1
|
Radaschin DS, Tatu A, Iancu AV, Beiu C, Popa LG. The Contribution of the Skin Microbiome to Psoriasis Pathogenesis and Its Implications for Therapeutic Strategies. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1619. [PMID: 39459406 PMCID: PMC11509136 DOI: 10.3390/medicina60101619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024]
Abstract
Psoriasis is a common chronic inflammatory skin disease, associated with significant morbidity and a considerable negative impact on the patients' quality of life. The complex pathogenesis of psoriasis is still incompletely understood. Genetic predisposition, environmental factors like smoking, alcohol consumption, psychological stress, consumption of certain drugs, and mechanical trauma, as well as specific immune dysfunctions, contribute to the onset of the disease. Mounting evidence indicate that skin dysbiosis plays a significant role in the development and exacerbation of psoriasis through loss of immune tolerance to commensal skin flora, an altered balance between Tregs and effector cells, and an excessive Th1 and Th17 polarization. While the implications of skin dysbiosis in psoriasis pathogenesis are only starting to be revealed, the progress in the characterization of the skin microbiome changes in psoriasis patients has opened a whole new avenue of research focusing on the modulation of the skin microbiome as an adjuvant treatment for psoriasis and as part of a long-term plan to prevent disease flares. The skin microbiome may also represent a valuable predictive marker of treatment response and may aid in the selection of the optimal personalized treatment. We present the current knowledge on the skin microbiome changes in psoriasis and the results of the studies that investigated the efficacy of the different skin microbiome modulation strategies in the management of psoriasis, and discuss the complex interaction between the host and skin commensal flora.
Collapse
Affiliation(s)
- Diana Sabina Radaschin
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Saint Parascheva” Infectious Disease Clinical Hospital, Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| | - Alin Tatu
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Saint Parascheva” Infectious Disease Clinical Hospital, Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| | - Alina Viorica Iancu
- Department of Morphological and Functional Sciences, “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| | - Cristina Beiu
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Liliana Gabriela Popa
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
2
|
Radaschin DS, Iancu AV, Ionescu AM, Gurau G, Niculet E, Bujoreanu FC, Beiu C, Tatu AL, Popa LG. Comparative Analysis of the Cutaneous Microbiome in Psoriasis Patients and Healthy Individuals-Insights into Microbial Dysbiosis: Final Results. Int J Mol Sci 2024; 25:10583. [PMID: 39408916 PMCID: PMC11477231 DOI: 10.3390/ijms251910583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Psoriasis is one of the most frequent chronic inflammatory skin diseases and exerts a significant psychological impact, causing stigmatization, low self-esteem and depression. The pathogenesis of psoriasis is remarkably complex, involving genetic, immune and environmental factors, some of which are still incompletely explored. The cutaneous microbiome has become more and more important in the pathogenesis of inflammatory skin diseases such as acne, rosacea, atopic dermatitis and psoriasis. Dysbiosis of the skin microbiome could be linked to acute flare ups in psoriatic disease, as recent studies suggest. Given this hypothesis, we conducted a study in which we evaluated the cutaneous microbiome of psoriasis patients and healthy individuals. In our study, we collected multiple samples using swab sampling, adhesive tape and punch biopsies. Our results are similar to other studies in which the qualitative and quantitative changes found in the cutaneous microbiome of psoriasis patients are different than healthy individuals. Larger, standardized studies are needed in order to elucidate the microbiome changes in psoriasis patients, clarify their role in the pathogenesis of psoriasis, decipher the interactions between the commensal microorganisms of the same and different niches and between microbiomes and the host and identify new therapeutic strategies.
Collapse
Affiliation(s)
- Diana Sabina Radaschin
- Department of Dermatology, “Saint Parascheva” Infectious Disease Clinical Hospital, 800179 Galati, Romania; (D.S.R.); (F.C.B.)
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800008 Galati, Romania
- Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University, 800008 Galati, Romania
| | - Alina Viorica Iancu
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800008 Galati, Romania; (A.V.I.); (G.G.); (E.N.)
| | | | - Gabriela Gurau
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800008 Galati, Romania; (A.V.I.); (G.G.); (E.N.)
| | - Elena Niculet
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800008 Galati, Romania; (A.V.I.); (G.G.); (E.N.)
| | - Florin Ciprian Bujoreanu
- Department of Dermatology, “Saint Parascheva” Infectious Disease Clinical Hospital, 800179 Galati, Romania; (D.S.R.); (F.C.B.)
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800008 Galati, Romania
- Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University, 800008 Galati, Romania
| | - Cristina Beiu
- Dermatology Department, Carol Davila University of Medicine and Pharmacy, 030167 Bucharest, Romania;
| | - Alin Laurentiu Tatu
- Department of Dermatology, “Saint Parascheva” Infectious Disease Clinical Hospital, 800179 Galati, Romania; (D.S.R.); (F.C.B.)
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800008 Galati, Romania
- Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University, 800008 Galati, Romania
| | - Liliana Gabriela Popa
- Dermatology Department, Carol Davila University of Medicine and Pharmacy, 030167 Bucharest, Romania;
| |
Collapse
|
3
|
Kline SN, Saito Y, Archer NK. Staphylococcus aureus Proteases: Orchestrators of Skin Inflammation. DNA Cell Biol 2024; 43:483-491. [PMID: 38957987 PMCID: PMC11535466 DOI: 10.1089/dna.2024.0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
Skin homeostasis relies on a delicate balance between host proteases and protease inhibitors along with those secreted from microbial communities, as disruption to this harmony contributes to the pathogenesis of inflammatory skin disorders, including atopic dermatitis and Netherton's syndrome. In addition to being a prominent cause of skin and soft tissue infections, the gram-positive bacterium Staphylococcus aureus is a key player in inflammatory skin conditions due to its array of 10 secreted proteases. Herein we review how S. aureus proteases augment the development of inflammation in skin disorders. These mechanisms include degradation of skin barrier integrity, immune dysregulation and pruritis, and impairment of host defenses. Delineating the diverse roles of S. aureus proteases has the potential to reveal novel therapeutic strategies, such as inhibitors of proteases or their cognate target, as well as neutralizing vaccines to alleviate the burden of inflammatory skin disorders in patients.
Collapse
Affiliation(s)
- Sabrina N. Kline
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Yoshine Saito
- School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Nathan K. Archer
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Chen G, Chen X, Duan X, Zhang R, Bai C. Unraveling the roles of IFIT3 gene and immune-metabolic pathways in psoriasis: a bioinformatics exploration for diagnostic markers and therapeutic targets. Front Mol Biosci 2024; 11:1439837. [PMID: 39239353 PMCID: PMC11374644 DOI: 10.3389/fmolb.2024.1439837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Background The functions and related signal pathways of the IFIT3 gene in the skin lesions of patients with psoriasis were explored through bioinformatics methods to determine the potential specific molecular markers of psoriasis. Methods The "limma" R package was used to analyze three datasets from the Gene Expression Omnibus database (GSE13355, GSE30999 and GSE106992), and the differential genes were screened. The STRING database was used for gene ontology (GO) enrichment analysis, Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis, and protein-protein interaction network integration. Then, the IFIT3 subnetwork was extracted and analyzed by gene set enrichment analysis (GSEA) using the Metascape database to verify the effectiveness of gene differentiation and disease tissue identification. Results In this study, 426 differential genes were obtained, of which 322 were significantly upregulated and 104 were significantly downregulated. GO enrichment analysis showed that the differential genes were mainly involved in immunity and metabolism; the KEGG pathway enrichment analysis mainly included the chemokine signal pathway, PPAR signal pathway, and IL-17 signal pathway, among others. Based on the IFIT3 subnetwork analysis, it was found that IFIT3 was mainly involved in the biological processes of viruses, bacteria, and other microorganisms. The pathways obtained by GSEA were mainly related to immunity, metabolism, and antiviral activities. IFIT3 was highly expressed in psoriatic lesions and may thus be helpful in the diagnosis of psoriasis. Conclusion The differential genes, biological processes, and signal pathways of psoriasis, especially information related to and diagnostic efficiency of the IFIT3 gene, were obtained by bioinformatics analysis. These results are expected to provide the theoretical basis and new directions for exploring the pathogenesis of psoriasis, in addition to helping with finding diagnostic markers and developing drug treatment targets.
Collapse
Affiliation(s)
- Guangshan Chen
- Department of Dermatology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xi Chen
- Department of Dermatology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xingwu Duan
- Department of Dermatology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Runtian Zhang
- Department of Dermatology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Chunxiao Bai
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
5
|
Xu Y, Gan Y, Qi F, Lu X, Zhang X, Zhang J, Wang H, Li Y, Zhou Z, Wang X, Zeng D, Lu F, Zhang C, Cheng B, Hu Z, Wang G. Innate lymphoid cell-based immunomodulatory hydrogel microspheres containing Cutibacterium acnes extracellular vesicles for the treatment of psoriasis. Acta Biomater 2024; 184:296-312. [PMID: 38871203 DOI: 10.1016/j.actbio.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
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.
Collapse
Affiliation(s)
- Yujie Xu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuyang Gan
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fangfang Qi
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xinyu Lu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaofei Zhang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiarui Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hailin Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yue Li
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhiyang Zhou
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xusheng Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Feng Lu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chunhua Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Biao Cheng
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command, PLA, Guangzhou 510010, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Gaofeng Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA.
| |
Collapse
|
6
|
Rademacher F, Scheel A, Gläser R, Schröder L, Heinemann N, Bartels J, Gerdes S, Stölzl D, Rodriguez E, Döhner K, Weidinger S, Werfel T, Harder J. Inhibition of RNase 7 by RNase inhibitor promotes inflammation and Staphylococcus aureus growth: Implications for atopic dermatitis. Allergy 2024; 79:1573-1583. [PMID: 38641894 DOI: 10.1111/all.16125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/14/2024] [Accepted: 03/07/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND The antimicrobial ribonuclease RNase 7 is abundantly expressed in the epidermis of lesional skin of atopic dermatitis (AD). Host RNase inhibitor (RI) binds to RNase 7 and blocks its ribonuclease activity. This study aimed to evaluate the impact of RNase 7-RI interactions on AD. METHODS Cultured human primary keratinocytes, with siRNA-mediated downregulation of RNase 7 and RI, were stimulated with the synthetic RNA polyinosinic-polycytidylic acid (poly I:C). Induction of proinflammatory mediators was analyzed by real-time PCR and ELISA. RI expression in AD non-lesional and lesional skin biopsies and healthy controls was analyzed by real-time PCR and immunostaining. RI protein release in vivo on the AD skin surface was determined by western blot. Antimicrobial and ribonuclease assays were used to investigate the functional role of RI. RESULTS RNase 7 inhibited the RNA-induced expression of proinflammatory mediators in keratinocytes. Accordingly, downregulation of RNase 7 in keratinocytes enhanced RNA-mediated induction of proinflammatory mediators, whereas downregulation of RI had the opposite effect. RI was released by damaged keratinocytes and epidermis. In vivo expression and release of RI on the skin surface were enhanced in lesional AD skin. Rinsing solution from the surface of lesional AD skin blocked the ribonuclease activity of RNase 7. The anti-Staphylococcus aureus activity of RNase 7 was abrogated by RI. CONCLUSIONS Our data suggest a novel role of RI as a trigger factor of inflammation in AD by blocking the ribonuclease and antimicrobial activity of RNase 7, thereby enhancing RNA-mediated inflammation and S. aureus growth.
Collapse
Affiliation(s)
- Franziska Rademacher
- Department of Dermatology, Quincke Research Center, Kiel University, Kiel, Germany
| | - Annika Scheel
- Department of Dermatology, Quincke Research Center, Kiel University, Kiel, Germany
| | - Regine Gläser
- Department of Dermatology, Quincke Research Center, Kiel University, Kiel, Germany
| | - Lena Schröder
- Department of Dermatology, Quincke Research Center, Kiel University, Kiel, Germany
| | - Nina Heinemann
- Department of Dermatology, Quincke Research Center, Kiel University, Kiel, Germany
| | - Joachim Bartels
- Department of Dermatology, Quincke Research Center, Kiel University, Kiel, Germany
| | - Sascha Gerdes
- Department of Dermatology, Quincke Research Center, Kiel University, Kiel, Germany
| | - Dora Stölzl
- Department of Dermatology, Quincke Research Center, Kiel University, Kiel, Germany
| | - Elke Rodriguez
- Department of Dermatology, Quincke Research Center, Kiel University, Kiel, Germany
| | - Katinka Döhner
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany
| | - Stephan Weidinger
- Department of Dermatology, Quincke Research Center, Kiel University, Kiel, Germany
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany
| | - Jürgen Harder
- Department of Dermatology, Quincke Research Center, Kiel University, Kiel, Germany
| |
Collapse
|
7
|
Chen Y, Peng C, Zhu L, Wang J, Cao Q, Chen X, Li J. Atopic Dermatitis and Psoriasis: Similarities and Differences in Metabolism and Microbiome. Clin Rev Allergy Immunol 2024; 66:294-315. [PMID: 38954264 DOI: 10.1007/s12016-024-08995-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2024] [Indexed: 07/04/2024]
Abstract
Atopic dermatitis and psoriasis are common chronic inflammatory diseases of high incidence that share some clinical features, including symptoms of pruritus and pain, scaly lesions, and histologically, acanthosis and hyperkeratosis. Meanwhile, they are both commonly comorbid with metabolic disorders such as obesity and diabetes, indicating that both diseases may exist with significant metabolic disturbances. Metabolomics reveals that both atopic dermatitis and psoriasis have abnormalities in a variety of metabolites, including lipids, amino acids, and glucose. Meanwhile, recent studies have highlighted the importance of the microbiome and its metabolites in the pathogenesis of atopic dermatitis and psoriasis. Metabolic alterations and microbiome dysbiosis can also affect the immune, inflammatory, and epidermal barrier, thereby influencing the development of atopic dermatitis and psoriasis. Focusing on the metabolic and microbiome levels, this review is devoted to elaborating the similarities and differences between atopic dermatitis and psoriasis, thus providing insights into the intricate relationship between both conditions.
