Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 375
Filter
1.
Skin Res Technol ; 30(9): e70035, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39218780

ABSTRACT

BACKGROUND: Atopic dermatitis (AD), psoriasis (PSO), rosacea, and other related immune skin diseases are affected by multiple complex factors such as genetic and microbial components. This research investigates the causal relationships between specific skin microbiota and these diseases by using Mendelian randomization (MR), and Bayesian weighted Mendelian randomization (BWMR). METHODS: We utilized genome-wide association study (GWAS) data to analyze the associations between various skin bacteria and three dermatological diseases. Single nucleotide polymorphisms (SNPs) served as instrumental variables (IVs) in MR methods, including inverse variance weighted (IVW), and MR Egger. BWMR was employed to validate results and address pleiotropy. RESULTS: The IVW analysis identified significant associations between specific skin microbiota and dermatological diseases. ASV006_Dry, ASV076_Dry, and Haemophilus_Dry were significantly positively associated with AD, whereas Kocuria_Dry was negatively associated. In PSO, ASV005_Dry was negatively associated, whereas ASV004_Dry, Rothia_Dry, and Streptococcus_Moist showed positive associations. For rosacea, ASV023_Dry was significantly positively associated, while ASV016_Moist, Finegoldia_Dry, and Rhodobacteraceae_Moist were significantly negatively associated. These results were corroborated by BWMR analysis. CONCLUSION: Bacterial species such as Finegoldia, Rothia, and Streptococcus play crucial roles in the pathogenesis of AD, PSO, and rosacea. Understanding these microbial interactions can aid in developing targeted treatments and preventive strategies, enhancing patient outcomes and quality of life.


Subject(s)
Bayes Theorem , Dermatitis, Atopic , Genome-Wide Association Study , Mendelian Randomization Analysis , Microbiota , Polymorphism, Single Nucleotide , Humans , Microbiota/genetics , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/genetics , Skin/microbiology , Rosacea/microbiology , Rosacea/genetics , Skin Diseases/microbiology , Skin Diseases/genetics , Psoriasis/microbiology , Psoriasis/genetics
2.
Sci Rep ; 14(1): 21743, 2024 09 18.
Article in English | MEDLINE | ID: mdl-39289407

ABSTRACT

Onychomycosis, a nail infection caused by dermatophytes, yeast, and molds makes up roughly half of all onychopathies and is the most prevalent nail condition in the world. Clinically, nail psoriasis and onychomycosis can frequently be difficult to distinguish from one another. To assess the prevalence of onychomycosis in patients with psoriasis. Fifty patients with psoriasis associated with nail disease were included in this study. After taking clinical history, nail samples were gathered for dermoscopic inspection, culture, direct microscopy with 20% KOH solution, and nail clipping with PAS stain. Of the 50 patients recruited, 43 were males and 7 were females, with mean age 6-71 years (mean ± SD 44.06 ± 16.2). Eleven patients (22%) tested positive for onychomycosis. Dermatophytes were isolated from 2% of patients, yeast from 14% of patients, and non-dermatophytic mold from 38% of patients. Histopathological results revealed fungal hyphae and spores in 18% of patients. The most prevalent dermoscopic sign in psoriatic patients with onychomycosis was spikes (81.8%) with statistical significance (P-value < 0.001), while nail pitting was the most prevalent dermoscopic feature in nail psoriasis. This study lays the way for an accurate diagnosis of nail lesions by highlighting the significance of cooperation between mycology, histology, and dermoscopy in the diagnosis of onychomycosis in patients with nail psoriasis.


Subject(s)
Dermoscopy , Onychomycosis , Psoriasis , Humans , Onychomycosis/microbiology , Onychomycosis/epidemiology , Onychomycosis/pathology , Onychomycosis/diagnostic imaging , Psoriasis/microbiology , Psoriasis/complications , Psoriasis/pathology , Female , Male , Middle Aged , Adult , Adolescent , Aged , Cross-Sectional Studies , Prevalence , Young Adult , Dermoscopy/methods , Child , Arthrodermataceae/isolation & purification , Nails/microbiology , Nails/pathology
3.
Sci Rep ; 14(1): 18402, 2024 08 08.
Article in English | MEDLINE | ID: mdl-39117753

