remain obscure. Guided by our in vivo observations, we modeled hibernation with a newly generated little brown bat (Myotis lucifugus) keratinocyte cell line. We uncovered the stealth intracellular lifestyle of P. destructans, which inhibits apoptosis of keratinocytes and spreads through the cells by two epidermal growth factor receptor (EGFR)-dependent mechanisms: active penetration during torpor and induced endocytosis during arousal. Melanin of endocytosed P. destructans blocks endolysosomal maturation, facilitating P. destructans survival and germination after return to torpor. Blockade of EGFR aborts P. destructans entry into keratinocytes.


Assuntos
Nível de Alerta , Ascomicetos , Quirópteros , Receptores ErbB , Hibernação , Queratinócitos , Animais , Apoptose , Ascomicetos/fisiologia , Ascomicetos/patogenicidade , Linhagem Celular , Quirópteros/microbiologia , Quirópteros/fisiologia , Endocitose , Receptores ErbB/metabolismo , Queratinócitos/microbiologia , Melaninas/metabolismo
4.
FASEB J ; 38(14): e23801, 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39018106

RESUMO

Intracellular pathogens including Staphylococcus aureus contribute to the non-healing phenotype of chronic wounds. Lactobacilli, well known as beneficial bacteria, are also reported to modulate the immune system, yet their role in cutaneous immunity remains largely unknown. We explored the therapeutic potential of bacteria-free postbiotics, bioactive lysates of lactobacilli, to reduce intracellular S. aureus colonization and promote healing. Fourteen postbiotics derived from various lactobacilli species were screened, and Latilactobacillus curvatus BGMK2-41 was selected for further analysis based on the most efficient ability to reduce intracellular infection by S. aureus diabetic foot ulcer clinical isolate and S. aureus USA300. Treatment of both infected keratinocytes in vitro and infected human skin ex vivo with BGMK2-41 postbiotic cleared S. aureus. Keratinocytes treated in vitro with BGMK2-41 upregulated expression of antimicrobial response genes, of which DEFB4, ANG, and RNASE7 were also found upregulated in treated ex vivo human skin together with CAMP exclusively upregulated ex vivo. Furthermore, BGMK2-41 postbiotic treatment has a multifaceted impact on the wound healing process. Treatment of keratinocytes stimulated cell migration and the expression of tight junction proteins, while in ex vivo human skin BGMK2-41 increased expression of anti-inflammatory cytokine IL-10, promoted re-epithelialization, and restored the epidermal barrier via upregulation of tight junction proteins. Together, this provides a potential therapeutic approach for persistent intracellular S. aureus infections.


Assuntos
Queratinócitos , Lactobacillus , Staphylococcus aureus , Humanos , Queratinócitos/microbiologia , Queratinócitos/metabolismo , Queratinócitos/efeitos dos fármacos , Pele/microbiologia , Pele/metabolismo , Cicatrização/efeitos dos fármacos , Probióticos/farmacologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/metabolismo , Ribonucleases/metabolismo
5.
Infect Immun ; 92(8): e0013324, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-38953668

RESUMO

Staphylococcus aureus α-hemolysin (Hla) is a pore-forming toxin critical for the pathogenesis of skin and soft tissue infections, which causes the pathognomonic lesion of cutaneous necrosis (dermonecrosis) in mouse models. To determine the mechanism by which dermonecrosis develops during S. aureus skin infection, mice were given control serum, Hla-neutralizing antiserum, or an inhibitor of Hla receptor [A-disintegrin and metalloprotease 10 (ADAM10) inhibitor] followed by subcutaneous infection by S. aureus, and the lesions were evaluated using immunohistochemistry and immunofluorescence. Hla induced apoptosis in the vascular endothelium at 6 hours post-infection (hpi), followed by apoptosis in keratinocytes at 24 hpi. The loss of vascular endothelial (VE)-cadherin expression preceded the loss of epithelial-cadherin expression. Hla also induced hypoxia in the keratinocytes at 24 hpi following vascular injury. Treatment with Hla-neutralizing antibody or ADAM10 inhibitor attenuated early cleavage of VE-cadherin, cutaneous hypoxia, and dermonecrosis. These findings suggest that Hla-mediated vascular injury with cutaneous hypoxia underlies the pathogenesis of S. aureus-induced dermonecrosis.