Collapse
Affiliation(s)
- Yihui Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Furong Laboratory, Changsha, 410008, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Furong Laboratory, Changsha, 410008, China
| | - Lei Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Furong Laboratory, Changsha, 410008, China
| | - Jiayi Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Furong Laboratory, Changsha, 410008, China
| | - Qiaozhi Cao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Furong Laboratory, Changsha, 410008, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Furong Laboratory, Changsha, 410008, China.
| | - Jie Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Furong Laboratory, Changsha, 410008, China.
| |
Collapse
|
8
|
Radaschin DS, Iancu AV, Ionescu AM, Gurau G, Niculet E, Bujoreanu FC, Nastase F, Radaschin T, Popa LG, Axente RE, Tatu AL. An Eastern County from an European Eastern Country-The Characteristics of Cutaneous Microbiome in Psoriasis Patients-Preliminary Results. Life (Basel) 2024; 14:678. [PMID: 38929663 PMCID: PMC11205136 DOI: 10.3390/life14060678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/18/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
The cutaneous microbiome represents a topic of high interest nowadays. Multiple studies have suggested the importance of the skin microbiome in different dermatological pathologies, highlighting the possible implications of cutaneous microorganisms in either the pathogenesis or prognosis of skin maladies. Psoriasis represents a common inflammatory skin disease, with a high prevalence in the worldwide population. The role of the cutaneous microbiome in psoriasis could explain a number of pathogenic theories and treatment objectives of this incurable skin disease. Our interest in the characteristics of the cutaneous microbiome, especially in psoriatic patients who attended a tertiary dermatological centre in Galati, Romania, is reflected in our current study, of which the preliminary results are discussed in this article. Using three types of skin sampling techniques (swabs, adhesive tape, and punch biopsies), we tried to characterise the microorganisms harboured in the skin of psoriatic patients and healthy individuals. This study was performed using culture-based probes, which were analysed using MALDI-TOF mass spectrometer equipment. Our preliminary results suggested that the greatest diversity was observed in the perilesional areas of psoriatic patients. The lowest cutaneous diversity was obtained from sampling psoriatic plaques. These results are similar to other studies of the cutaneous microbiome in psoriasis. The most frequent microorganisms found in all groups studied were of the Staphylococcus species: Staphylococcus epidermidis, Staphylococcus hominis, and Staphylococcus aureus. Analysing the living environment of each individual from this study, our preliminary results suggested different results from other studies, as higher diversity and heterogenicity was observed in urban environments than in rural living areas. Regarding the differences between sexes, our preliminary results showed higher quantitative and qualitative changes in the skin microbiome of male participants than female participants, opposite to the results found in other studies of the cutaneous microbiome in psoriasis. Given these preliminary results, we can conclude that we have found important differences by studying the cutaneous microbiome of psoriatic patients and healthy control individuals from a population that, to our knowledge, has not been yet studied from this point of view. Our results showed important characteristics of the skin microbiome in an Eastern European population, where cultural and environmental living habits could influence the cutaneous microbiome.
Collapse
Affiliation(s)
- Diana Sabina Radaschin
- Department of Dermatology, “Saint Parascheva” Infectious Disease Clinical Hospital, 800179 Galati, Romania; (D.S.R.); (A.L.T.)
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800385 Galati, Romania
- Multidisciplinary Integrated Centre of Dermatological Interface Research (MICDIR), “Dunarea de Jos” University of Galati, 800385 Galati, Romania
| | - Alina Viorica Iancu
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800385 Galati, Romania; (A.V.I.)
| | | | - Gabriela Gurau
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800385 Galati, Romania; (A.V.I.)
| | - Elena Niculet
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800385 Galati, Romania; (A.V.I.)
| | - Florin Ciprian Bujoreanu
- Department of Dermatology, “Saint Parascheva” Infectious Disease Clinical Hospital, 800179 Galati, Romania; (D.S.R.); (A.L.T.)
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800385 Galati, Romania
- Multidisciplinary Integrated Centre of Dermatological Interface Research (MICDIR), “Dunarea de Jos” University of Galati, 800385 Galati, Romania
| | - Florentina Nastase
- Department of Neuropsychomotor Rehabilitation, “Sf. Ioan” Clinical Hospital for Children, 800487 Galati, Romania
| | - Teodora Radaschin
- Radiology Department, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Liliana Gabriela Popa
- Dermatology Department, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Roxana Elena Axente
- Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800385 Galati, Romania;
| | - Alin Laurentiu Tatu
- Department of Dermatology, “Saint Parascheva” Infectious Disease Clinical Hospital, 800179 Galati, Romania; (D.S.R.); (A.L.T.)
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800385 Galati, Romania
- Multidisciplinary Integrated Centre of Dermatological Interface Research (MICDIR), “Dunarea de Jos” University of Galati, 800385 Galati, Romania
| |
Collapse
|
9
|
Liu S, He M, Jiang J, Duan X, Chai B, Zhang J, Tao Q, Chen H. Triggers for the onset and recurrence of psoriasis: a review and update. Cell Commun Signal 2024; 22:108. [PMID: 38347543 PMCID: PMC10860266 DOI: 10.1186/s12964-023-01381-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/02/2023] [Indexed: 02/15/2024] Open
Abstract
Psoriasis is an immune-mediated inflammatory skin disease, involving a complex interplay between genetic and environmental factors. Previous studies have demonstrated that genetic factors play a major role in the pathogenesis of psoriasis. However, non-genetic factors are also necessary to trigger the onset and recurrence of psoriasis in genetically predisposed individuals, which include infections, microbiota dysbiosis of the skin and gut, dysregulated lipid metabolism, dysregulated sex hormones, and mental illness. Psoriasis can also be induced by other environmental triggers, such as skin trauma, unhealthy lifestyles, and medications. Understanding how these triggers play a role in the onset and recurrence of psoriasis provides insights into psoriasis pathogenesis, as well as better clinical administration. In this review, we summarize the triggers for the onset and recurrence of psoriasis and update the current evidence on the underlying mechanism of how these factors elicit the disease. Video Abstract.
Collapse
Grants
- No.82173423, No.81974475, No.82103731 the National Natural Science Foundation of China
- No.82173423, No.81974475, No.82103731 the National Natural Science Foundation of China
- No.82173423, No.81974475, No.82103731 the National Natural Science Foundation of China
- Basic Research Project, No. JCYJ20190809103805589 Shenzhen Natural Science Foundation
- Basic Research Project, No. JCYJ20190809103805589 Shenzhen Natural Science Foundation
- Basic Research Project, No. JCYJ20190809103805589 Shenzhen Natural Science Foundation
- Key Project, No.2019003 Shenzhen Nanshan District Science and Technology Project
- Key Project, No.2019003 Shenzhen Nanshan District Science and Technology Project
- Key Project, No.2019003 Shenzhen Nanshan District Science and Technology Project
Collapse
Affiliation(s)
- Suwen Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Mengwen He
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Jian Jiang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoru Duan
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bao Chai
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
- Department of Dermatology, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Jingyu Zhang
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
- Department of Dermatology, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Qingxiao Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongxiang Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China.
| |
Collapse
|
10
|
Almenara-Blasco M, Gracia-Cazaña T, Poblador-Plou B, Laguna-Berna C, Carmona-Pírez J, Navarro-Bielsa A, Prados-Torres A, Gimeno-Miguel A, Gilaberte Y. Multimorbidity of Psoriasis: A Large-Scale Population Study of Its Associated Comorbidities. J Clin Med 2024; 13:492. [PMID: 38256625 PMCID: PMC10816233 DOI: 10.3390/jcm13020492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/06/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
INTRODUCTION Psoriasis is a chronic disease of the skin with a prevalence of 2% in the general population. The high prevalence of psoriasis has prompted the study of its comorbidities in recent decades. We designed a study to determine the prevalence of psoriasis in a large-scale, population-based cohort, to exhaustively describe its comorbidities, and to analyze which diseases are associated with psoriasis. METHODS Retrospective, observational study based on the clinical information contained in the electronic health records of the individuals in the EpiChron Cohort with a diagnosis of psoriasis (31,178 individuals) in 2019. We used logistic regression models and calculated the likelihood of the occurrence of each comorbidity based on the presence of psoriasis (p-value < 0.05). RESULTS The prevalence of psoriasis was 2.84%, and it was more prevalent in men (3.31% vs. 2.43%). The most frequent chronic comorbidities were disorders of lipid metabolism (35.87%), hypertension (35.50%), and other nutritional-endocrine-metabolic disorders (21.79%). The conditions most associated with psoriasis were (odds ratio; 95% confidence interval) tuberculosis (2.36; 1.24-4.49), cystic fibrosis (2.15; 1.25-3.69), amongst others. We did not find a significant association between psoriasis and hypertension or neoplasms (0.90; 0.86-0.95). CONCLUSIONS This study revealed significant associations between psoriasis and cardiac, psychological, and musculoskeletal comorbidities.
Collapse
Affiliation(s)
- Manuel Almenara-Blasco
- Department of Dermatology, Miguel Servet University Hospital, IIS Aragon, Paseo Isabel la Católica 1-3, 50009 Zaragoza, Spain; (M.A.-B.)
| | - Tamara Gracia-Cazaña
- Department of Dermatology, Miguel Servet University Hospital, IIS Aragon, Paseo Isabel la Católica 1-3, 50009 Zaragoza, Spain; (M.A.-B.)
| | - Beatriz Poblador-Plou
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), Aragon Health Research Institute (IIS Aragón), Miguel Servet University Hospital, 50009 Zaragoza, Spain
- Research Network on Chronicity, Primary Care, and Health Promotion (RICAPPS), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Clara Laguna-Berna
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), Aragon Health Research Institute (IIS Aragón), Miguel Servet University Hospital, 50009 Zaragoza, Spain
- Research Network on Chronicity, Primary Care, and Health Promotion (RICAPPS), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Jonás Carmona-Pírez
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), Aragon Health Research Institute (IIS Aragón), Miguel Servet University Hospital, 50009 Zaragoza, Spain
- Research Network on Chronicity, Primary Care, and Health Promotion (RICAPPS), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- Subdirección Técnica Asesora de Gestión de la Información, Andalusian Health Service, 41071 Sevilla, Spain
| | - Alba Navarro-Bielsa
- Department of Dermatology, Miguel Servet University Hospital, IIS Aragon, Paseo Isabel la Católica 1-3, 50009 Zaragoza, Spain; (M.A.-B.)
| | - Alexandra Prados-Torres
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), Aragon Health Research Institute (IIS Aragón), Miguel Servet University Hospital, 50009 Zaragoza, Spain
- Research Network on Chronicity, Primary Care, and Health Promotion (RICAPPS), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Antonio Gimeno-Miguel
- EpiChron Research Group, Aragon Health Sciences Institute (IACS), Aragon Health Research Institute (IIS Aragón), Miguel Servet University Hospital, 50009 Zaragoza, Spain
- Research Network on Chronicity, Primary Care, and Health Promotion (RICAPPS), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Yolanda Gilaberte
- Department of Dermatology, Miguel Servet University Hospital, IIS Aragon, Paseo Isabel la Católica 1-3, 50009 Zaragoza, Spain; (M.A.-B.)
| |
Collapse
|
11
|
Jia Y, Guan Z, Liu C, Huang M, Li J, Feng J, Shen B, Yang G. Staphylococcus aureus β-hemolysin causes skin inflammation by acting as an agonist of epidermal growth factor receptor. Microbiol Spectr 2024; 12:e0222723. [PMID: 38059627 PMCID: PMC10783061 DOI: 10.1128/spectrum.02227-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 11/08/2023] [Indexed: 12/08/2023] Open
Abstract
IMPORTANCE Staphylococcus aureus is a Gram-positive opportunistic bacterium that is responsible for the majority of skin infections in humans. Our study provides important molecular insights into the pathogenesis of S. aureus skin infections and identifies a potential therapeutic target for the treatment of these infections. Our findings also indicate that β-hemolysin (Hlb) secreted by colonized S. aureus is a risk factor for epidermal growth factor receptor (EGFR)-related diseases by acting as an agonist of EGFR. The neutralized monoclonal antibody we have developed for the first time will provide a functional inhibitor of Hlb. This study provides important insights to better understand the relationship between the skin colonization of S. aureus and inflammatory skin diseases.