ABSTRACT

Gaining a comprehensive understanding of the role played by the oral microbiome in moderate to severe plaque psoriasis and its potential implications for disease management and development holds significant importance. With the objective of exploring correlations between the oral microbiota and severe psoriasis, this study involved 72 severe psoriasis patients and 16 healthy individuals, whose clinical manifestations and living habits were carefully recorded. Cutting-edge techniques such as 16S rRNA gene sequencing and bioinformatics analysis were employed to compare the microbial flora, investigating dynamic changes among severe plaque psoriasis patients, psoriatic arthritis patients and healthy individuals. The findings revealed noteworthy patterns including increased levels of Aggregatibacter in the psoriatic arthritis group, accompanied by a decrease in the level of Prevotella. Moreover, the enrichment o Capnocytandophaga (P = 0.009), Campylobacter (P = 0.0022), and Acetobacter (P = 0.0292) was notably more substantial in the psoriasis group compared to the control group, whereas certain bacterial species such as Bacteroides (P = 0.0049), Muribaculaceae (P = 0.0048) demonstrated decreased enrichment. Additionally, the psoriatic arthritis group exhibited significantly higher levels of Ralstonia, Bifidobacterium and Micromonospora. Based on these findings, it can be inferred that individuals with lower levels of Prevotella and higher levels of Corynebacterium may be more susceptible to psoriasis exacerbation.


Subject(s)
Arthritis, Psoriatic , Microbiota , Psoriasis , RNA, Ribosomal, 16S , Humans , Arthritis, Psoriatic/microbiology , Female , Male , Psoriasis/microbiology , Microbiota/genetics , Adult , Middle Aged , RNA, Ribosomal, 16S/genetics , Mouth/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Nail Diseases/microbiology , Case-Control Studies
4.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125666

ABSTRACT

In recent years, the incidence of metabolic syndrome (MS) has increased due to lifestyle-related factors in developed countries. MS represents a group of conditions that increase the risk of diabetes, cardiovascular diseases, and other severe health problems. Low-grade chronic inflammation is now considered one of the key aspects of MS and could be defined as a new cardiovascular risk factor. Indeed, an increase in visceral adipose tissue, typical of obesity, contributes to the development of an inflammatory state, which, in turn, induces the production of several proinflammatory cytokines responsible for insulin resistance. Psoriasis is a chronic relapsing inflammatory skin disease and is characterized by the increased release of pro-inflammatory cytokines, which can contribute to different pathological conditions within the spectrum of MS. A link between metabolic disorders and Psoriasis has emerged from evidence indicating that weight loss obtained through healthy diets and exercise was able to improve the clinical course and therapeutic response of Psoriasis in patients with obesity or overweight patients and even prevent its occurrence. A key factor in this balance is the gut microbiota; it is an extremely dynamic system, and this makes its manipulation through diet possible via probiotic, prebiotic, and symbiotic compounds. Given this, the gut microbiota represents an additional therapeutic target that can improve metabolism in different clinical conditions.


Subject(s)
Gastrointestinal Microbiome , Inflammation , Metabolic Syndrome , Psoriasis , Psoriasis/microbiology , Psoriasis/metabolism , Psoriasis/complications , Humans , Metabolic Syndrome/microbiology , Metabolic Syndrome/metabolism , Metabolic Syndrome/complications , Inflammation/metabolism , Animals , Obesity/complications , Obesity/microbiology , Obesity/metabolism
5.
Int Immunopharmacol ; 141: 112658, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39137625

ABSTRACT

Atopic dermatitis (AD) and psoriasis are chronic skin diseases with a global impact, posing significant challenges to public health systems and severely affecting patients' quality of life. This review delves into the key role of the gut microbiota in these diseases, emphasizing the importance of the gut-skin axis in inflammatory mediators and immune regulation and revealing a complex bidirectional communication system. We comprehensively assessed the pathogenesis, clinical manifestations, and treatment strategies for AD and psoriasis, with a particular focus on how the gut microbiota and their metabolites influence disease progression via the gut-skin axis. In addition, personalized treatment plans based on individual patient microbiome characteristics have been proposed, offering new perspectives for future treatment approaches. We call for enhanced interdisciplinary cooperation to further explore the interactions between gut microbiota and skin diseases and to assess the potential of drugs and natural products in modulating the gut-skin axis, aiming to advance the treatment of skin diseases.