Assuntos
Proteína ADAM10 , Toxinas Bacterianas , Caderinas , Proteínas Hemolisinas , Queratinócitos , Necrose , Staphylococcus aureus , Animais , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/toxicidade , Camundongos , Toxinas Bacterianas/toxicidade , Toxinas Bacterianas/metabolismo , Staphylococcus aureus/patogenicidade , Queratinócitos/microbiologia , Queratinócitos/metabolismo , Proteína ADAM10/metabolismo , Caderinas/metabolismo , Apoptose , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Antígenos CD/metabolismo , Proteínas de Membrana/metabolismo , Infecções Cutâneas Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/patologia , Infecções Cutâneas Estafilocócicas/imunologia , Pele/patologia , Pele/microbiologia , Feminino , Endotélio Vascular/patologia , Endotélio Vascular/microbiologia , Endotélio Vascular/metabolismo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/patologia , Modelos Animais de Doenças
6.
Sci Adv ; 10(26): eadj2020, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38924411

RESUMO

Chronic wounds are a common and costly complication of diabetes, where multifactorial defects contribute to dysregulated skin repair, inflammation, tissue damage, and infection. We previously showed that aspects of the diabetic foot ulcer microbiota were correlated with poor healing outcomes, but many microbial species recovered remain uninvestigated with respect to wound healing. Here, we focused on Alcaligenes faecalis, a Gram-negative bacterium that is frequently recovered from chronic wounds but rarely causes infection. Treatment of diabetic wounds with A. faecalis accelerated healing during early stages. We investigated the underlying mechanisms and found that A. faecalis treatment promotes reepithelialization of diabetic keratinocytes, a process that is necessary for healing but deficient in chronic wounds. Overexpression of matrix metalloproteinases in diabetes contributes to failed epithelialization, and we found that A. faecalis treatment balances this overexpression to allow proper healing. This work uncovers a mechanism of bacterial-driven wound repair and provides a foundation for the development of microbiota-based wound interventions.


Assuntos
Alcaligenes faecalis , Queratinócitos , Metaloproteinases da Matriz , Cicatrização , Alcaligenes faecalis/metabolismo , Animais , Queratinócitos/metabolismo , Queratinócitos/microbiologia , Humanos , Metaloproteinases da Matriz/metabolismo , Metaloproteinases da Matriz/genética , Pé Diabético/microbiologia , Pé Diabético/patologia , Pé Diabético/metabolismo , Camundongos , Reepitelização , Masculino
7.
Cell Host Microbe ; 32(7): 1129-1146.e8, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38936370

RESUMO

Cutibacterium acnes is the most abundant bacterium of the human skin microbiome since adolescence, participating in both skin homeostasis and diseases. Here, we demonstrate individual and niche heterogeneity of C. acnes from 1,234 isolate genomes. Skin disease (atopic dermatitis and acne) and body site shape genomic differences of C. acnes, stemming from horizontal gene transfer and selection pressure. C. acnes harbors characteristic metabolic functions, fewer antibiotic resistance genes and virulence factors, and a more stable genome compared with Staphylococcus epidermidis. Integrated genome, transcriptome, and metabolome analysis at the strain level unveils the functional characteristics of C. acnes. Consistent with the transcriptome signature, C. acnes in a sebum-rich environment induces toxic and pro-inflammatory effects on keratinocytes. L-carnosine, an anti-oxidative stress metabolite, is up-regulated in the C. acnes metabolome from atopic dermatitis and attenuates skin inflammation. Collectively, our study reveals the joint impact of genes and the microenvironment on C. acnes function.


Assuntos
Acne Vulgar , Dermatite Atópica , Queratinócitos , Propionibacterium acnes , Pele , Humanos , Pele/microbiologia , Dermatite Atópica/microbiologia , Dermatite Atópica/genética , Queratinócitos/microbiologia , Acne Vulgar/microbiologia , Propionibacterium acnes/genética , Genômica , Genoma Bacteriano , Staphylococcus epidermidis/genética , Transcriptoma , Fatores de Virulência/genética , Propionibacteriaceae/genética , Metaboloma , Metabolômica , Microbiota/genética , Multiômica
8.
Elife ; 132024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38767331

RESUMO

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.