Collapse
Affiliation(s)
- Yonggen Jia
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhangchun Guan
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chenghua Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Minjun Huang
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jingjing Li
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jiannan Feng
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Beifen Shen
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Guang Yang
- Beijing Institute of Tropical Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
12
|
Chen TC, Wang TC, Yiu ZZN, Lee MS, Chen LC, Chan KA, Griffiths CEM, Ashcroft DM. Risk of serious infection and infection mortality in patients with psoriasis: A nationwide cohort study using the Taiwan National Health Insurance claims database. J Eur Acad Dermatol Venereol 2024; 38:136-144. [PMID: 37611288 DOI: 10.1111/jdv.19466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND The risks of serious infections that lead to hospitalization and mortality in patients with psoriasis in Asia have not been comprehensively studied. OBJECTIVES We examined the incidence of serious infection and infection mortality in patients with psoriasis. METHODS This population-based retrospective cohort study used the Taiwan National Health Insurance claims database from 2000 to 2017. Adult patients with psoriasis were identified by a relevant International Classification of Diseases (ICD) code and matched to six comparators without psoriasis on age and sex. Psoriasis patients were categorized as having moderate-to-severe disease once exposed to systemic therapies, phototherapy or biologic therapies. The incidence of serious infection and infection mortality were identified by ICD codes from inpatient hospitalization and death registration. Cox proportional hazard models were used to compare the risk, and the results were adjusted for covariates and presented as adjusted hazard ratios (aHR) and 95% confidence interval (95% CI). RESULTS Overall, 185,434 psoriasis patients and 1,112,581 comparators were included. A higher rate of serious infection (aHR: 1.21, 95% CI: 1.19-1.22) was found in patients with psoriasis compared to matched comparators without psoriasis, and the risk was enhanced when patients had moderate-to-severe psoriasis (aHR: 1.30, 95% CI: 1.27-1.34). Specifically, there was an increased risk of serious infection due to respiratory infections (aHR: 1.11, 95% CI: 1.09-1.13), skin/soft-tissue infections (aHR: 1.57, 95% CI: 1.52-1.62), sepsis (aHR: 1.23, 95% CI: 1.19-1.27), urinary tract infections (aHR: 1.11, 95% CI: 1.08-1.14), hepatitis B (aHR: 1.18, 95% CI: 1.06-1.30) and hepatitis C (aHR: 1.49, 95% CI: 1.32-1.69). Furthermore, psoriasis patients were associated with a higher risk of infection-related mortality (aHR: 1.15, 95% CI: 1.11-1.18) compared to matched comparators. CONCLUSION Patients with psoriasis had a higher risk of serious infection and infection mortality, which was enhanced by moderate-to-severe psoriasis. Practitioners should be aware of the increased risk in patients with psoriasis, but it should not be a barrier to offering effective treatment.
Collapse
Affiliation(s)
- Teng-Chou Chen
- Centre for Pharmacoepidemiology and Drug Safety, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Department of Pharmacy, School of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Chun Wang
- Health Data Research Center, National Taiwan University, Taipei, Taiwan
| | - Zenas Z N Yiu
- Dermatology Centre, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Meng-Sui Lee
- Department of Dermatology, Taipei City Hospital, Taipei, Taiwan
- Department of Dermatology, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Li-Chia Chen
- Centre for Pharmacoepidemiology and Drug Safety, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - K Arnold Chan
- Health Data Research Center, National Taiwan University, Taipei, Taiwan
| | - Christopher E M Griffiths
- Dermatology Centre, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Darren M Ashcroft
- Centre for Pharmacoepidemiology and Drug Safety, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- NIHR Greater Manchester Patient Safety Translational Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
13
|
Si W, Li M, Wang K, Li J, Xu M, Zhou X, Bai J, Qu Z, Song G, Wu X, Guo Y, Hu H, Fu D, Yang Z, Wu M, Yan D, Song X, Tian Z. Staphylococcus warneri strain XSB102 exacerbates psoriasis and promotes keratinocyte proliferation in imiquimod-induced psoriasis-like dermatitis mice. Arch Microbiol 2023; 206:3. [PMID: 37991548 DOI: 10.1007/s00203-023-03726-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/19/2023] [Accepted: 10/29/2023] [Indexed: 11/23/2023]
Abstract
Psoriasis is one of the common chronic inflammatory skin diseases worldwide. The skin microbiota plays a role in psoriasis through regulating skin homeostasis. However, the studies on the interactions between symbiotic microbial strains and psoriasis are limited. In this study, Staphylococcus strain XSB102 was isolated from the skin of human, which was identified as Staphylococcus warneri using VITEK2 Compact. To reveal the roles of Staphylococcus warneri on psoriasis, XSB102 were applied on the back of imiquimod-induced psoriasis-like dermatitis mice. The results indicated that it exacerbated the psoriasis and significantly increased the thickening of the epidermis. Furthermore, in vitro experiments confirmed that inactivated strain XSB102 could promote the proliferation of human epidermal keratinocytes (HaCaT) cell. However, real-time quantitative PCR and immunofluorescence results suggested that the expression of inflammatory factors such as IL-17a, IL-6, and so on were not significantly increased, while extracellular matrix related factors such as Col6a3 and TGIF2 were significantly increased after XSB102 administration. This study indicates that Staphylococcus warneri XSB102 can exacerbate psoriasis and promote keratinocyte proliferation independently of inflammatory factors, which paves the way for further exploration of the relationship between skin microbiota and psoriasis.
Collapse
Affiliation(s)
- Wenhao Si
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- Department of Dermatology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Min Li
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Kuan Wang
- Department of Dermatology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Jialin Li
- Department of Dermatology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Mengke Xu
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xiaoyue Zhou
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Jie Bai
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Zhiyuan Qu
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Guoyan Song
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xueya Wu
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Yuqi Guo
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Hua Hu
- Department of Dermatology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Dandan Fu
- Department of Dermatology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Zishan Yang
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Minna Wu
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Dong Yan
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| | - Xiangfeng Song
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| | - Zhongwei Tian
- Department of Dermatology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| |
Collapse
|
14
|
Esquivel-García R, Seker A, Abu-Lail NI, García-Pérez M, Ochoa-Zarzosa A, García-Pérez ME. Ethanolic extract, solvent fractions, and bio-oils from Urtica subincisa: Chemical composition, toxicity, and anti-IL-17 activity on HaCaT keratinocytes. J Herb Med 2022. [DOI: 10.1016/j.hermed.2022.100599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
15
|
Lee HJ, Kim M. Skin Barrier Function and the Microbiome. Int J Mol Sci 2022; 23:13071. [PMID: 36361857 PMCID: PMC9654002 DOI: 10.3390/ijms232113071] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 08/27/2023] Open
Abstract
Human skin is the largest organ and serves as the first line of defense against environmental factors. The human microbiota is defined as the total microbial community that coexists in the human body, while the microbiome refers to the collective genome of these microorganisms. Skin microbes do not simply reside on the skin but interact with the skin in a variety of ways, significantly affecting the skin barrier function. Here, we discuss recent insights into the symbiotic relationships between the microbiome and the skin barrier in physical, chemical, and innate/adaptive immunological ways. We discuss the gut-skin axis that affects skin barrier function. Finally, we examine the effects of microbiome dysbiosis on skin barrier function and the role of these effects in inflammatory skin diseases, such as acne, atopic dermatitis, and psoriasis. Microbiome cosmetics can help restore skin barrier function and improve these diseases.
Collapse
Affiliation(s)
| | - Miri Kim
- Department of Dermatology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, #10, 63-ro, Yeongdeungpo-gu, Seoul 07345, Korea
| |
Collapse
|
16
|
Rizzetto G, Molinelli E, Radi G, Cirioni O, Brescini L, Giacometti A, Offidani A, Simonetti O. MRSA and Skin Infections in Psoriatic Patients: Therapeutic Options and New Perspectives. Antibiotics (Basel) 2022; 11:1504. [PMID: 36358159 PMCID: PMC9686594 DOI: 10.3390/antibiotics11111504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/23/2022] [Accepted: 10/27/2022] [Indexed: 09/29/2023] Open
Abstract
Psoriatic patients present various infectious risk factors, but there are few studies in the literature evaluating the actual impact of psoriasis in severe staphylococcal skin infections. Our narrative review of the literature suggests that psoriatic patients are at increased risk of both colonization and severe infection, during hospitalization, by S. aureus. The latter also appears to play a role in the pathogenesis of psoriasis through the production of exotoxins. Hospitalized psoriatic patients are also at increased risk of MRSA skin infections. For this reason, new molecules are needed that could both overcome bacterial resistance and inhibit exotoxin production. In our opinion, in the near future, topical quorum sensing inhibitors in combination with current anti-MRSA therapies will be able to overcome the increasing resistance and block exotoxin production. Supplementation with Vitamin E (VE) or derivatives could also enhance the effect of anti-MRSA antibiotics, considering that psoriatic patients with metabolic comorbidities show a low intake of VE and low serum levels, making VE supplementation an interesting new perspective.
Collapse
Affiliation(s)
- Giulio Rizzetto
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Elisa Molinelli
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Giulia Radi
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Oscar Cirioni
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Lucia Brescini
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Andrea Giacometti
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Annamaria Offidani
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Oriana Simonetti
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| |
Collapse
|
17
|
Staphylococcus aureus Biofilm Inhibiting Activity of Advanced Glycation Endproduct Crosslink Breaking and Glycation Inhibiting Compounds. Antibiotics (Basel) 2022; 11:antibiotics11101412. [PMID: 36290070 PMCID: PMC9598957 DOI: 10.3390/antibiotics11101412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022] Open
Abstract
Staphylococcus aureus is a Gram-positive bacterium that plays a role in the pathogenesis of skin lesions in diabetes mellitus, atopic dermatitis, and psoriasis, all of which are associated with elevated non-enzymatic glycation biomarkers. The production of biofilm protects resident bacteria from host immune defenses and antibiotic interventions, prolonging pathogen survival, and risking recurrence after treatment. Glycated proteins formed from keratin and glucose induce biofilm formation in S. aureus, promoting dysbiosis and increasing pathogenicity. In this study, several glycation-inhibiting and advanced glycation endproduct (AGE) crosslink-breaking compounds were assayed for their ability to inhibit glycated keratin-induced biofilm formation as preliminary screening for clinical testing candidates. Ascorbic acid, astaxanthin, clove extract, n-phenacylthiazolium bromide, and rosemary extract were examined in an in vitro static biofilm model with S. aureus strain ATCC 12600. Near complete biofilm inhibition was achieved with astaxanthin (ED50 = 0.060 mg/mL), clove extract (ED50 = 0.0087 mg/mL), n-phenacylthiazolium bromide (ED50 = 5.3 mg/mL), and rosemary extract (ED50 = 1.5 mg/mL). The dosage necessary for biofilm inhibition was not significantly correlated with growth inhibition (R2 = 0.055. p = 0.49). Anti-glycation and AGE breaking compounds with biofilm inhibitory activity are ideal candidates for treatment of S. aureus dysbiosis and skin infection that is associated with elevated skin glycation.