Subject(s)
Dermatitis, Atopic , Gastrointestinal Microbiome , Psoriasis , Skin , Humans , Dermatitis, Atopic/immunology , Dermatitis, Atopic/microbiology , Psoriasis/immunology , Psoriasis/microbiology , Gastrointestinal Microbiome/immunology , Skin/microbiology , Skin/immunology , Skin/pathology , Animals
6.
Skin Res Technol ; 30(8): e70005, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39167010

ABSTRACT

BACKGROUND: Psoriasis is a prevalent inflammatory skin condition that can be recognized by silvery-white scales on plaques and erythematous papules, despite the fact that psoriasis appears to have multiple causes. Helicobacter pylori (H. pylori) has been investigated recently as a potential infectious etiological component. AIMS: The objective of the study was to evaluate the prevalence of H. pylori infection in psoriatic patients compared to that of healthy controls and determine whether the degree of psoriasis and H. pylori infection were related. PATIENTS AND METHODS: The dermatology, venerology, and andrology department at South Valley University Outpatient Clinic carried out this cross-sectional study. Psoriatic patients of both sexes and ages were included. In addition to the control group, H. Pylori antigen was measured from psoriatic and control groups by using H. pylori stool antigen-enzyme linked immunosorbent assay (HpSA-ELISA), a test for H. pylori stool antigen. More than 20 ng/mL of antigen proved positive, or less than 15 ng/mL proved negative. RESULTS: There was a significant difference between psoriatic patients and control regarding H. pylori infection (p = 0.046): (30.66%) positive in controls, (45.33%) positive in psoriatic patients. Both groups were matched for age (p = 0.908), that is, the mean age of psoriatic patients was 37.44 ± 15.79 years, and the control group was 37.15 ± 15.15 years. Twenty-five psoriatic patients in each group: mild, moderate, and severe psoriasis according to the Psoriasis Area Severity Index (PASI) score. No significant correlation between H. pylori infection and PASI, age, or duration of illness in psoriatic patients. CONCLUSIONS: Patients with psoriasis had greater rates of H. pylori infection but didn't affect the severity of psoriasis.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Psoriasis , Severity of Illness Index , Humans , Helicobacter Infections/epidemiology , Psoriasis/microbiology , Psoriasis/complications , Psoriasis/epidemiology , Female , Male , Cross-Sectional Studies , Adult , Middle Aged , Prevalence , Young Adult
7.
Front Cell Infect Microbiol ; 14: 1430586, 2024.
Article in English | MEDLINE | ID: mdl-39170985

ABSTRACT

Background: Psoriasis is one of the most common autoimmune skin diseases. Increasing evidence shows that alterations in the diversity and function of microbiota can participate in the pathogenesis of psoriasis through various pathways and mechanisms. Objective: To review the connection between microbial changes and psoriasis, how microbial-targeted therapy can be used to treat psoriasis, as well as the potential of prebiotics, probiotics, synbiotics, fecal microbiota transplantation, diet, and Traditional Chinese Medicine as supplementary and adjunctive therapies. Methods: Literature related to the relationship between psoriasis and gut microbiota was searched in PubMed and CNKI. Results: Adjunct therapies such as dietary interventions, traditional Chinese medicine, and probiotics can enhance gut microbiota abundance and diversity in patients with psoriasis. These therapies stimulate immune mediators including IL-23, IL-17, IL-22, and modulate gamma interferon (IFN-γ) along with the NF-kB pathway, thereby suppressing the release of pro-inflammatory cytokines and ameliorating systemic inflammatory conditions. Conclusion: This article discusses the direction of future research and clinical treatment of psoriasis from the perspective of intestinal microbiota and the mechanism of traditional Chinese medicine, so as to provide clinicians with more comprehensive diagnosis and treatment options and bring greater hope to patients with psoriasis.


Subject(s)
Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Medicine, Chinese Traditional , Probiotics , Psoriasis , Psoriasis/therapy , Psoriasis/microbiology , Psoriasis/drug therapy , Humans , Probiotics/therapeutic use , Prebiotics , Cytokines/metabolism , Interleukin-17/metabolism
9.
Arch Dermatol Res ; 316(8): 495, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39073596