Assuntos
Enterococcus faecalis , Infecções por Bactérias Gram-Positivas , Queratinócitos , Cicatrização , Enterococcus faecalis/fisiologia , Enterococcus faecalis/genética , Animais , Camundongos , Infecções por Bactérias Gram-Positivas/microbiologia , Queratinócitos/microbiologia , Queratinócitos/metabolismo , Macrófagos/microbiologia , Macrófagos/metabolismo , Macrófagos/imunologia , Modelos Animais de Doenças , Infecção dos Ferimentos/microbiologia , Transcriptoma , Camundongos Endogâmicos C57BL , Análise de Célula Única , Transição Epitelial-Mesenquimal/genética , Masculino , Fibroblastos/microbiologia , Fibroblastos/metabolismo
9.
Infect Immun ; 92(6): e0008324, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38712951

RESUMO

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.


Assuntos
Proteínas de Bactérias , Regulação Bacteriana da Expressão Gênica , Streptococcus pyogenes , Fatores de Virulência , Streptococcus pyogenes/patogenicidade , Streptococcus pyogenes/genética , Streptococcus pyogenes/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Virulência , Fosforilação , Humanos , Regulon , Óperon , Infecções Estreptocócicas/microbiologia , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/metabolismo , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/genética , Queratinócitos/microbiologia
10.
Mycopathologia ; 189(3): 37, 2024 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-38704808

RESUMO

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.


Assuntos
Processamento Alternativo , Arthrodermataceae , Regulação Fúngica da Expressão Gênica , Proteínas Quinases Ativadas por Mitógeno , Tinha , Fatores de Transcrição , Humanos , Arthrodermataceae/genética , Arthrodermataceae/metabolismo , Glucose/metabolismo , Queratinócitos/microbiologia , Queratinas/metabolismo , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Tinha/metabolismo , Tinha/microbiologia
11.
Microbiol Spectr ; 12(6): e0034724, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38700333

RESUMO

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.


Assuntos
Biofilmes , Escherichia coli , Queratinócitos , Probióticos , Pseudomonas aeruginosa , Staphylococcus aureus , Humanos , Queratinócitos/microbiologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/fisiologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia , Streptococcus pyogenes/efeitos dos fármacos , Streptococcus pyogenes/crescimento & desenvolvimento , Streptococcus pyogenes/fisiologia , Acinetobacter baumannii/efeitos dos fármacos , Acinetobacter baumannii/fisiologia , Acinetobacter baumannii/crescimento & desenvolvimento , Infecção dos Ferimentos/microbiologia
12.
mSystems ; 9(5): e0017924, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38656122

RESUMO

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.


Assuntos
Trifosfato de Adenosina , Interações Hospedeiro-Patógeno , Queratinócitos , Macrófagos , Staphylococcus aureus , Humanos , Staphylococcus aureus/metabolismo , Trifosfato de Adenosina/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Macrófagos/microbiologia , Macrófagos/metabolismo , Macrófagos/imunologia , Queratinócitos/microbiologia , Queratinócitos/metabolismo , Queratinócitos/imunologia , Células THP-1 , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Proteômica/métodos , Proteínas de Bactérias/metabolismo , Células HaCaT
13.
Exp Dermatol ; 33(5): e15087, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38685821

RESUMO

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.


Assuntos
Hidradenite Supurativa , Queratinócitos , Queratinócitos/imunologia , Queratinócitos/microbiologia , Queratinócitos/metabolismo , Humanos , Animais , Camundongos , Hidradenite Supurativa/microbiologia , Hidradenite Supurativa/imunologia , Staphylococcus aureus/imunologia , Staphylococcus epidermidis/imunologia , Fusobacterium nucleatum/imunologia , Transcriptoma , Citocinas/metabolismo , Bactérias Anaeróbias , Interleucina-17/metabolismo , Microbiota , Prevotella/imunologia
14.
Future Microbiol ; 19(7): 585-594, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38629904

RESUMO

Aim: To evaluate the behavior of oral keratinocytes in the presence of Vitamin C (Vit C) and its anti-inflammatory potential. Materials & methods: Oral keratinocytes were initially exposed to 0.1-2.5 mM of Vit C and the metabolic activity and cell migration were evaluated using MTS assay and Ibidi culture inserts, respectively. After, the cells were challenged with Candida albicans and inflammatory markers were analyzed by qPCR. Results: The treatment was not cytotoxic, and the highest concentrations increased the metabolic activity at 24 h. Vit C delayed the cell migration at 48 and 72 h. Interestingly, it downregulated the genes IL-8 and IL-1ß. Conclusion: Vit C could be an interesting adjuvant to anti-fungal treatment due to its anti-inflammatory potential.