Collapse
|
18
|
Hooper MJ, LeWitt TM, Veon FL, Pang Y, Chlipala GE, Feferman L, Green SJ, Sweeney D, Bagnowski KT, Burns MB, Seed PC, Guitart J, Zhou XA. Nasal Dysbiosis in Cutaneous T-Cell Lymphoma Is Characterized by Shifts in Relative Abundances of Non- Staphylococcus Bacteria. JID INNOVATIONS 2022; 2:100132. [PMID: 36161104 PMCID: PMC9500465 DOI: 10.1016/j.xjidi.2022.100132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 12/26/2022] Open
Abstract
The nasal microbiome of patients with cutaneous T-cell lymphoma (CTCL) remains unexplored despite growing evidence connecting nasal bacteria to skin health and disease. Nasal swabs from 45 patients with CTCL (40 with mycosis fungoides, 5 with Sézary syndrome) and 20 healthy controls from the same geographical region (Chicago Metropolitan Area, Chicago, IL) were analyzed using sequencing of 16S ribosomal RNA and tuf2 gene amplicons. Nasal α-diversity did not differ between mycosis fungoides/Sézary syndrome and healthy controls (Shannon index, genus level, P = 0.201), but distinct microbial communities were identified at the class (R2 = 0.104, P = 0.023) and order (R2 = 0.0904, P = 0.038) levels. Increased relative abundance of the genera Catenococcus, Vibrio, Roseomonas, Acinetobacter, and unclassified Clostridiales was associated with increased skin disease burden (P < 0.005, q < 0.05). Performed to accurately resolve nasal Staphylococcus at the species level, tuf2 gene amplicon sequencing revealed no significant differences between mycosis fungoides/Sézary syndrome and healthy controls. Although S. aureus has been shown to worsen CTCL through its toxins, no increase in the relative abundance of this taxon was observed in nasal samples. Despite the lack of differences in Staphylococcus, the CTCL nasal microbiome was characterized by shifts in numerous other bacterial taxa. These data add to our understanding of the greater CTCL microbiome and provide context for comprehending nasal-skin and host‒tumor‒microbial relationships.
Collapse
Affiliation(s)
- Madeline J. Hooper
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tessa M. LeWitt
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Francesca L. Veon
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yanzhen Pang
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - George E. Chlipala
- Research Informatics Core, Research Resources Center, University of Illinois Chicago, Chicago, Illinois, USA
| | - Leo Feferman
- Research Informatics Core, Research Resources Center, University of Illinois Chicago, Chicago, Illinois, USA
| | - Stefan J. Green
- Rush Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, Illinois, USA
| | - Dagmar Sweeney
- Genome Research Core, Genome Research Division, Research Resources Center, University of Illinois Chicago, Chicago, Illinois, USA
| | - Katherine T. Bagnowski
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michael B. Burns
- Department of Biology, Loyola University Chicago, Chicago, Illinois, USA
| | - Patrick C. Seed
- Division of Pediatric Infectious Diseases, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | - Joan Guitart
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Xiaolong A. Zhou
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
19
|
Potential Therapeutic Skin Microbiomes Suppressing Staphylococcus aureus-Derived Immune Responses and Upregulating Skin Barrier Function-Related Genes via the AhR Signaling Pathway. Int J Mol Sci 2022; 23:ijms23179551. [PMID: 36076953 PMCID: PMC9455615 DOI: 10.3390/ijms23179551] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/21/2022] [Accepted: 08/21/2022] [Indexed: 11/17/2022] Open
Abstract
Disruption of the skin microbial balance can exacerbate certain skin diseases and affect prognosis and treatment. Changes in the distribution and prevalence of certain microbial species on the skin, such as Staphylococcus aureus (SA), can impact the development of severe atopic dermatitis (AD) or psoriasis (Pso). A dysfunctional skin barrier develops in AD and Pso due to SA colonization, resulting in keratinization and chronic or progressive chronic inflammation. Disruption of the skin barrier following SA colonization can elevate the production of T helper 2 (Th2)-derived cytokines, which can cause an imbalance in Th1, Th2, and Th17 cells. This study examined the ability of potential therapeutic skin microbiomes, such as Cutibacterium avidum R-CH3 and Staphylococcus hominis R9, to inhibit SA biofilm formation and restore skin barrier function-related genes through the activation of the aryl hydrocarbon receptor (AhR) and the nuclear factor erythroid-2-related factor 2 (Nrf2) downstream target. We observed that IL-4/IL-13-induced downregulation of FLG, LOR, and IVL induced by SA colonization could be reversed by dual AhR/Nrf2 activation. Further, OVOL1 expression may be modulated by functional microbiomes via dual AhR/Nrf2 activation. Our results suggest that our potential therapeutic skin microbiomes can prevent SA-derived Th2-biased skin barrier disruption via IL-13 and IL-4-dependent FLG deregulation, STAT3 activation, and AhR-mediated STAT6 expression.
Collapse
|
20
|
Rungjang A, Meephansan J, Payungporn S, Sawaswong V, Chanchaem P, Pureesrisak P, Wongpiyabovorn J, Thio HB. Skin Microbiota Profiles from Tape Stripping and Skin Biopsy Samples of Patients with Psoriasis Treated with Narrowband Ultraviolet B. Clin Cosmet Investig Dermatol 2022; 15:1767-1778. [PMID: 36065340 PMCID: PMC9440725 DOI: 10.2147/ccid.s374871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/12/2022] [Indexed: 11/23/2022]
Abstract
Purpose Although the pathogenesis of psoriasis involves the dermis, most previous studies collected samples using the swab technique. A recent study examining the microbiomes obtained via both skin biopsies and swabs revealed a significant difference in normal skin. We hypothesized that the microbiome profile of patients with psoriasis from tape stripping and skin biopsy might be different. This study sought to contribute to microbiome research on psoriasis by investigating the changes in the microbiome during narrowband ultraviolet B (NBUVB) therapy by comparing the results from the different sampling techniques of tape stripping and skin biopsy. Patients and Methods Twenty-three participants, including 14 patients with chronic plaque psoriasis and nine healthy controls, were recruited, and nine patients with psoriasis completed 20-sessions of NBUVB treatment. Skin microbiota from both techniques was analyzed using the 16S rRNA gene at baseline and after treatment. Results A clear difference was observed between the results from the two sampling techniques. Alpha diversity of the microbiota obtained from tape stripping was higher than that of the microbiota from skin biopsy, whereas beta diversity was clustered into two groups by sampling technique. The microbiome was altered during NBUVB treatment using both sampling techniques. Conclusion Different sampling techniques resulted in different microbiome profiles in patients with psoriasis. Tape stripping and swabs are feasible procedures and are mostly used in psoriasis and other skin microbiome studies; however, skin biopsy may also expand our understanding of psoriasis and other skin diseases that pathophysiology involves deeper to the dermis or subcutaneous tissue.
Collapse
Affiliation(s)
- Atiya Rungjang
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Jitlada Meephansan
- Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
- Correspondence: Jitlada Meephansan, Division of Dermatology, Chulabhorn International College of Medicine, Thammasat University, Rangsit Campus, Klong Luang, Pathum Thani, 12120, Thailand, Tel +66 0 2564-4444, ext.1535, Fax +66 0 2564-4440, ext.7594, Email
| | - Sunchai Payungporn
- The Research Unit of Systems Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Vorthon Sawaswong
- The Research Unit of Systems Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Prangwalai Chanchaem
- The Research Unit of Systems Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Purit Pureesrisak
- Division of Dermatology, Department of Medicine, Rajavithi Hospital, Ministry of Public Health, Bangkok, Thailand
| | - Jongkonnee Wongpiyabovorn
- Division of Immunology, Department of Microbiology, Faculty of Medicine, Center of Excellence in Immunology and Immune Mediated Diseases, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Hok Bing Thio
- Department of Dermatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
21
|
Javia A, Misra A, Thakkar H. Liposomes encapsulating novel antimicrobial peptide Omiganan: Characterization and its pharmacodynamic evaluation in atopic dermatitis and psoriasis mice model. Int J Pharm 2022; 624:122045. [PMID: 35878872 DOI: 10.1016/j.ijpharm.2022.122045] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/18/2022] [Accepted: 07/20/2022] [Indexed: 11/28/2022]
Abstract
Omiganan is a novel 12 amino acid synthetic cationic peptide from the cathelicidin family. Omiganan possesses antimicrobial action against a wide range of microbes, including gram-positive and gram-negative bacteria and fungi. Omiganan mainly acts by depolarizing the cytoplasmic membrane, resulting in cellular disruption and death. Apart from its antimicrobial effect, Omiganan also has anti-inflammatory activity. The present investigation aimed to evaluate and compare the efficacy of Omiganan liposomal gel with conventional formulations (Omiganan gel and lotion) in atopic dermatitis (AD) and psoriasis mice animal models. Liposomes encapsulating Omiganan were prepared using the reverse-phase evaporation technique and incorporated into Carbopol 934P gel. The optimized Omiganan liposomes were then characterized for various physicochemical parameters such as vesicle size, shape and surface morphology, zeta-potential, rheological parameters, in-vitro drug release, ex-vivo skin permeation/deposition, in-vitro antimicrobial activity, proteolytic stability, and cellular toxicity and uptake studies. Liposomes exhibited 72 % encapsulation with 7.8 % loading efficacy, a vesicle size, and zeta potential of 120 nm and - 17.2 mv, respectively. Moreover, Omiganan liposomal gel demonstrated controlled release and a better permeation profile than conventional formulations. A substantial reduction in levels of pro-inflammatory cytokines and improvement in AD and psoriatic lesions were achieved by Omiganan liposomal gel compared to Omiganan gel and lotion-based formulations. The present study confirms that Omiganan liposomal formulation can be an effective, safe, and novel alternative treatment approach in atopic dermatitis and psoriasis.
Collapse
Affiliation(s)
- Ankit Javia
- Pharmacy Department, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat-390001, India
| | - Ambikanandan Misra
- Pharmacy Department, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat-390001, India
| | - Hetal Thakkar
- Pharmacy Department, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat-390001, India.
| |
Collapse
|
22
|
Exploring the Role of Staphylococcus aureus in Inflammatory Diseases. Toxins (Basel) 2022; 14:toxins14070464. [PMID: 35878202 PMCID: PMC9318596 DOI: 10.3390/toxins14070464] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 02/04/2023] Open
Abstract
Staphylococcus aureus is a very common Gram-positive bacterium, and S. aureus infections play an extremely important role in a variety of diseases. This paper describes the types of virulence factors involved, the inflammatory cells activated, the process of host cell death, and the associated diseases caused by S. aureus. S. aureus can secrete a variety of enterotoxins and other toxins to trigger inflammatory responses and activate inflammatory cells, such as keratinocytes, helper T cells, innate lymphoid cells, macrophages, dendritic cells, mast cells, neutrophils, eosinophils, and basophils. Activated inflammatory cells can express various cytokines and induce an inflammatory response. S. aureus can also induce host cell death through pyroptosis, apoptosis, necroptosis, autophagy, etc. This article discusses S. aureus and MRSA (methicillin-resistant S. aureus) in atopic dermatitis, psoriasis, pulmonary cystic fibrosis, allergic asthma, food poisoning, sarcoidosis, multiple sclerosis, and osteomyelitis. Summarizing the pathogenic mechanism of Staphylococcus aureus provides a basis for the targeted treatment of Staphylococcus aureus infection.
Collapse
|
23
|
Simioni YR, Perez NS, Barbosa LR, Perez AP, Schilrreff P, Romero EL, Morilla MJ. Enhancing the anti-psoriatic activity of vitamin D3 employing nanostructured archaeolipid carriers. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
24
|
Bu J, Ding R, Zhou L, Chen X, Shen E. Epidemiology of Psoriasis and Comorbid Diseases: A Narrative Review. Front Immunol 2022; 13:880201. [PMID: 35757712 PMCID: PMC9226890 DOI: 10.3389/fimmu.2022.880201] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/28/2022] [Indexed: 11/18/2022] Open
Abstract
Psoriasis is a chronic autoimmune inflammatory disease that remains active for a long period, even for life in most patients. The impact of psoriasis on health is not only limited to the skin, but also influences multiple systems of the body, even mental health. With the increasing of literature on the association between psoriasis and extracutaneous systems, a better understanding of psoriasis as an autoimmune disease with systemic inflammation is created. Except for cardiometabolic diseases, gastrointestinal diseases, chronic kidney diseases, malignancy, and infections that have received much attention, the association between psoriasis and more systemic diseases, including the skin system, reproductive system, and oral and ocular systems has also been revealed, and mental health diseases draw more attention not just because of the negative mental and mood influence caused by skin lesions, but a common immune-inflammatory mechanism identified of the two systemic diseases. This review summarizes the epidemiological evidence supporting the association between psoriasis and important and/or newly reported systemic diseases in the past 5 years, and may help to comprehensively recognize the comorbidity burden related to psoriasis, further to improve the management of people with psoriasis.