ABSTRACT

The scalp microbiome represents an array of microorganisms important in maintaining scalp homeostasis and mediating inflammation. Scalp microbial dysregulation has been implicated in dermatologic conditions including alopecia areata (AA), dandruff/seborrheic dermatitis (D/SD), scalp psoriasis (SP) and folliculitis decalvans (FD). Understanding the impact of scalp microbial dysbiosis gives insight on disease pathophysiology and guides therapeutic decision making. Herein we review the scalp microbiome and its functional role in scalp conditions by analysis of metagenomic medical literature in alopecia, D/SD, SP, and other dermatologic disease.Increased abundance of Malassezia, Staphylococcus, and Brevibacterium was associated with SD compared to healthy controls. A higher proportion of Corynebacterium, actinobacteria, and firmicutes are present in AA patients, and lower proportions of Staphylococcus caprae are associated with worse clinical outcomes. Decreased prevalence of actinobacteria and Propionibacterium and increased firmicutes, staphylococcus, and streptococcus are associated with scalp psoriasis. Studies of central centrifugal cicatricial alopecia (CCCA) suggest scalp microbial composition contributes to CCCA's pro-inflammatory status. The most common organisms associated with FD include methicillin-resistant S. aureus and S. lugdunensis. Antifungals have been a mainstay treatment for these diseases, while other alternatives including coconut oils and shampoos with heat-killed probiotics have shown considerable potential efficacy by replenishing the scalp microbiome.


Subject(s)
Microbiota , Scalp , Humans , Microbiota/drug effects , Microbiota/immunology , Scalp/microbiology , Scalp Dermatoses/microbiology , Scalp Dermatoses/drug therapy , Scalp Dermatoses/therapy , Dysbiosis/microbiology , Dysbiosis/immunology , Folliculitis/microbiology , Folliculitis/diagnosis , Folliculitis/drug therapy , Folliculitis/therapy , Psoriasis/microbiology , Psoriasis/drug therapy , Psoriasis/immunology , Psoriasis/therapy , Dermatitis, Seborrheic/microbiology , Dermatitis, Seborrheic/drug therapy , Dermatitis, Seborrheic/therapy , Alopecia Areata/microbiology , Alopecia Areata/immunology , Alopecia Areata/therapy , Alopecia Areata/drug therapy , Dandruff/microbiology , Dandruff/drug therapy
10.
Skin Res Technol ; 30(7): e13822, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38970783

ABSTRACT

PURPOSE: In present, the diagnosis of psoriasis is mainly based on the patient's typical clinical manifestations, dermoscopy and skin biopsy, and unlike other immune diseases, psoriasis lacks specific indicators in the blood. Therefore, we are required to search novel biomarkers for the diagnosis of psoriasis. METHODS: In this study, we analyzed the composition and the differences of intestinal fungal communities composition between psoriasis patients and healthy individuals in order to find the intestinal fungal communities associated with the diagnosis of psoriasis. We built a machine learning model and identified potential microbial markers for the diagnosis of psoriasis. RESULTS: The results of AUROC (area under ROC) showed that Aspergillus puulaauensis (AUROC = 0.779), Kazachstania africana (AUROC = 0.750) and Torulaspora delbrueckii (AUROC = 0.745) had high predictive ability (AUROC > 0.7) for predicting psoriasis, While Fusarium keratoplasticum (AUROC = 0.670) was relatively lower (AUROC < 0.7). CONCLUSION: The strategy based on the prediction of intestinal fungal communities provides a new idea for the diagnosis of psoriasis and is expected to become an auxiliary diagnostic method for psoriasis.


Subject(s)
Biomarkers , Gastrointestinal Microbiome , Metagenomics , Mycobiome , Psoriasis , Humans , Psoriasis/microbiology , Psoriasis/blood , Female , Adult , Male , Biomarkers/blood , Metagenomics/methods , Middle Aged , Machine Learning , Feces/microbiology , Young Adult , Aspergillus
11.
Int J Mol Sci ; 25(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39062988

ABSTRACT

Biological agents used to treat severe psoriasis may alter the gut microbiota, though current knowledge is limited. This study examines whether switching from TNFα inhibitors, from which patients had reduced or lost effect, to brodalumab, an IL-17 inhibitor, affects the gut microbiota in patients with psoriasis and how these changes correlate with the clinical variables of psoriasis severity and depressive symptoms. Fecal samples from patients were collected before the treatment switch and 12 weeks after the switch and were analyzed for the microbiota composition using next-generation sequencing targeting the V3-V5 region of the 16S rRNA gene, followed by bioinformatics analysis. No significant changes in overall gut microbiota composition were observed after the treatment switch, although individual variations in the Firmicutes/Bacteroidetes ratio were noted, and no significant correlations with clinical variables were found. These findings suggest that short-term changes in gut microbiota in patients with psoriasis are limited and that dysbiosis may be influenced by the interplay of various microbial populations rather than specific taxa. This study provides a foundation for further research into the effects of biological treatments on the gut microbiota in patients with psoriasis.