Vitamin C, also known as ascorbic acid, is a vitamin commonly found in fruits and vegetables. It is popular for supporting our immune system, so is commonly taken as a supplement. We looked at the action of vitamin C on cells from the mouth and its potential to reduce inflammation in a fungal disease of the mouth ­ oral candidiasis. We showed that vitamin C is not toxic to cells of the mouth and may reduce inflammation in cells infected by the fungus. This suggests that vitamin C could be used as a complementary therapy for oral candidiasis.


Assuntos
Anti-Inflamatórios , Ácido Ascórbico , Candida albicans , Movimento Celular , Queratinócitos , Candida albicans/efeitos dos fármacos , Candida albicans/imunologia , Ácido Ascórbico/farmacologia , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/imunologia , Queratinócitos/microbiologia , Queratinócitos/metabolismo , Anti-Inflamatórios/farmacologia , Movimento Celular/efeitos dos fármacos , Interleucina-8/metabolismo , Interleucina-8/genética , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Inflamação , Antifúngicos/farmacologia
15.
Cell Rep ; 43(4): 114022, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38568806

RESUMO

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.


Assuntos
Aderência Bacteriana , Queratinócitos , Pele , Staphylococcus aureus , Staphylococcus aureus/metabolismo , Humanos , Pele/microbiologia , Pele/metabolismo , Queratinócitos/microbiologia , Queratinócitos/metabolismo , Lectinas/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Filogenia , Ligação Proteica
16.
Int J Cosmet Sci ; 46(3): 414-423, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38229273

RESUMO

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.


Assuntos
Face , Microbiota , Pele , Humanos , Pele/microbiologia , Face/microbiologia , Adulto , Feminino , Queratinócitos/microbiologia , Pessoa de Meia-Idade
17.
J Innate Immun ; 15(1): 822-835, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37903473

RESUMO

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.


Assuntos
Acne Vulgar , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas do Tecido Nervoso , Propionibacteriaceae , Fatores de Processamento de RNA , RNA Circular , Humanos , Propionibacteriaceae/fisiologia , Acne Vulgar/imunologia , Acne Vulgar/microbiologia , Células Cultivadas , Queratinócitos/imunologia , Queratinócitos/microbiologia , RNA Circular/genética , Regulação para Baixo , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Transporte Biológico Ativo , Fatores de Processamento de RNA/metabolismo , Proteínas do Tecido Nervoso/metabolismo
18.
Indian J Med Microbiol ; 40(2): 239-244, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35131138

RESUMO

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.


Assuntos
Infecções Estafilocócicas , Staphylococcus epidermidis , Humanos , Queratinócitos/microbiologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus
19.
Emerg Microbes Infect ; 10(1): 2326-2339, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34821529

RESUMO

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.


Assuntos
Burkholderia pseudomallei/fisiologia , Burkholderia/fisiologia , Epiderme/microbiologia , Inflamassomos/metabolismo , Queratinócitos/microbiologia , Melioidose/microbiologia , Pele/microbiologia , Ferimentos e Lesões/microbiologia , Células Cultivadas , Epiderme/metabolismo , Humanos , Queratinócitos/metabolismo , Melioidose/metabolismo , Melioidose/patologia , Modelos Biológicos , Pele/metabolismo , Pele/patologia , Ferimentos e Lesões/metabolismo , Ferimentos e Lesões/patologia
20.
Int J Mol Sci ; 22(22)2021 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-34830121

RESUMO

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.


Assuntos
Tecido Adiposo/citologia , Meios de Cultivo Condicionados/farmacologia , Hidrogéis/farmacologia , Células-Tronco Mesenquimais/metabolismo , Secretoma/metabolismo , Pele/metabolismo , Anti-Infecciosos/química , Anti-Infecciosos/farmacologia , Bactérias/classificação , Bactérias/efeitos dos fármacos , Bactérias/crescimento & desenvolvimento , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Meios de Cultivo Condicionados/química , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/microbiologia , Humanos , Hidrogéis/química , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/microbiologia , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , Pele/citologia , Pele/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...