Collapse
Affiliation(s)
- Jin Bu
- Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Ruilian Ding
- Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Liangjia Zhou
- Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xiangming Chen
- Sino-French Hoffmann Institute, School of Basic Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Erxia Shen
- Sino-French Hoffmann Institute, School of Basic Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- The State Key Laboratory of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
25
|
Effect of Melatonin on Psoriatic Phenotype in Human Reconstructed Skin Model. Biomedicines 2022; 10:biomedicines10040752. [PMID: 35453501 PMCID: PMC9032986 DOI: 10.3390/biomedicines10040752] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/15/2022] [Accepted: 03/22/2022] [Indexed: 02/06/2023] Open
Abstract
Psoriasis is an inflammatory and auto-immune skin-disease characterized by uncontrolled keratinocyte proliferation. Its pathogenesis is not still fully understood; however, an aberrant and excessive inflammatory and immune response can contribute to its progression. Recently, more attention has been given to the anti-inflammatory and immunomodulators effects of melatonin in inflammatory diseases. The aim of this paper was to investigate the effect of melatonin on psoriatic phenotype and also in S. aureus infection-associated psoriasis, with an in vitro model using Skinethic Reconstructed Human Epidermis (RHE). An in vitro model was constructed using the RHE, a three-dimensional-model obtained from human primary-keratinocytes. RHE-cells were exposed to a mix of pro-inflammatory cytokines, to induce a psoriatic phenotype; cells were also infected with S. aureus to aggravate psoriasis disease, and then were treated with melatonin at the concentrations of 1 nM, 10 nM, and 50 nM. Our results demonstrated that melatonin at higher concentrations significantly reduced histological damage, compared to the cytokine and S. aureus groups. Additionally, the treatment with melatonin restored tight-junction expression and reduced pro-inflammatory cytokine levels, such as interleukin-1β and interleukin-12. Our results suggest that melatonin could be considered a promising strategy for psoriasis-like skin inflammation, as well as complications of psoriasis, such as S. aureus infection.
Collapse
|
26
|
von Köckritz-Blickwede M, Winstel V. Molecular Prerequisites for Neutrophil Extracellular Trap Formation and Evasion Mechanisms of Staphylococcus aureus. Front Immunol 2022; 13:836278. [PMID: 35237275 PMCID: PMC8884242 DOI: 10.3389/fimmu.2022.836278] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/19/2022] [Indexed: 12/15/2022] Open
Abstract
NETosis is a multi-facetted cellular process that promotes the formation of neutrophil extracellular traps (NETs). NETs as web-like structures consist of DNA fibers armed with granular proteins, histones, and microbicidal peptides, thereby exhibiting pathogen-immobilizing and antimicrobial attributes that maximize innate immune defenses against invading microbes. However, clinically relevant pathogens often tolerate entrapment and even take advantage of the remnants of NETs to cause persistent infections in mammalian hosts. Here, we briefly summarize how Staphylococcus aureus, a high-priority pathogen and causative agent of fatal diseases in humans as well as animals, catalyzes and concurrently exploits NETs during pathogenesis and recurrent infections. Specifically, we focus on toxigenic and immunomodulatory effector molecules produced by staphylococci that prime NET formation, and further highlight the molecular and underlying principles of suicidal NETosis compared to vital NET-formation by viable neutrophils in response to these stimuli. We also discuss the inflammatory potential of NET-controlled microenvironments, as excessive expulsion of NETs from activated neutrophils provokes local tissue injury and may therefore amplify staphylococcal disease severity in hospitalized or chronically ill patients. Combined with an overview of adaptation and counteracting strategies evolved by S. aureus to impede NET-mediated killing, these insights may stimulate biomedical research activities to uncover novel aspects of NET biology at the host-microbe interface.
Collapse
Affiliation(s)
- Maren von Köckritz-Blickwede
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Volker Winstel
- Research Group Pathogenesis of Bacterial Infections, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
- *Correspondence: Volker Winstel,
| |
Collapse
|
27
|
Patel A, Venter F, Grewal K, Sharma R, Petersen G, Heidari A. Erythrodermic Psoriasis and Staph-Infective Endocarditis—A Conundrum in Succession. J Investig Med High Impact Case Rep 2022; 10:23247096221105243. [PMID: 35726179 PMCID: PMC9218485 DOI: 10.1177/23247096221105243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Erythrodermic psoriasis is a rare subtype of psoriasis vulgaris that presents with diffuse erythema and desquamation over greater than 75% of the body’s surface area. We present a case of a 57-year-old male who was admitted with a diffuse, erythematous scaly rash covering his entire body, with associated subjective fevers. Skin biopsy revealed erythrodermic psoriasis, and blood cultures were positive for methicillin-sensitive Staphylococcus aureus. Echocardiogram revealed a mitral valve vegetation. Clinical improvement was achieved with intravenous antibiotic administration and topical corticosteroids without the use of immunomodulators.
Collapse
Affiliation(s)
- Arti Patel
- Department of Internal Medicine, Kern Medical, Bakersfield, CA, USA
| | - Frederick Venter
- Department of Internal Medicine, Kern Medical, Bakersfield, CA, USA
| | - Kulraj Grewal
- Department of Internal Medicine, Kern Medical, Bakersfield, CA, USA
| | - Rupam Sharma
- Department of Internal Medicine, Kern Medical, Bakersfield, CA, USA
| | - Greti Petersen
- Department of Internal Medicine, Kern Medical, Bakersfield, CA, USA
| | - Arash Heidari
- Department of Internal Medicine, Kern Medical, Bakersfield, CA, USA
- Division of Infectious Disease, Kern Medical, Bakersfield, CA, USA
| |
Collapse
|
28
|
Nikolaeva MY, Monakhov KN, Sokolovskiy EV. Disorders of the skin microbiome in atopic dermatitis and psoriasis. VESTNIK DERMATOLOGII I VENEROLOGII 2021. [DOI: 10.25208/vdv1282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
The notion of skin microbiome encompasses a heterogeneous group of microorganisms that belong to various taxonomic units, such as bacteria, archaea, viruses, and fungi. The impact of these microbial community constituents upon the epidermal barrier condition, and upon the immune system functioning, is being intensely scrutinized. There is a particular interest in studying the role that the microorganisms of genus Staphylococcus spp. play in the course of physiological and pathological processes occurring in the skin. This review examines in detail the interaction of the microorganisms of genus Staphylococcus spp. with the microbial community constituents, as well as with the skin immune system in normal condition and in the condition associated with inflammatory dermatoses. There are also the data given on S. aureus pathogenicity factors, the data on the impact of this microorganism upon the course of atopic dermatitis, and upon the course of psoriasis. The review examines the role that coagulase-negative staphylococci, S. epidermidis in particular, play in maintaining the microbiome homeostasis. The review as well examines the impact of the skin microbiome upon the development and activity of the skin immune system, and upon maintaining the integrity of the epidermal barrier.
Collapse
|
29
|
Liang X, Ou C, Zhuang J, Li J, Zhang F, Zhong Y, Chen Y. Interplay Between Skin Microbiota Dysbiosis and the Host Immune System in Psoriasis: Potential Pathogenesis. Front Immunol 2021; 12:764384. [PMID: 34733291 PMCID: PMC8558530 DOI: 10.3389/fimmu.2021.764384] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Psoriasis is a multifactorial immune-mediated disease. The highly effective and eligible treatment for psoriasis is limited, for its specific pathogenesis is incompletely elucidated. Skin microbiota is a research hotspot in the pathogenesis of immune-mediated inflammatory skin diseases nowadays, and it may have significant involvement in the provocation or exacerbation of psoriasis with broadly applicable prospects. It is postulated that skin microbiota alternation may interplay with innate immunity such as antimicrobial peptides and Toll-like receptors to stimulate T-cell populations, resulting in immune cascade responses and ultimately psoriasis. Achieving a thorough understanding of its underlying pathogenesis is crucial. Herein, we discuss the potential immunopathogenesis of psoriasis from the aspect of skin microbiota in an attempt to yield insights for novel therapeutic and preventive modalities for psoriasis.
Collapse
Affiliation(s)
- Xiaoqian Liang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Caixin Ou
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Jiayi Zhuang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Jinsheng Li
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Fangfei Zhang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Yuanqiu Zhong
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Yongfeng Chen
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
30
|
Orsmond A, Bereza-Malcolm L, Lynch T, March L, Xue M. Skin Barrier Dysregulation in Psoriasis. Int J Mol Sci 2021; 22:10841. [PMID: 34639182 PMCID: PMC8509518 DOI: 10.3390/ijms221910841] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023] Open
Abstract
The skin barrier is broadly composed of two elements-a physical barrier mostly localised in the epidermis, and an immune barrier localised in both the dermis and epidermis. These two systems interact cooperatively to maintain skin homeostasis and overall human health. However, if dysregulated, several skin diseases may arise. Psoriasis is one of the most prevalent skin diseases associated with disrupted barrier function. It is characterised by the formation of psoriatic lesions, the aberrant differentiation and proliferation of keratinocytes, and excessive inflammation. In this review, we summarize recent discoveries in disease pathogenesis, including the contribution of keratinocytes, immune cells, genetic and environmental factors, and how they advance current and future treatments.
Collapse
Affiliation(s)
- Andreas Orsmond
- Sutton Arthritis Research Laboratory, Faculty of Medicine and Health, Institute of Bone and Joint Research, Kolling Institute, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (A.O.); (L.B.-M.)
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Faculty of Medicine and Health, Institute of Bone and Joint Research, Kolling Institute, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (T.L.); (L.M.)
| | - Lara Bereza-Malcolm
- Sutton Arthritis Research Laboratory, Faculty of Medicine and Health, Institute of Bone and Joint Research, Kolling Institute, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (A.O.); (L.B.-M.)
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Faculty of Medicine and Health, Institute of Bone and Joint Research, Kolling Institute, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (T.L.); (L.M.)
| | - Tom Lynch
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Faculty of Medicine and Health, Institute of Bone and Joint Research, Kolling Institute, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (T.L.); (L.M.)
| | - Lyn March
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Faculty of Medicine and Health, Institute of Bone and Joint Research, Kolling Institute, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (T.L.); (L.M.)
| | - Meilang Xue
- Sutton Arthritis Research Laboratory, Faculty of Medicine and Health, Institute of Bone and Joint Research, Kolling Institute, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (A.O.); (L.B.-M.)
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Faculty of Medicine and Health, Institute of Bone and Joint Research, Kolling Institute, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (T.L.); (L.M.)
| |
Collapse
|
31
|
Pistone D, Meroni G, Panelli S, D’Auria E, Acunzo M, Pasala AR, Zuccotti GV, Bandi C, Drago L. A Journey on the Skin Microbiome: Pitfalls and Opportunities. Int J Mol Sci 2021; 22:9846. [PMID: 34576010 PMCID: PMC8469928 DOI: 10.3390/ijms22189846] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/22/2022] Open
Abstract
The human skin microbiota is essential for maintaining homeostasis and ensuring barrier functions. Over the years, the characterization of its composition and taxonomic diversity has reached outstanding goals, with more than 10 million bacterial genes collected and cataloged. Nevertheless, the study of the skin microbiota presents specific challenges that need to be addressed in study design. Benchmarking procedures and reproducible and robust analysis workflows for increasing comparability among studies are required. For various reasons and because of specific technical problems, these issues have been investigated in gut microbiota studies, but they have been largely overlooked for skin microbiota. After a short description of the skin microbiota, the review tackles methodological aspects and their pitfalls, covering NGS approaches and high throughput culture-based techniques. Recent insights into the "core" and "transient" types of skin microbiota and how the manipulation of these communities can prevent or combat skin diseases are also covered. Finally, this review includes an overview of the main dermatological diseases, the changes in the microbiota composition associated with them, and the recommended skin sampling procedures. The last section focuses on topical and oral probiotics to improve and maintain skin health, considering their possible applications for skin diseases.
Collapse
Affiliation(s)
- Dario Pistone
- Pediatric Clinical Research Center “Invernizzi”, Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy; (S.P.); (A.R.P.); (G.V.Z.)
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy;
| | - Gabriele Meroni
- Department of Biomedical Surgical and Dental Sciences-One Health Unit, University of Milan, 20133 Milan, Italy;
| | - Simona Panelli
- Pediatric Clinical Research Center “Invernizzi”, Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy; (S.P.); (A.R.P.); (G.V.Z.)
| | - Enza D’Auria
- Department of Pediatrics, Children’s Hospital Vittore Buzzi, University of Milan, 20154 Milan, Italy; (E.D.); (M.A.)
| | - Miriam Acunzo
- Department of Pediatrics, Children’s Hospital Vittore Buzzi, University of Milan, 20154 Milan, Italy; (E.D.); (M.A.)
| | - Ajay Ratan Pasala
- Pediatric Clinical Research Center “Invernizzi”, Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy; (S.P.); (A.R.P.); (G.V.Z.)
| | - Gian Vincenzo Zuccotti
- Pediatric Clinical Research Center “Invernizzi”, Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy; (S.P.); (A.R.P.); (G.V.Z.)