Subject(s)
Antibodies, Monoclonal, Humanized , Gastrointestinal Microbiome , Psoriasis , Humans , Psoriasis/drug therapy , Psoriasis/microbiology , Gastrointestinal Microbiome/drug effects , Female , Male , Middle Aged , Adult , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , RNA, Ribosomal, 16S/genetics , Feces/microbiology , Aged , Dysbiosis/microbiology , Interleukin-17/metabolism
12.
Sci Rep ; 14(1): 17495, 2024 07 30.
Article in English | MEDLINE | ID: mdl-39079980

ABSTRACT

Short-chain fatty acids (SCFAs) have been proposed to have anti-inflammatory effects and improve immune homeostasis. We aimed to examine the effects of SCFAs on skin phenotype, systemic inflammation, and gut microbiota in mice with psoriasis-like inflammation. Imiquimod (IMQ)-treated C57BL/6 mice served as the study model. We conducted a metagenomic association study of IMQ-mice treated with SCFAs or anti-IL-17 antibody using whole-genome shotgun sequencing. The associations among SCFA supplements, skin thickness, circulating inflammatory profiles, and fecal microbiota profiles were investigated. The microbiome study was performed using pipelines for phylogenetic analysis, functional gene analysis, and pathway analysis. In IMQ-treated mice, there were increases in skin thickness and splenic weight, as well as unique fecal microbial profiles. SCFAs ameliorated IMQ-induced skin thickening, splenic weight gain, and serum IL-17F levels, with results that were comparable with those receiving anti-IL-17 treatment. IMQ-treated mice receiving SCFAs had greater microbial diversity than mice treated with IMQ alone. SCFAs and anti-IL17 treatment were associated with alteration of gut microbiota, with increased prevalences of Oscillospiraceae and Lachnopiraceae and decreased prevalences of Muribaculaceae and Bacteroides, which have been predicted to be associated with increased glycan degradation, phenylalanine metabolism, and xylene degradation. SCFAs may mitigate IMQ-induced skin thickening and IL-17F levels and alter fecal microbiota profiles in IMQ-treated mice.


Subject(s)
Fatty Acids, Volatile , Gastrointestinal Microbiome , Imiquimod , Interleukin-17 , Mice, Inbred C57BL , Skin , Animals , Imiquimod/adverse effects , Gastrointestinal Microbiome/drug effects , Interleukin-17/metabolism , Fatty Acids, Volatile/metabolism , Mice , Skin/drug effects , Skin/pathology , Skin/microbiology , Skin/metabolism , Metagenomics/methods , Psoriasis/drug therapy , Psoriasis/chemically induced , Psoriasis/microbiology , Metagenome , Feces/microbiology
13.
Acta Biomater ; 184: 296-312, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38871203

ABSTRACT

Psoriasis is a chronic skin inflammation influenced by dysregulated skin microbiota, with the role of microbiota in psoriasis gaining increasing prominence. Bacterial extracellular vesicles (bEVs) serve as crucial regulators in the interaction between hosts and microbiota. However, the mechanism underlying the therapeutic potential of bEVs from commensal bacteria in psoriasis remains unclear. Here, we investigated the therapeutic role of Cutibacterium acnes (C. acnes)-derived extracellular vesicles (CA-EVs) in psoriasis treatment. To prolong the active duration of CA-EVs, we encapsulated them in gelatin methacrylate (GelMA) to fabricate hydrogel microspheres (CA-EVs@GHM) with sustained release properties. As GelMA degraded, CA-EVs were gradually released, maintaining a high concentration in mouse skin even 96 h post-treatment. In human keratinocyte cells (HaCaT), CA-EVs@GHM enhanced resistance to Staphylococcus aureus (S. aureus), promoted proliferation and migration of HaCaT cells exposed to S. aureus, and significantly reduced the expression of inflammatory genes such as interleukin (IL)-6 and C-X-C motif chemokine ligand 8 (CXCL8). In vivo, CA-EVs@GHM, more potent than CA-EVs alone, markedly attenuated proinflammatory gene expression, including tumor necrosis factor (TNF), Il6, Il17a, Il22 and Il23a in imiquimod (IMQ)-induced psoriasis-like mice, and restored skin barrier function. 16S rRNA sequencing revealed that CA-EVs@GHM might provide therapeutic effects against psoriasis by restoring microbiota diversity on the back skin of mice, reducing Staphylococcus colonization, and augmenting lipid metabolism. Furthermore, flow cytometry analysis showed that CA-EVs@GHM prevented the conversion of type 2 innate lymphoid cells (ILC2) to type 3 innate lymphoid cells (ILC3) in psoriasis-like mouse skin, reducing the pathogenic ILC3 population and suppressing the secretion of IL-17 and IL-22. In summary, our findings demonstrate that the long-term sustained release of CA-EVs alleviated psoriasis symptoms by controlling the transformation of innate lymphoid cells (ILCs) subgroups and restoring skin microbiota homeostasis, thus offering a promising therapy for psoriasis treatment. STATEMENT OF SIGNIFICANCE: Cutibacterium acnes, which is reduced in psoriasis skin, has been reported to promote skin homeostasis by regulating immune balance. Compared to live bacteria, bacterial extracellular vesicles (bEVs) are less prone to toxicity and safety concerns. bEVs play a pivotal role in maintaining bacterial homeostasis and modulating the immune system. However,bEVs without sustained release materials are unable to function continuously in chronic diseases. Therefore, we utilized hydrogel microspheres to encapsulate Cutibacterium acnes (C. acnes)-derived extracellular vesicles (CA-EVs), enabling long term sustained release. Our findings indicate that, CA-EVs loaded gelatin methacrylate hydrogel microspheres (CA-EVs@GHM) showed superior therapeutic effects in treating psoriasis compared to CA-EVs. CA-EVs@GHM exhibited a more significant regulation of pathological type 3 innate lymphoid cells (ILC3) and skin microbiota, providing a promising approach for microbiota-derived extracellular vesicle therapy in the treatment of skin inflammation.


Subject(s)
Extracellular Vesicles , Hydrogels , Lymphocytes , Microspheres , Psoriasis , Extracellular Vesicles/metabolism , Animals , Humans , Psoriasis/pathology , Psoriasis/immunology , Psoriasis/therapy , Psoriasis/microbiology , Hydrogels/chemistry , Hydrogels/pharmacology , Mice , Lymphocytes/immunology , Lymphocytes/metabolism , Immunity, Innate/drug effects , Staphylococcus aureus , HaCaT Cells
14.
Arch Dermatol Res ; 316(7): 374, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38850443

ABSTRACT

The microbiome is intricately linked to the development of psoriasis, serving as both a potential cause and consequence of the psoriatic process. In recent years, there has been growing interest among psoriasis researchers in exploring how psoriasis treatments affect the skin and gut microbiome. However, a comprehensive evaluation of the impact of modern treatment approaches on the microbiome has yet to be conducted. In this systematic review, we analyze studies investigating alterations in the skin and gut microbiome resulting from psoriasis treatment, aiming to understand how current therapies influence the role of the microbiome in psoriasis development. The systematic review was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. PubMed and Scopus databases were searched for eligible studies from the inception dates until July 5, 2023. Study selection, data extraction, and risk of bias assessment were carried out by three overlapping pairs of reviewers, resolving any disagreements through consensus. Our analysis of various treatments, including biologics, conventional medications, phototherapy, and probiotics, reveals significant shifts in microbial diversity and abundance. Importantly, favorable treatment outcomes are associated with microbiota alterations that approach those observed in healthy individuals. While the studies reviewed exhibit varying degrees of bias, underscoring the need for further research, this review supports the potential of microbiome modulation as both a preventive and therapeutic strategy for psoriasis patients. The findings underscore the importance of personalized therapeutic approaches, recognizing the profound impact of treatment on the microbiome. They also highlight the promise of probiotics, prebiotics, and dietary interventions in psoriasis management.


Subject(s)
Gastrointestinal Microbiome , Probiotics , Psoriasis , Skin , Psoriasis/microbiology , Psoriasis/immunology , Psoriasis/therapy , Humans , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Skin/microbiology , Probiotics/administration & dosage , Phototherapy/methods , Biological Products/therapeutic use , Treatment Outcome , Dermatologic Agents/therapeutic use , Dermatologic Agents/administration & dosage
15.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38928354