- Department of Pediatrics, Children’s Hospital Vittore Buzzi, University of Milan, 20154 Milan, Italy; (E.D.); (M.A.)
| | - Claudio Bandi
- Pediatric Clinical Research Center “Invernizzi”, Department of Biosciences, University of Milan, 20133 Milan, Italy;
| | - Lorenzo Drago
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy;
| |
Collapse
|
32
|
IL-17C and IL-17RE Promote Wound Closure in a Staphylococcus aureus-Based Murine Wound Infection Model. Microorganisms 2021; 9:microorganisms9091821. [PMID: 34576717 PMCID: PMC8469012 DOI: 10.3390/microorganisms9091821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 11/17/2022] Open
Abstract
The epithelial cytokine interleukin-17C (IL-17C) mediates inflammation through the interleukin 17 receptor E (IL-17RE). Prior studies showed a detrimental role of IL-17C in the pathogenesis of immune-mediated skin diseases (e.g., psoriasis). Here, we examined the role of IL-17C/IL-17RE in wound closure in a Staphylococcus aureus wound infection model. We demonstrate that wound closure is significantly delayed in IL-17RE (Il-17re−/−)- and 17C (Il-17c−/−)-deficient mice. There was no significant difference between WT, Il-17re−/−, and Il-17c−/− mice in the absence of infection. Deficiency for IL-17RE and IL-17C did not significantly affect the elimination of bacteria. IL-17C expression was increased in the epidermis of human S. aureus-infected skin. Our results indicate that the IL-17C/IL-17RE axis contributes to the closure of infected wounds but does not contribute to the elimination of S. aureus.
Collapse
|
33
|
Gundogan B, Dowlut N, Rajmohan S, Borrelli MR, Millip M, Iosifidis C, Udeaja YZ, Mathew G, Fowler A, Agha R. Assessing the compliance of systematic review articles published in leading dermatology journals with the PRISMA statement guidelines: A systematic review. JAAD Int 2021; 1:157-174. [PMID: 34409336 PMCID: PMC8361930 DOI: 10.1016/j.jdin.2020.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2020] [Indexed: 11/24/2022] Open
Abstract
Background Reporting quality of systematic reviews and meta-analyses is of critical importance in dermatology because of their key role in informing health care decisions. Objective To assess the compliance of systematic reviews and meta-analyses in leading dermatology journals with the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statement guidelines. Methods This review was carried out in accordance with PRISMA guidelines. Included studies were reviews published across 6 years in the top 4 highest-impact-factor dermatology journals of 2017. Records and full texts were screened independently. Data analysis was conducted with univariate multivariable linear regression. The primary outcome was to assess the compliance of systematic reviews and meta-analyses in leading dermatology journals with the PRISMA statement. Results A total of 166 studies were included and mean PRISMA compliance across all articles was 73%. Compliance significantly improved over time (β = .016; P = <.001). The worst reported checklist item was item 5 (reporting on protocol existence), with a compliance of 15% of articles. Conclusion PRISMA compliance within leading dermatology journals could be improved; however, it is steadily improving.
Collapse
Affiliation(s)
- Buket Gundogan
- University College London Hospital, London, United Kingdom
| | - Naeem Dowlut
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | | | - Mimi R Borrelli
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Mirabel Millip
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Christos Iosifidis
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Yagazie Z Udeaja
- Luton and Dunstable University Hospital NHS Foundation Trust, Luton, United Kingdom
| | - Ginimol Mathew
- University College London Medical School, Gower Street, London, United Kingdom
| | | | - Riaz Agha
- Bart's Health NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
34
|
Alenius H, Sinkko H, Moitinho-Silva L, Rodriguez E, Broderick C, Alexander H, Reiger M, Hjort Hjelmsø M, Fyhrquist N, Olah P, Bryce P, Smith C, Koning F, Eyerich K, Greco D, van den Bogaard EH, Neumann AU, Traidl-Hoffmann C, Homey B, Flohr C, Bønnelykke K, Stokholm J, Weidinger S. The power and potential of BIOMAP to elucidate host-microbiome interplay in skin inflammatory diseases. Exp Dermatol 2021; 30:1517-1531. [PMID: 34387406 DOI: 10.1111/exd.14446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/02/2021] [Accepted: 08/02/2021] [Indexed: 11/27/2022]
Abstract
The two most common chronic inflammatory skin diseases are atopic dermatitis (AD) and psoriasis. The underpinnings of the remarkable degree of clinical heterogeneity of AD and psoriasis are poorly understood and, as a consequence, disease onset and progression are unpredictable and the optimal type and time-point for intervention are as yet unknown. The BIOMAP project is the first IMI (Innovative Medicines Initiative) project dedicated to investigating the causes and mechanisms of AD and psoriasis and to identify potential biomarkers responsible for the variation in disease outcome. The consortium includes 7 large pharmaceutical companies and 25 non-industry partners including academia. Since there is mounting evidence supporting an important role for microbial exposures and our microbiota as factors mediating immune polarization and AD and psoriasis pathogenesis, an entire work package is dedicated to the investigation of skin and gut microbiome linked to AD or psoriasis. The large collaborative BIOMAP project will enable the integration of patient cohorts, data and knowledge in unprecedented proportions. The project has a unique opportunity with a potential to bridge and fill the gaps between current problems and solutions. This review highlights the power and potential of BIOMAP project in the investigation of microbe-host interplay in AD and psoriasis.
Collapse
Affiliation(s)
- Harri Alenius
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, 171 77, Stockholm, Sweden.,Human Microbiome Research Program (HUMI), Faculty of Medicine, University of Helsinki, Finland
| | - Hanna Sinkko
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, 171 77, Stockholm, Sweden.,Human Microbiome Research Program (HUMI), Faculty of Medicine, University of Helsinki, Finland
| | - Lucas Moitinho-Silva
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.,Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Elke Rodriguez
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Conor Broderick
- Unit for Population-Based Dermatology Research, St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Helen Alexander
- Unit for Population-Based Dermatology Research, St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Matthias Reiger
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany.,Chair of Environmental Medicine, Technical University Munich, Munich, Germany
| | - Mathis Hjort Hjelmsø
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nanna Fyhrquist
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, 171 77, Stockholm, Sweden
| | - Peter Olah
- Department of Dermatology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.,Department of Dermatology, Venereology and Oncodermatology, Medical Faculty, University of Pécs, Hungary
| | - Paul Bryce
- Type 2 Inflammation & Fibrosis Cluster, Immunology & Inflammation Therapeutic Area, Sanofi US, Cambridge, MA, United States of America
| | - Catherine Smith
- St John's Institute of Dermatology, Kings College London, and Guys and St Thomas' NHS Foundation Trust, 9th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Frits Koning
- Department of Immunology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Kilian Eyerich
- Department of Medicine, Karolinska Institutet, Solna, Sweden
| | - Dario Greco
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.,Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ellen H van den Bogaard
- Department of Dermatology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Avidan U Neumann
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
| | - Claudia Traidl-Hoffmann
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany.,Chair of Environmental Medicine, Technical University Munich, Munich, Germany.,CK CARE, Christine Kühne Center for Allergy Research and Education, Davos, Switzerland.,ZIEL - Institute for Food & Health, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Bernhard Homey
- Department of Dermatology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Carsten Flohr
- Unit for Population-Based Dermatology Research, St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, United Kingdom
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Stokholm
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.,Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Stephan Weidinger
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
35
|
[Dysbalance between the immune system and skin microbiome in chronic inflammatory dermatoses]. Hautarzt 2021; 72:570-577. [PMID: 34136940 DOI: 10.1007/s00105-021-04832-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND The skin is an organ frequently affected by chronic diseases. Inflammatory, immune-mediated dermatoses such as atopic dermatitis and psoriasis show a high prevalence as well as a significant impact on the quality of life of those affected. In a large proportion of cases, atopic dermatitis is associated with a marked change in microbial colonization of both clinically healthy and affected skin. In psoriasis, changes to this effect have been described, but clinical relevance remains elusive. AIM In recent years, increasing knowledge has been gained in microbiome research with resulting clinical relevance. The present article deals with the disturbed balance of the immune system and the skin microbiome in chronic inflammatory dermatoses on the basis of atopic dermatitis and psoriasis vulgaris. MATERIALS AND METHODS A literature search was performed in PubMed and Medline databases (entries until 09 April 2021). RESULTS Staphylococcus aureus is known to play a central pathophysiological role in atopic dermatitis. This is revisited in light of new insights regarding biodiversity and immunoregulatory processes. In psoriasis, a more heterogeneous body of data emerges regarding the microbiome and its contribution to disease development. DISCUSSION While topical applications to directly influence the microbiome are already being tested in atopic dermatitis, further knowledge regarding the pathophysiological significance of the microbiota is still needed in psoriasis.
Collapse
|
36
|
Teng Y, Xie W, Tao X, Liu N, Yu Y, Huang Y, Xu D, Fan Y. Infection-provoked psoriasis: Induced or aggravated (Review). Exp Ther Med 2021; 21:567. [PMID: 33850539 PMCID: PMC8027725 DOI: 10.3892/etm.2021.9999] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
Psoriasis is a common chronic, immune-mediated, inflammatory skin disorder, with a reported prevalence of 0.0-2.1% among children and 0.91-8.50% among adults, worldwide. Psoriasis is induced by several environmental factors, including infection, alcohol consumption, drugs, trauma, acute withdrawal of systemic or potent topical corticosteroids, body mass index and endocrine disorders. Increasing evidence suggest that a variety of microorganisms play key roles in the induction and exacerbation of psoriasis. Pathogens, such as streptococci and staphylococci are considered causal factors, presumably via superantigen activation of skin-seeking T cells. In addition, fungal pathogens, such as Candida and Malassezia, and viral agents, such as human immunodeficiency virus, hepatitis C virus infection and human papillomavirus, are also closely associated with psoriasis. Recently, several types of pathogens, such as Helicobacter pylori infection, Zika virus and scabies, have been reported to potentially trigger psoriasis. The present review discusses the underlying molecular mechanisms by which these infections influence psoriasis to provide a better understanding of the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Yan Teng
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Wenqing Xie
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiaohua Tao
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Na Liu
- Department of Dermatology, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Yong Yu
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Youming Huang
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Danfeng Xu
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Yibin Fan
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
37
|
Jia J, Wang N, Zheng Y, Mo X, Zhang Y, Ye S, Liu J, Yan F, Li H, Chen D. RAS-association domain family 1A regulates the abnormal cell proliferation in psoriasis via inhibition of Yes-associated protein. J Cell Mol Med 2021; 25:5070-5081. [PMID: 33960627 PMCID: PMC8178269 DOI: 10.1111/jcmm.16489] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 03/04/2021] [Accepted: 03/12/2021] [Indexed: 12/17/2022] Open
Abstract
Psoriasis is a chronic, inflammatory skin disease with a high incidence and recurrence; however, its exact pathogenesis and aetiology remain unclear. This study aimed to analyse the effect of the upstream negative regulator RAS‐association domain family 1A (RASSF1A) on Yes‐associated protein (YAP) in psoriasis. Skin lesions of 22 patients with psoriasis and 19 healthy controls were used. Human epidermal keratinocytes stimulated by M5 (IL‐1α, IL‐17, IL‐22, TNF‐α and oncostatin M) were used to establish a psoriatic cell model. BALB/c mice treated with topical imiquimod were used to establish a psoriatic mouse model. As the methylation level of RASSF1A increased, its expression in psoriatic patients and mice model decreased. Addition of the methylation inhibitor 5‐Aza‐CdR or RASSF1A‐overexpressing lentivirus vector increased RASSF1A and reduced YAP expression; meanwhile improved skin lesions, reduced cell proliferation, induced cell cycle arrest in the G0/G1 phase, increased apoptosis, reduced inflammatory cytokines and activities of ERK, STAT3 and NF‐κB signalling pathways. The results indicated that RASSF1A could play a role in the treatment of psoriasis by inhibiting YAP expression. Based on these findings, targeted drugs that can inhibit the methylation or increase the expression of RASSF1A may be useful for treating psoriasis.
Collapse
Affiliation(s)
- Jinjing Jia
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, Guangzhou, China
| | - Ning Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Zheng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiumei Mo
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, Guangzhou, China
| | - Yu Zhang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, Guangzhou, China
| | - Siqi Ye
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, Guangzhou, China
| | - Junfeng Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, Guangzhou, China
| | - Fenggen Yan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, Guangzhou, China
| | - Hongyi Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, Guangzhou, China
| | - Dacan Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Disease, Guangzhou, China
| |
Collapse
|
38
|
Wang G, Sweren E, Liu H, Wier E, Alphonse MP, Chen R, Islam N, Li A, Xue Y, Chen J, Park S, Chen Y, Lee S, Wang Y, Wang S, Archer NK, Andrews W, Kane MA, Dare E, Reddy SK, Hu Z, Grice EA, Miller LS, Garza LA. Bacteria induce skin regeneration via IL-1β signaling. Cell Host Microbe 2021; 29:777-791.e6. [PMID: 33798492 PMCID: PMC8122070 DOI: 10.1016/j.chom.2021.03.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/03/2021] [Accepted: 02/24/2021] [Indexed: 02/08/2023]
Abstract
Environmental factors that enhance regeneration are largely unknown. The immune system and microbiome are attributed roles in repairing and regenerating structure but their precise interplay is unclear. Here, we assessed the function of skin bacteria in wound healing and wound-induced hair follicle neogenesis (WIHN), a rare adult organogenesis model. WIHN levels and stem cell markers correlate with bacterial counts, being lowest in germ-free (GF), intermediate in conventional specific pathogen-free (SPF), and highest in wild-type mice, even those infected with pathogenic Staphylococcus aureus. Reducing skin microbiota via cage changes or topical antibiotics decreased WIHN. Inflammatory cytokine IL-1β and keratinocyte-dependent IL-1R-MyD88 signaling are necessary and sufficient for bacteria to promote regeneration. Finally, in a small trial, a topical broad-spectrum antibiotic also slowed skin wound healing in adult volunteers. These results demonstrate a role for IL-1β to control morphogenesis and support the need to reconsider routine applications of topical prophylactic antibiotics.
Collapse
Affiliation(s)
- Gaofeng Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Evan Sweren
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Eric Wier
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Ruosi Chen
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Nasif Islam
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Ang Li
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Yingchao Xue
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Junjie Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21210, USA
| | - Seungman Park
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21210, USA
| | - Yun Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21210, USA
| | - Sam Lee
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Saifeng Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Nate K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - William Andrews
- Department of Pharmaceutical Sciences, School of Pharmacy Mass Spectrometry Center, University of Maryland, MD 21201, USA
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, School of Pharmacy Mass Spectrometry Center, University of Maryland, MD 21201, USA
| | - Erika Dare
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Sashank K Reddy
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Elizabeth A Grice
- Department of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Luis A Garza
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA.
| |
Collapse
|
39
|
Patrick MT, Zhang H, Wasikowski R, Prens EP, Weidinger S, Gudjonsson JE, Elder JT, He K, Tsoi LC. Associations between COVID-19 and skin conditions identified through epidemiology and genomic studies. J Allergy Clin Immunol 2021; 147:857-869.e7. [PMID: 33485957 PMCID: PMC7825803 DOI: 10.1016/j.jaci.2021.01.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is commonly associated with skin manifestations, and may also exacerbate existing skin diseases, yet the relationship between COVID-19 and skin diseases remains unclear. OBJECTIVE By investigating this relationship through a multiomics approach, we sought to ascertain whether patients with skin conditions are more susceptible to COVID-19. METHODS We conducted an epidemiological study and then compared gene expression across 9 different inflammatory skin conditions and severe acute respiratory syndrome coronavirus 2-infected bronchial epithelial cell lines, and then performed a genome-wide association study transdisease meta-analysis between COVID-19 susceptibility and 2 skin diseases (psoriasis and atopic dermatitis). RESULTS Skin conditions, including psoriasis and atopic dermatitis, increase the risk of COVID-19 (odds ratio, 1.55; P = 1.4 × 10-9) but decrease the risk of mechanical ventilation (odds ratio, 0.22; P = 8.5 × 10-5). We observed significant overlap in gene expression between the infected normal bronchial epithelial cells and inflammatory skin diseases, such as psoriasis and atopic dermatitis. For genes that are commonly induced in both the severe acute respiratory syndrome coronavirus 2 infection and skin diseases, there are 4 S100 family members located in the epidermal differentiation complex, and we also identified the "IL-17 signaling pathway" (P = 4.9 × 10-77) as one of the most significantly enriched pathways. Furthermore, a shared genome-wide significant locus in the epidermal differentiation complex was identified between psoriasis and severe acute respiratory syndrome coronavirus 2 infection, with the lead marker being a significant expression quantitative trait locus for S100A12 (P = 3.3 × 10-7). CONCLUSIONS Together our findings suggest association between inflammatory skin conditions and higher risk of COVID-19, but with less severe course, and highlight shared components involved in anti-COVID-19 immune response.
Collapse
Affiliation(s)
- Matthew T Patrick
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Mich.
| | - Haihan Zhang
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Mich
| | - Rachael Wasikowski
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Mich
| | - Errol P Prens
- Department of Dermatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Stephan Weidinger
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Johann E Gudjonsson
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Mich
| | - James T Elder
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Mich; Ann Arbor Veterans Affairs Hospital, Ann Arbor, Mich
| | - Kevin He
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Mich
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Mich; Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Mich; Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, Mich.
| |
Collapse
|
40
|
Moosbrugger-Martinz V, Hackl H, Gruber R, Pilecky M, Knabl L, Orth-Höller D, Dubrac S. Initial Evidence of Distinguishable Bacterial and Fungal Dysbiosis in the Skin of Patients with Atopic Dermatitis or Netherton Syndrome. J Invest Dermatol 2021; 141:114-123. [PMID: 32553662 DOI: 10.1016/j.jid.2020.05.102] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 04/29/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
Atopic dermatitis (AD) is an inflammatory skin disease in which epidermal barrier impairment, often owing to FLG null mutations, precedes immune hyperresponsiveness. Ichthyosis vulgaris is characterized by FLG null mutations and noninflamed dry skin. Netherton syndrome (NS), caused by SPINK5 null mutations, is characterized by generalized erythroderma with scaling and atopic manifestations. The goal of this work was to evaluate associations between specific skin disease features, such as ichthyotic and/or atopic manifestations, and the skin bacterial and fungal microbiota. Taxon diversity showed greater variation in the bacterial microbiota than in the fungal microbiota in the skin diseases. The relative abundances of Firmicutes (Staphylococcus) and Actinobacteria (Corynebacterium) were augmented in ichthyosis vulgaris, AD, and NS, whereas those of Proteobacteria/Enhydrobacter and Bacteroidetes were reduced, regardless of body site. Furthermore, proportions of Staphylococcus were correlated with transepidermal water loss and serum IgE levels. Nevertheless, the skin of patients with low to mild AD was overcolonized with Staphylococcus epidermidis and not with Staphylococcus aureus. Ascomycota were increased in both AD and NS, but from expansion of different fungal species. Finally, the expansion of pathologic bacteria in AD and NS might be supported by surrounding fungi. Thus, distinguishable bacterial and fungal skin dysbiosis in AD, NS, and ichthyosis vulgaris emphasizes disease-specific pathomechanisms.
Collapse
Affiliation(s)
- Verena Moosbrugger-Martinz
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hubert Hackl
- Division of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Robert Gruber
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Matthias Pilecky
- Center for Biomedical Technology, Department for Health Sciences and Biomedicine, Danube University Krems, Krems, Austria
| | - Ludwig Knabl
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dorothea Orth-Höller
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
41
|
Yiu ZZN, Parisi R, Lunt M, Warren RB, Griffiths CEM, Langan SM, Ashcroft DM. Risk of hospitalization and death due to infection in people with psoriasis: a population-based cohort study using the Clinical Practice Research Datalink. Br J Dermatol 2021; 184:78-86. [PMID: 32222069 DOI: 10.1111/bjd.19052] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Psoriasis is associated with risk factors for serious infections, but the independent relationship between psoriasis and serious infection is as yet unclear. OBJECTIVES To determine whether people with psoriasis have a higher risk of hospitalization due to any infection, respiratory infections, soft-tissue and skin infections, or a higher risk of death due to infection. METHODS We conducted a cohort study of people (≥ 18 years) with psoriasis using the UK Clinical Practice Research Datalink (CPRD GOLD) linked to Hospital Episode Statistics (HES) and Office for National Statistics (ONS) mortality records between 1 April 2003 and 31 December 2016, and matched with up to six comparators on age, sex and general practice. Hospitalization was ascertained from HES records; death was ascertained from ONS mortality records. Stratified Cox proportional hazard models were estimated, with stepwise adjustment in different models for potential confounders or mediators between psoriasis and serious infection. RESULTS There were 69 315 people with psoriasis and 338 620 comparators who were followed up for a median (interquartile range) of 4·9 (5·9) and 5·1 (6·3) years, respectively. People with psoriasis had a higher incidence rate of serious infection [20·5 per 1000 person-years, 95% confidence interval (CI) 20·0-21·0, n = 7631] compared with those without psoriasis (16·1 per 1000 person-years, 95% CI 15·9-16·3, n = 30 761). The fully adjusted hazard ratio for the association between psoriasis and serious infection was 1·36 (95% CI 1·31-1·40), with similar results across the other outcomes. CONCLUSIONS Psoriasis is associated with a small increase in the risk of serious infection. Further research is needed to understand how psoriasis predisposes to a higher risk of infection.
Collapse
Affiliation(s)
- Z Z N Yiu
- Dermatology Centre, Salford Royal NHS Foundation Trust, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - R Parisi
- Division of Informatics, Imaging & Data Sciences, School of Health Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - M Lunt
- Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, School of Biological Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - R B Warren
- Dermatology Centre, Salford Royal NHS Foundation Trust, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - C E M Griffiths
- Dermatology Centre, Salford Royal NHS Foundation Trust, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - S M Langan
- Faculty of Epidemiology, and Population Health, London School of Hygiene and Tropical Medicine, London, UK.,Health Data Research UK
| | - D M Ashcroft
- Centre for Pharmacoepidemiology and Drug Safety, School of Health Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
42
|
Paluch Z, Biriczová L, Pallag G, Carvalheiro Marques E, Vargová N, Kmoníčková E. The therapeutic effects of Agrimonia eupatoria L. Physiol Res 2020; 69:S555-S571. [PMID: 33646008 DOI: 10.33549/physiolres.934641] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Agrimonia eupatoria L. is an herb of the Rosaceae family, widely used in traditional (folk) medicine for its beneficial effects. Its water extracts (infusions and decoctions) are used in the treatment of airway and urinary system diseases, digestive tract diseases, and chronic wounds. Phytochemical analyses of Agrimonia eupatoria L. identified a variety of bioactive compounds including tannins, flavonoids, phenolic acids, triterpenoids and volatile oils possessing antioxidant, immunomodulatory and antimicrobial activities. The authors review the available literature sources examining and discussing the therapeutic and pharmacological effects of Agrimonia eupatoria L. at the molecular level in vitro and in vivo.
Collapse
Affiliation(s)
- Z Paluch
- Department of Pharmacology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
43
|
Chen L, Li J, Zhu W, Kuang Y, Liu T, Zhang W, Chen X, Peng C. Skin and Gut Microbiome in Psoriasis: Gaining Insight Into the Pathophysiology of It and Finding Novel Therapeutic Strategies. Front Microbiol 2020; 11:589726. [PMID: 33384669 PMCID: PMC7769758 DOI: 10.3389/fmicb.2020.589726] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Psoriasis affects the health of myriad populations around the world. The pathogenesis is multifactorial, and the exact driving factor remains unclear. This condition arises from the interaction between hyperproliferative keratinocytes and infiltrating immune cells, with poor prognosis and high recurrence. Better clinical treatments remain to be explored. There is much evidence that alterations in the skin and intestinal microbiome play an important role in the pathogenesis of psoriasis, and restoration of the microbiome is a promising preventive and therapeutic strategy for psoriasis. Herein, we have reviewed recent studies on the psoriasis-related microbiome in an attempt to confidently identify the “core” microbiome of psoriasis patients, understand the role of microbiome in the pathogenesis of psoriasis, and explore new therapeutic strategies for psoriasis through microbial intervention.
Collapse
Affiliation(s)
- Lihui Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Jie Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Wu Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Yehong Kuang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Tao Liu
- Central Laboratory, Shenzhen Center for Chronic Disease Control and Prevention, Shenzhen, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China
| |
Collapse
|
44
|
Nørreslet LB, Edslev SM, Andersen PS, Plum F, Holt J, Kjerulf A, Ebbehøj NE, Clausen ML, Flachs EM, Agner T. Colonization with Staphylococcus aureus in patients with hand eczema: Prevalence and association with severity, atopic dermatitis, subtype and nasal colonization. Contact Dermatitis 2020; 83:442-449. [PMID: 32720317 DOI: 10.1111/cod.13679] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/24/2020] [Accepted: 07/26/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND While Staphylococcus aureus (S. aureus) colonization has been thoroughly studied in atopic dermatitis (AD), where S. aureus is related to flares and considered a trigger factor, S. aureus colonization in hand eczema (HE) has only been sparsely studied. OBJECTIVES To examine the 1-week prevalence of S. aureus colonization in HE patients, and its association with severity, HE subtype, AD, and nasal S. aureus colonization compared with healthy controls. METHODS In a case-control study of 50 adult HE patients and 50 healthy controls, bacterial swabs from lesional skin (patients only), non-lesional skin (dorsal hand), and the nasal cavity were sampled for culturing of S. aureus on days 1, 3, 5 and 8. Participants were characterized by demographics, AD, HE subtype, filaggrin gene mutation status, and HE severity. RESULTS Twenty-seven HE patients (54%) were colonized with S. aureus on the hand compared to one control (2%) (P < .01). Nasal S. aureus colonization was found in 72% of patients and 22% of controls (P < .01). For patients, S. aureus colonization on the hands was associated with an atopic HE subtype and HE severity (P = .01 and P < .01, respectively). CONCLUSIONS Both hand and nasal S. aureus colonization were highly prevalent among HE-patients and may have an impact on the persistence of HE.
Collapse
Affiliation(s)
- Line B Nørreslet
- Department of Dermatology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Sofie M Edslev
- Department of Bacteria, Parasites, and Fungi, Statens Serum Institute, Copenhagen, Denmark
| | - Paal S Andersen
- Department of Bacteria, Parasites, and Fungi, Statens Serum Institute, Copenhagen, Denmark
| | - Frederik Plum
- Department of Dermatology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jette Holt
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institute, Copenhagen, Denmark
| | - Anne Kjerulf
- Department of Infectious Disease Epidemiology and Prevention, Statens Serum Institute, Copenhagen, Denmark
| | - Niels E Ebbehøj
- Department of Occupational and Environmental Medicine, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Maja-Lisa Clausen
- Department of Dermatology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Esben M Flachs
- Department of Occupational and Environmental Medicine, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Tove Agner
- Department of Dermatology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
45
|
Hashida Y, Higuchi T, Tanaka M, Shibata Y, Nakajima K, Sano S, Daibata M. Prevalence and Viral Loads of Cutaneous Human Polyomaviruses in the Skin of Patients With Chronic Inflammatory Skin Diseases. J Infect Dis 2020; 219:1564-1573. [PMID: 30357388 DOI: 10.1093/infdis/jiy618] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/21/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Human skin microorganisms have been associated with various skin diseases. However, most studies have focused on bacterial communities, and little is known about normally resident skin viruses such as the Polyomaviridae and their association with cutaneous disorders. METHODS We investigated the infection levels of Merkel cell polyomavirus (MCPyV), human polyomavirus 6 (HPyV6), and human polyomavirus 7 (HPyV7), using triplet skin swabs collected from lesional and nonlesional skins of 86 Japanese patients with inflammatory skin diseases and mycosis fungoides, and from 149 healthy control individuals. RESULTS This age-matched case-control study provides the first analyses of the loads of polyomaviruses in association with various skin diseases. The viral loads were significantly higher for HPyV6/HPyV7 and lower for MCPyV in patients with psoriasis. The viral load variation was observed not only at lesion sites, but also at clinically unaffected skin sites in most of the patients. The viral strains tested were all of the Asian/Japanese genotype. CONCLUSIONS Our findings suggest a covariation in the infection levels of cutaneous polyomaviruses in certain inflammatory skin conditions. Worldwide prospective longitudinal studies are warranted to understand the influence of such alterations on the pathogenesis of inflammatory skin disorders.
Collapse
Affiliation(s)
- Yumiko Hashida
- Department of Microbiology and Infection, Kochi Medical School, Kochi University, Japan
| | - Tomonori Higuchi
- Department of Microbiology and Infection, Kochi Medical School, Kochi University, Japan
| | - Moe Tanaka
- Department of Microbiology and Infection, Kochi Medical School, Kochi University, Japan
| | - Yuka Shibata
- Department of Dermatology, Kochi Medical School, Kochi University, Japan
| | - Kimiko Nakajima
- Department of Dermatology, Kochi Medical School, Kochi University, Japan
| | - Shigetoshi Sano
- Department of Dermatology, Kochi Medical School, Kochi University, Japan
| | - Masanori Daibata
- Department of Microbiology and Infection, Kochi Medical School, Kochi University, Japan
| |
Collapse
|
46
|
Ellis SR, Nguyen M, Vaughn AR, Notay M, Burney WA, Sandhu S, Sivamani RK. The Skin and Gut Microbiome and Its Role in Common Dermatologic Conditions. Microorganisms 2019; 7:microorganisms7110550. [PMID: 31717915 PMCID: PMC6920876 DOI: 10.3390/microorganisms7110550] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 10/27/2019] [Accepted: 11/06/2019] [Indexed: 02/08/2023] Open
Abstract
Microorganisms inhabit various areas of the body, including the gut and skin, and are important in maintaining homeostasis. Changes to the normal microflora due to genetic or environmental factors can contribute to the development of various disease states. In this review, we will discuss the relationship between the gut and skin microbiome and various dermatological diseases including acne, psoriasis, rosacea, and atopic dermatitis. In addition, we will discuss the impact of treatment on the microbiome and the role of probiotics.
Collapse
Affiliation(s)
- Samantha R. Ellis
- PotozkinMD Skincare Center, Danville, CA 94526, USA;
- Department of Dermatology, University of California-Davis, Sacramento, CA 95816, USA; (A.R.V.); (M.N.); (W.A.B.)
| | - Mimi Nguyen
- School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (M.N.); (S.S.)
| | - Alexandra R. Vaughn
- Department of Dermatology, University of California-Davis, Sacramento, CA 95816, USA; (A.R.V.); (M.N.); (W.A.B.)
| | - Manisha Notay
- Department of Dermatology, University of California-Davis, Sacramento, CA 95816, USA; (A.R.V.); (M.N.); (W.A.B.)
| | - Waqas A. Burney
- Department of Dermatology, University of California-Davis, Sacramento, CA 95816, USA; (A.R.V.); (M.N.); (W.A.B.)
- Department of Biological Sciences, California State University, Sacramento, CA 95819, USA
| | - Simran Sandhu
- School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (M.N.); (S.S.)
| | - Raja K. Sivamani
- Department of Dermatology, University of California-Davis, Sacramento, CA 95816, USA; (A.R.V.); (M.N.); (W.A.B.)
- Department of Biological Sciences, California State University, Sacramento, CA 95819, USA
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
- Pacific Skin Institute, Sacramento, CA 95815, USA
- Zen Dermatology, Sacramento, CA 95819, USA
- Correspondence:
| |
Collapse
|
47
|
Tsai YC, Tsai TF. A review of antibiotics and psoriasis: induction, exacerbation, and amelioration. Expert Rev Clin Pharmacol 2019; 12:981-989. [PMID: 31498683 DOI: 10.1080/17512433.2019.1665027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Psoriasis is affected by many environmental factors, including infections and antibiotics. However, the relationship between antibiotics and psoriasis is inadequately studied. Some antibiotics were listed as triggering factors; others showed benefit for psoriasis control. The aim of this article is to review current evidence that may help identify appropriate antibiotics for patients with psoriasis. Areas covered: The PubMed, Embase, Clinicalkey databases, and google scholar were searched for relevant articles published up to May 2019. Literature regarding antibiotics and psoriasis were included. Six randomized controlled trials and four controlled or cohort studies were identified in 13 kinds of antibiotics. Expert opinion: Macrolides and rifampin showed decrease of psoriasis area and severity index score in plaque-type psoriasis, while penicillin revealed no statistically significant improvement in guttate psoriasis. Previously tetracyclines were considered as triggering factors, but data were found only in cases or retrospective studies. Mechanisms were thought to be related to immunomodulation rather than bacteria inhibition. Research gap in the influence of genetic susceptibility, the impact on microbiota, and the mode of actions remain to be investigated.
Collapse
Affiliation(s)
- Ya-Chu Tsai
- Department of Dermatology, Far Eastern Memorial Hospital , New Taipei , Taiwan
| | - Tsen-Fang Tsai
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine , Taipei , Taiwan
| |
Collapse
|
48
|
Sirobhushanam S, Parsa N, Reed TJ, Kahlenberg JM. Chromagar™ requires secondary confirmation strategies to minimize false positive/negative results for detection of Staphylococcus aureus. J Microbiol Methods 2019; 161:71-73. [PMID: 31014957 PMCID: PMC6534454 DOI: 10.1016/j.mimet.2019.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/18/2019] [Accepted: 04/18/2019] [Indexed: 10/27/2022]
Abstract
Chromagar is a medium that is used in culture-based colonization studies of Staphylococcus aureus. We have found that S. aureus negative colonies often fit the color recommendations for S. aureus identification and can cause overestimation of colonization rates. Confirmation of suspect colonies is important to minimize false negative/positive results.
Collapse
Affiliation(s)
- Sirisha Sirobhushanam
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | - Tamra J Reed
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - J Michelle Kahlenberg
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
49
|
Triplett KD, Pokhrel S, Castleman MJ, Daly SM, Elmore BO, Joyner JA, Sharma G, Herbert G, Campen MJ, Hathaway HJ, Prossnitz ER, Hall PR. GPER activation protects against epithelial barrier disruption by Staphylococcus aureus α-toxin. Sci Rep 2019; 9:1343. [PMID: 30718654 PMCID: PMC6362070 DOI: 10.1038/s41598-018-37951-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 12/07/2018] [Indexed: 02/07/2023] Open
Abstract
Sex bias in innate defense against Staphylococcus aureus skin and soft tissue infection (SSTI) is dependent on both estrogen production by the host and S. aureus secretion of the virulence factor, α-hemolysin (Hla). The impact of estrogen signaling on the immune system is most often studied in terms of the nuclear estrogen receptors ERα and ERβ. However, the potential contribution of the G protein-coupled estrogen receptor (GPER) to innate defense against infectious disease, particularly with respect to skin infection, has not been addressed. Using a murine model of SSTI, we found that GPER activation with the highly selective agonist G-1 limits S. aureus SSTI and Hla-mediated pathogenesis, effects that were absent in GPER knockout mice. Specifically, G-1 reduced Hla-mediated skin lesion formation and pro-inflammatory cytokine production, while increasing bacterial clearance. In vitro, G-1 reduced surface expression of the Hla receptor, ADAM10, in a human keratinocyte cell line and increased resistance to Hla-mediated permeability barrier disruption. This novel role for GPER activation in skin innate defense against infectious disease suggests that G-1 may have clinical utility in patients with epithelial permeability barrier dysfunction or who are otherwise at increased risk of S. aureus infection, including those with atopic dermatitis or cancer.
Collapse
Affiliation(s)
- Kathleen D Triplett
- University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA
| | - Srijana Pokhrel
- University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA
| | - Moriah J Castleman
- University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA
| | - Seth M Daly
- University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA
| | - Bradley O Elmore
- University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA
| | - Jason A Joyner
- University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA
| | - Geetanjali Sharma
- University of New Mexico School of Medicine, Department of Internal Medicine, Albuquerque, NM, 87131, USA
| | - Guy Herbert
- University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA
| | - Matthew J Campen
- University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA
| | - Helen J Hathaway
- University of New Mexico School of Medicine, Department of Cell Biology & Physiology, Albuquerque, NM, 87131, USA
| | - Eric R Prossnitz
- University of New Mexico School of Medicine, Department of Internal Medicine, Albuquerque, NM, 87131, USA
| | - Pamela R Hall
- University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA.
| |
Collapse
|
50
|
Liu H, Archer NK, Dillen CA, Wang Y, Ashbaugh AG, Ortines RV, Kao T, Lee SK, Cai SS, Miller RJ, Marchitto MC, Zhang E, Riggins DP, Plaut RD, Stibitz S, Geha RS, Miller LS. Staphylococcus aureus Epicutaneous Exposure Drives Skin Inflammation via IL-36-Mediated T Cell Responses. Cell Host Microbe 2018; 22:653-666.e5. [PMID: 29120743 DOI: 10.1016/j.chom.2017.10.006] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/16/2017] [Accepted: 09/29/2017] [Indexed: 11/16/2022]
Abstract
Staphylococcus aureus colonization contributes to skin inflammation in diseases such as atopic dermatitis, but the signaling pathways involved are unclear. Herein, epicutaneous S. aureus exposure to mouse skin promoted MyD88-dependent skin inflammation initiated by IL-36, but not IL-1α/β, IL-18, or IL-33. By contrast, an intradermal S. aureus challenge promoted MyD88-dependent host defense initiated by IL-1β rather than IL-36, suggesting that different IL-1 cytokines trigger MyD88 signaling depending on the anatomical depth of S. aureus cutaneous exposure. The bacterial virulence factor PSMα, but not α-toxin or δ-toxin, contributed to the skin inflammation, which was driven by IL-17-producing γδ and CD4+ T cells via direct IL-36R signaling in the T cells. Finally, adoptive transfer of IL-36R-expressing T cells to IL-36R-deficient mice was sufficient for mediating S. aureus-induced skin inflammation. Together, this study defines a previously unknown pathway by which S. aureus epicutaneous exposure promotes skin inflammation involving IL-36R/MyD88-dependent IL-17 T cell responses.
Collapse
Affiliation(s)
- Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Carly A Dillen
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Alyssa G Ashbaugh
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Roger V Ortines
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Tracy Kao
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Steven K Lee
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Shuting S Cai
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Robert J Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Mark C Marchitto
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Emily Zhang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Daniel P Riggins
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20852, USA
| | - Roger D Plaut
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20852, USA
| | - Scott Stibitz
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20852, USA
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|