ABSTRACT

Psoriasis is an inflammatory dermatosis with a complex pathogenesis, significantly impacting the quality of life of patients. The role of oxidative stress and gut microbiota in the pathogenesis of this disease is increasingly studied, appearing to underlie the comorbidities associated with this condition. We present the first prospective observational study conducted in Romania evaluating the interrelationship between gut microbiota and hematological, inflammatory, biochemical, and oxidative stress parameters in treatment-naïve psoriasis patients. Significant differences were observed in terms of microbiota composition, with lower levels of Firmicutes and Enterobacteriaceae in the psoriasis group compared to the control group. Moreover, a negative correlation was found between the serum triglyceride levels in patients with psoriasis and the Enterobacteriaceae family (p = 0.018, r = -0.722), and a positive correlation was found between the serum glucose levels and the Firmicutes/Bacteroides ratio (p = 0.03, r = 0.682). Regarding the oxidant-antioxidant status, a significant correlation was found between the FORT level and Lactobacillus (p = 0.034, r = 0.669). Lastly, the Firmicutes level negatively correlated with the DLQI level, independent of the clinical severity of the disease (p = 0.02, r = -0.685). In conclusion, even though the number of included patients is small, these results may serve as a starting point for future research into the involvement of the microbiota-inflammation-oxidative stress axis in psoriasis development.


Subject(s)
Gastrointestinal Microbiome , Oxidative Stress , Psoriasis , Humans , Psoriasis/microbiology , Psoriasis/blood , Psoriasis/metabolism , Female , Male , Adult , Middle Aged , Inflammation , Prospective Studies
16.
EBioMedicine ; 105: 105222, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38924840

ABSTRACT

BACKGROUND: Psoriasis (Pso) is a chronic inflammatory skin disease that poses both physical and psychological challenges. Dysbiosis of the skin microbiome has been implicated in Pso, yet a comprehensive multi-omics analysis of host-microbe interactions is still lacking. To bridge this gap, we conducted an exploratory study by adopting the integrated approach that combines whole metagenomic shotgun sequencing with skin transcriptomics. METHODS: This was a cross-sectional study, adult patients with plaque-type Psoriasis (Pso) and healthy volunteers were included. Skin microbiota samples and biopsies were collected from both lesional and non-lesional skin areas on the lower back. Weighted Gene Correlation Network Analysis (WGCNA) was employed for co-expression network analysis, and cell deconvolution was conducted to estimate cell fractions. Taxonomic and functional features of the microbiome were identified using whole metagenomic shotgun sequencing. Association between host genes and microbes was analyzed using Spearman correlation. FINDINGS: Host anti-viral responses and interferon-related networks were identified and correlated with the severity of psoriasis. The skin microbiome showed a greater prevalence of Corynebacterium simulans in the PASI severe-moderate groups, which correlated with interferon-induced host genes. Two distinct psoriatic clusters with varying disease severities were identified. Variations in the expression of cell apoptosis-associated antimicrobial peptides (AMPs) and microbial aerobic respiration I pathway may partly account for these differences in disease severity. INTERPRETATION: Our multi-omics analysis revealed for the first time anti-viral responses and the presence of C. simulans associated with psoriasis severity. It also identified two psoriatic subtypes with distinct AMP and metabolic pathway expression. Our study provides new insights into understanding the host-microbe interaction in psoriasis and lays the groundwork for developing subtype-specific strategies for managing this chronic skin disease. FUNDING: The research has received funding from the FP7 (MAARS-Grant 261366) and the Innovative Medicines Initiative 2 Joint Undertaking (JU) under grant agreement No 821511 (BIOMAP). The JU receives support from the European Union's Horizon 2020 research and innovation programme and EFPIA. This publication reflects only the author's view and the JU is not responsible for any use that may be made of the information it contains. GAM was supported by a scholarship provided by CAPES-PRINT, financed by the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior - CAPES (Brazilian Government Agency). The authors thank all patients who participated in our study.


Subject(s)
Host Microbial Interactions , Metagenomics , Microbiota , Psoriasis , Severity of Illness Index , Skin , Humans , Psoriasis/microbiology , Psoriasis/genetics , Psoriasis/metabolism , Metagenomics/methods , Skin/microbiology , Skin/metabolism , Skin/pathology , Female , Male , Adult , Host Microbial Interactions/genetics , Middle Aged , Cross-Sectional Studies , Metagenome , Gene Expression Profiling , Transcriptome , Gene Regulatory Networks , Host-Pathogen Interactions/genetics , Computational Biology/methods , Multiomics
17.
Acta Derm Venereol ; 104: adv34892, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898675

ABSTRACT

Psoriasis results from both genetic predisposition and environmental triggers, such as Streptococcal infections. This study aimed to explore the correlation between the abundance of the Streptococcus genus on the skin and psoriasis severity in individuals carrying specific psoriasis-associated genetic variants. Studying 39 chronic plaque psoriasis patients, the elbow skin microbiome and 49 psoriasis-related single nucleotide polymorphisms (SNPs) were analysed using a MiSeq instrument for 16S rDNA sequencing, and CLC Genomic Workbench for processing and analysis. Through multivariate linear regression analysis, a positive correlation was found between Streptococcus genus abundance and psoriasis severity in patients with certain FBXL19 gene-related heterozygous SNPs (rs12924903, rs10782001, rs12445568). Conversely, a negative association was observed in patients with homozygous genotypes. Moreover, we identified an association between Streptococcus abundance and psoriasis severity in patients with genetic variants related to IL-22, ERAP1, NOS2, and ILF3. This is the first study highlighting a positive association between Streptococcus skin colonization and psoriasis severity in patients with heterozygous genotypes within the FBXL19 gene region. FXBL19 targets the IL-33/IL1RL1 axis, crucial in infectious diseases and innate immunity promotion. These novel results suggests an intricate interaction among host genetics, Streptococcus skin colonization, and psoriasis inflammation, offering potential avenues for novel treatment approaches.


Subject(s)
F-Box Proteins , Polymorphism, Single Nucleotide , Psoriasis , Severity of Illness Index , Skin , Streptococcus , Humans , Male , Psoriasis/genetics , Psoriasis/microbiology , Female , Middle Aged , Adult , Skin/microbiology , Streptococcus/genetics , Streptococcus/isolation & purification , F-Box Proteins/genetics , Genetic Predisposition to Disease , Phenotype , Heterozygote , Host-Pathogen Interactions , Homozygote , Ribotyping , Aged
18.
Science ; 384(6699): 1023-1030, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38815037

ABSTRACT

Seamless interfaces between electronic devices and biological tissues stand to revolutionize disease diagnosis and treatment. However, biological and biomechanical disparities between synthetic materials and living tissues present challenges at bioelectrical signal transduction interfaces. We introduce the active biointegrated living electronics (ABLE) platform, encompassing capabilities across the biogenic, biomechanical, and bioelectrical properties simultaneously. The living biointerface, comprising a bioelectronics layout and a Staphylococcus epidermidis-laden hydrogel composite, enables multimodal signal transduction at the microbial-mammalian nexus. The extracellular components of the living hydrogels, prepared through thermal release of naturally occurring amylose polymer chains, are viscoelastic, capable of sustaining the bacteria with high viability. Through electrophysiological recordings and wireless probing of skin electrical impedance, body temperature, and humidity, ABLE monitors microbial-driven intervention in psoriasis.


Subject(s)
Hydrogels , Psoriasis , Skin , Staphylococcus epidermidis , Animals , Humans , Mice , Body Temperature , Electric Impedance , Electronics , Humidity , Hydrogels/chemistry , Inflammation/microbiology , Inflammation/therapy , Skin/microbiology , Wearable Electronic Devices , Wireless Technology , Psoriasis/microbiology , Psoriasis/therapy , Mice, Knockout , Toll-Like Receptor 2/genetics
19.
Australas J Dermatol ; 65(4): 319-327, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38419189

ABSTRACT

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


Subject(s)
Acne Vulgaris , Dermatitis, Atopic , Gastrointestinal Microbiome , Mendelian Randomization Analysis , Psoriasis , Humans , Gastrointestinal Microbiome/genetics , Psoriasis/microbiology , Psoriasis/genetics , Acne Vulgaris/microbiology , Dermatitis, Atopic/microbiology , Urticaria/microbiology , Genome-Wide Association Study
20.
Cells ; 12(22)2023 11 14.
Article in English | MEDLINE | ID: mdl-37998359

ABSTRACT

The skin presents a multifaceted microbiome, a balanced coexistence of bacteria, fungi, and viruses. These resident microorganisms are fundamental in upholding skin health by both countering detrimental pathogens and working in tandem with the skin's immunity. Disruptions in this balance, known as dysbiosis, can lead to disorders like psoriasis and atopic dermatitis. Central to the skin's defense system are mast cells. These are strategically positioned within the skin layers, primed for rapid response to any potential foreign threats. Recent investigations have started to unravel the complex interplay between these mast cells and the diverse entities within the skin's microbiome. This relationship, especially during times of both balance and imbalance, is proving to be more integral to skin health than previously recognized. In this review, we illuminate the latest findings on the ties between mast cells and commensal skin microorganisms, shedding light on their combined effects on skin health and maladies.


Subject(s)
Dermatitis, Atopic , Microbiota , Psoriasis , Humans , Mast Cells , Skin , Psoriasis/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL