101
|
Dysregulation of the gut-brain-skin axis and key overlapping inflammatory and immune mechanisms of psoriasis and depression. Biomed Pharmacother 2021; 137:111065. [PMID: 33540138 DOI: 10.1016/j.biopha.2020.111065] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/14/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022] Open
Abstract
The occurrence, progression and recurrence of psoriasis are thought to be related to mood and psychological disorders such as depression. Psoriasis can lead to depression, and depression, in turn, exacerbates psoriasis. No specific mechanism can explain the association between psoriasis and depression. The gut-brain-skin axis has been used to explain correlations among the gut microbiota, emotional states and systemic and skin inflammation, and this axis may be associated with overlapping mechanisms between psoriasis and depression. Therefore, in the context of the gut-brain-skin axis, we systematically summarized and comparatively analysed the inflammatory and immune mechanisms of psoriasis and depression and illustrated the dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis and the gut microbiota. This review provides a theoretical basis and new targets for the treatment of psoriasis and depression.
Collapse
|
102
|
Valentini V, Silvestri V, Marraffa F, Greco G, Bucalo A, Grassi S, Gagliardi A, Mazzotta A, Ottini L, Richetta AG. Gut microbiome profile in psoriatic patients treated and untreated with biologic therapy. J Dermatol 2021; 48:786-793. [PMID: 33511673 DOI: 10.1111/1346-8138.15680] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/09/2020] [Indexed: 01/06/2023]
Abstract
There are increasing data about the role of the gut microbiome in various autoimmune diseases, including psoriasis, a chronic inflammatory and immune-mediated disease. Current treatment strategies in psoriasis include immunomodulating biologic agents. A variable response to this type of therapy has been reported in psoriatic patients. A possible effect of biologic therapy on the gut microbiome composition has been suggested, but data are still limited. The aim of this study was to compare the gut microbiome composition between psoriatic patients treated and untreated with biologic drugs in order to identify differences which may highlight the potential impact of the treatment on the gut microbiome. 16S rRNA sequencing and bioinformatic analyses were performed on the fecal samples of 30 psoriatic patients with similar clinicopathological features, 10 of whom were undergoing biologic therapy and 20 not receiving systemic therapy. Alpha and beta diversity significantly differed between the two groups of patients. A reduced bacterial biodiversity in the group of treated patients compared with the group of untreated patients was observed. Differential relative abundances of key gut microbial communities, including Akkermansia muciniphila and Bacteroides plebeius, were identified between the two groups of patients. This study showed that biologic therapy may have an impact on the composition of the gut microbiome of psoriatic patients. Gut microbiome composition could be used as an indicator of response to therapy and the modulation of the microbial composition could help to restore the intestinal symbiosis in psoriatic patients.
Collapse
Affiliation(s)
- Virginia Valentini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Federica Marraffa
- Unit of Dermatology, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Giulia Greco
- Unit of Dermatology, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Agostino Bucalo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Sara Grassi
- Unit of Dermatology, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Antonella Gagliardi
- Section of Microbiology, Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | | | - Laura Ottini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonio Giovanni Richetta
- Unit of Dermatology, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
103
|
Myers B, Vidhatha R, Nicholas B, Stephanie C, Quinn T, Chang HW, Bhutani T, Liao W. Sleep and the gut microbiome in psoriasis: clinical implications for disease progression and the development of cardiometabolic comorbidities. JOURNAL OF PSORIASIS AND PSORIATIC ARTHRITIS 2021; 6:27-37. [PMID: 35784674 PMCID: PMC9249097 DOI: 10.1177/2475530320964781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Background Sleep dysfunction and sleep disorders are important comorbidities of psoriasis. Not only do these sleep comorbidities contribute to reduced quality of life, but they may also lead to worsening psoriasis and increased susceptibility to cardiometabolic diseases. While psoriasis and sleep dysfunction are thought to be linked by itch, depression, and immune system dysregulation, the relationship between psoriasis and sleep dysfunction is not yet fully understood. Objective We sought to compare previous studies characterizing the gut microbiome in psoriasis and sleep dysfunction and examine the potential relevance of shared findings on cardiometabolic and overall health. Methods We performed literature searches of PubMed and Embase databases to find studies evaluating the gut microbiome in psoriasis, sleep dysfunction, and cardiometabolic diseases. Results Studies characterizing the gut microbiome in psoriasis and sleep dysfunction reveal shared findings, specifically an increased Firmicutes to Bacteroidetes ratio and reduced abundance of short chain fatty acid-producing bacteria. These dysbiotic features have also been shown to promote systemic inflammation and cardiometabolic disease. Conclusion In favoring an increased Firmicutes to Bacteroidetes ratio and reduced abundance of short chain fatty acid-producing bacteria, sleep dysfunction could be contributing to worsening psoriasis and cardiometabolic comorbidities through intestinal dysbiosis. Future studies are needed to determine whether gut- and sleep-targeting interventions could be therapeutic in psoriasis patients with poor sleep.
Collapse
Affiliation(s)
- Bridget Myers
- University of California, San Francisco, Department of Dermatology
| | - Reddy Vidhatha
- University of California, San Francisco, Department of Dermatology
| | | | - Chan Stephanie
- University of California, San Francisco, Department of Dermatology
| | - Thibodeaux Quinn
- University of California, San Francisco, Department of Dermatology
| | - Hsin-Wen Chang
- University of California, San Francisco, Department of Dermatology
| | - Tina Bhutani
- University of California, San Francisco, Department of Dermatology
| | - Wilson Liao
- University of California, San Francisco, Department of Dermatology
| |
Collapse
|
104
|
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: 61] [Impact Index Per Article: 15.3] [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
|
105
|
Ni Q, Ye Z, Wang Y, Chen J, Zhang W, Ma C, Li K, Liu Y, Liu L, Han Z, Gao T, Jian Z, Li S, Li C. Gut Microbial Dysbiosis and Plasma Metabolic Profile in Individuals With Vitiligo. Front Microbiol 2020; 11:592248. [PMID: 33381090 PMCID: PMC7768019 DOI: 10.3389/fmicb.2020.592248] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
Autoimmune diseases are increasingly linked to aberrant gut microbiome and relevant metabolites. However, the association between vitiligo and the gut microbiome remains to be elucidated. Thus, we conducted a case-control study through 16S rRNA sequencing and serum untargeted-metabolomic profiling based on 30 vitiligo patients and 30 matched healthy controls. In vitiligo patients, the microbial composition was distinct from that of healthy controls according to the analysis on α- and β-diversity (P < 0.05), with a characteristic decreased Bacteroidetes: Firmicutes ratio. Meanwhile, the levels of 23 serum metabolites (including taurochenodeoxycholate and L-NG-monomethyl-arginine) in the vitiligo patients were different from those in the healthy individuals and showed significant correlations with some microbial markers. We found that Corynebacterium 1, Ruminococcus 2, Jeotgalibaca and Psychrobacter were correlated significantly with disease duration and serum IL-1β level in vitiligo patients. And Psychrobacter was identified as the most predictive features for vitiligo by machine learning analysis ("importance" = 0.0236). Finally, combining multi-omics data and joint prediction models with accuracies up to 0.929 were established with dominant contribution of Corynebacterium 1 and Psychrobacter. Our findings replenished the previously unknown relationship between gut dysbiosis and vitiligo circulating metabolome and enrolled the gut-skin axis into the understanding of vitiligo pathogenesis.
Collapse
Affiliation(s)
- Qingrong Ni
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhubiao Ye
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yinghan Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jianru Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Weigang Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Cuiling Ma
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Kai Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yu Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ling Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zheyi Han
- Department of Gastroenterology, Xijing Hospital, Fourth Military Medical University, Xi'an, China; National Clinical Research Center for Digestive Diseases, Xi'an, China
| | - Tianwen Gao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhe Jian
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shuli Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
106
|
Chen YJ, Ho HJ, Wu CY, Juan CK, Wu CY. Infantile infection and antibiotic exposure in association with pediatric psoriasis development: A nationwide nested case-control study. J Am Acad Dermatol 2020; 85:626-635. [PMID: 33321158 DOI: 10.1016/j.jaad.2020.12.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/05/2020] [Accepted: 12/03/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Microbiol dysbiosis and antibiotic exposure have been implicated in the pathogenesis of pediatric inflammatory diseases. OBJECTIVES To investigate the impacts of infantile infection and antibiotic exposure on pediatric psoriasis development. METHODS This is a nationwide nested case-control study. From the National Health Insurance Research Database of Taiwan, a total of 1527 patients with pediatric psoriasis were identified and matched with 15,270 reference individuals without psoriasis, for the period of 2000 to 2017. Demographic characteristics and comorbidities were compared. Conditional stepwise logistic regression analysis was conducted to examine the associations. RESULTS The mean ages were 9.9 ± 3.7 years in both groups. Atopic dermatitis (adjusted odds ratio [aOR], 2.07; 95% confidence interval [CI], 1.84-2.32) and family history of psoriasis, especially of the mother (aOR, 9.86; 95% CI, 6.89-14.10) or other first-degree relatives (aOR, 5.49; 95% CI, 3.91-7.70), were independently associated with pediatric psoriasis on multivariate analyses. Skin viral and bacterial infections (aOR, 1.35; 95% CI, 1.13-1.62) and fungal infections (aOR, 1.71; 95% CI, 1.44-2.04) in the first 2 years of life were significantly associated with pediatric psoriasis. Systemic antibiotic exposure was not. These results were consistent at different time periods across sensitivity analyses. LIMITATION Information about diet and lifestyle was not available. CONCLUSION Skin infections at an early age were associated with pediatric psoriasis development.
Collapse
Affiliation(s)
- Yi-Ju Chen
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan; Faculty of Medicine and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Taiwan Microbiota Consortium, Taipei, Taiwan.
| | - Hsiu J Ho
- Taiwan Microbiota Consortium, Taipei, Taiwan; Institute of Biomedical Informatics, Institute of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Chen-Yi Wu
- Taiwan Microbiota Consortium, Taipei, Taiwan; Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Chao-Kuei Juan
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan; Faculty of Medicine and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chun-Ying Wu
- Faculty of Medicine and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Taiwan Microbiota Consortium, Taipei, Taiwan; Institute of Biomedical Informatics, Institute of Public Health, National Yang-Ming University, Taipei, Taiwan; Division of Translational Research and Center of Excellence for Cancer Research, Taipei Veterans General Hospital, Taipei, Taiwan; College of Public Health, China Medical University, Taichung, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan.
| |
Collapse
|
107
|
Chen G, Chen ZM, Fan XY, Jin YL, Li X, Wu SR, Ge WW, Lv CH, Wang YK, Chen JG. Gut-Brain-Skin Axis in Psoriasis: A Review. Dermatol Ther (Heidelb) 2020; 11:25-38. [PMID: 33206326 PMCID: PMC7859123 DOI: 10.1007/s13555-020-00466-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction Psoriasis is a common skin disease, with chronic inflammation and a complex etiology. It has long been recognized that chronic skin conditions and mental health disorders are often co-morbid. Thus, the concept of the gut–brain–skin axis emphasized in mental health disorders may also regulate the health of skin. Results The gut microbiota has been found to be the bridge between the immune system and nervous system. By leveraging clinical cases and animal models of psoriasis, an important communication pathway has been identified along the gut–brain–skin axis that is associated with the modulation of neurotransmitters from the microbiota. Furthermore, mammalian neurotransmitters, including dopamine, serotonin, or γ-aminobutyric acid (GABA), can be produced and/or consumed by several types of bacteria. Other studies suggest that manipulating these neurotransmitters by bacteria may have an effect on host physiology, and the levels of neurotransmitter can be altered by microbiota-based interventions. Conclusions Nonetheless, it is unknown whether or not the manipulation of neurotransmitter levels by bacteria can affect the occurrence and development of psoriasis. Notably, preliminary experiments found that oral consumption of probiotics improves the clinical symptoms in patients with psoriasis, perhaps correlated with the gut microbiome-mediated crosstalk between the immune system and the nervous system by secreting neurotransmitters in psoriasis. In this review, the communication along the gut–brain–skin axis is discussed.
Collapse
Affiliation(s)
- Guang Chen
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, China.,Department of Dermatology, Taizhou Second People's Hospital, No 2 Shuinan east road, Tiantai Country, Taizhou, China
| | - Zai-Ming Chen
- Department of Dermatology, Taizhou Second People's Hospital, No 2 Shuinan east road, Tiantai Country, Taizhou, China
| | - Xiao-Yan Fan
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, China
| | - Yue-Lei Jin
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, China
| | - Xin Li
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, China.,Department of Medicine, Jiamusi University, No 148 Xuefu road, Xiangyang District, Jiamusi, China
| | - Shi-Ren Wu
- Department of Dermatology, Taizhou Second People's Hospital, No 2 Shuinan east road, Tiantai Country, Taizhou, China
| | - Wei-Wei Ge
- Department of Dermatology, Taizhou Second People's Hospital, No 2 Shuinan east road, Tiantai Country, Taizhou, China
| | - Cao-Hua Lv
- Department of Dermatology, Taizhou Second People's Hospital, No 2 Shuinan east road, Tiantai Country, Taizhou, China
| | - Yao-Kun Wang
- Department of Medicine, Jiamusi University, No 148 Xuefu road, Xiangyang District, Jiamusi, China
| | - Jin-Guang Chen
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, China.
| |
Collapse
|
108
|
Gabashvili IS. Cutaneous Bacteria in the Gut Microbiome as Biomarkers of Systemic Malodor and People Are Allergic to Me (PATM) Conditions: Insights From a Virtually Conducted Clinical Trial. JMIR DERMATOLOGY 2020. [DOI: 10.2196/10508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background
The skin is a dynamic ecosystem of microbes and the source of many chemical compounds that affect human health. Skin-microbiome interactions can cause persistent, psychosocially devastating body smell despite good hygiene. Since odor production is often transient, malodors may not be perceptible during medical examinations. Therefore, having odor complaints can be diagnosed as body dysmorphic disorder and referred for psychological evaluations. Development of simple at-home tests and virtual care programs could improve the diagnosis and management of socially debilitating malodor conditions.
Objective
The aim of this study was to assess potential effectiveness of at-home gut microbiome testing in the diagnosis and management of idiopathic body and breath odor and in people are allergic to me (PATM) syndrome.
Methods
We contacted participants of prior metabolic body odor (MEBO) and PATM studies and online support groups by email or social media. Individuals who consented to participate were mailed test kits for at-home collection of gut microbiome samples. Participants completed an online survey (specially developed for this study) addressing their symptoms and other quality-of-life indicators at baseline and after sampling. Participants collected stool samples after flare-ups or symptom improvements and mailed them to the laboratory to be processed and analyzed. We evaluated between-group differences in symptom severity, as well as symptom improvement observations for the same individuals. For differential abundance testing of microbial taxa, we performed nonparametric statistical analyses using Mann-Whitney U tests for unpaired samples and Wilcoxon signed rank test for paired samples.
Results
A total of 112 individuals from 21 countries consented to participate. About half the participants had been tested for the metabolic disorder trimethylaminuria, and about half of those tested were diagnosed with the disorder. The levels of bacteria previously associated with cutaneous body odor were significantly elevated in gut samples. For the combination of species from Anaerococcus, Corynebacterium, Campylobacter, and Propionibacterium genera, the differences were P=.002 for active (73 participants, 182 samples) versus regression or remission groups (30 participants, 51 samples); P=.01 for those experiencing symptoms most or all of the time (46 participants, 88 samples) versus those who had symptoms sometimes, rarely, or never (25 participants, 74 samples); and P<.001 for improvement of symptoms in the same individuals (22 participants, 43 sets of matched samples). Changes in microbial diversity were significant for between- but not within-participant comparisons.
Conclusions
Changes in the gut microbiome composition affect MEBO and PATM severity. In particular, an increase in intestinal bacteria producing odor when in skin flexures was associated with increased intensity of self-reported symptoms. The changes were consistent in the within-group and between-group analyses. Our findings support the feasibility of remote and decentralized clinical studies of malodor conditions. Supplementary sample collection procedures may help to meet established research quality standards.
Trial Registration
ClinicalTrials.gov NCT03582826; http://clinicaltrials.gov/ct2/show/NCT03582826
International Registered Report Identifier (IRRID)
RR2-10.1101/2020.08.21.20179242
Collapse
|
109
|
Raftery AL, Tsantikos E, Harris NL, Hibbs ML. Links Between Inflammatory Bowel Disease and Chronic Obstructive Pulmonary Disease. Front Immunol 2020; 11:2144. [PMID: 33042125 PMCID: PMC7517908 DOI: 10.3389/fimmu.2020.02144] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) and chronic obstructive pulmonary disease (COPD) are chronic inflammatory diseases of the gastrointestinal and respiratory tracts, respectively. These mucosal tissues bear commonalities in embryology, structure and physiology. Inherent similarities in immune responses at the two sites, as well as overlapping environmental risk factors, help to explain the increase in prevalence of IBD amongst COPD patients. Over the past decade, a tremendous amount of research has been conducted to define the microbiological makeup of the intestine, known as the intestinal microbiota, and determine its contribution to health and disease. Intestinal microbial dysbiosis is now known to be associated with IBD where it impacts upon intestinal epithelial barrier integrity and leads to augmented immune responses and the perpetuation of chronic inflammation. While much less is known about the lung microbiota, like the intestine, it has its own distinct, diverse microflora, with dysbiosis being reported in respiratory disease settings such as COPD. Recent research has begun to delineate the interaction or crosstalk between the lung and the intestine and how this may influence, or be influenced by, the microbiota. It is now known that microbial products and metabolites can be transferred from the intestine to the lung via the bloodstream, providing a mechanism for communication. While recent studies indicate that intestinal microbiota can influence respiratory health, intestinal dysbiosis in COPD has not yet been described although it is anticipated since factors that lead to dysbiosis are similarly associated with COPD. This review will focus on the gut-lung axis in the context of IBD and COPD, highlighting the role of environmental and genetic factors and the impact of microbial dysbiosis on chronic inflammation in the intestinal tract and lung.
Collapse
Affiliation(s)
- April L Raftery
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Evelyn Tsantikos
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Nicola L Harris
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Margaret L Hibbs
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
110
|
Metagenomic analysis of gut microbiota in non-treated plaque psoriasis patients stratified by disease severity: development of a new Psoriasis-Microbiome Index. Sci Rep 2020; 10:12754. [PMID: 32728075 PMCID: PMC7391695 DOI: 10.1038/s41598-020-69537-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
Psoriasis is an immune-mediated skin disorder. Imbalance of gut microbial populations has been implicated in many diseases. We aimed to investigate whether there were differences in gut microbiota in psoriasis patients vs non-psoriasis controls and between psoriasis severity groups. 55 psoriasis patients and 27 controls were included. V3–V4 regions of the 16S rRNA gene of fecal samples were analyzed using Illumina MiSeq. Bioinformatic analysis was performed. We found changes in gut microbiome composition depending on their psoriasis status as determined by weighted unifrac (p < 0.05), in particular an increase in Firmicutes and depletion of Bacteroidetes in psoriasis patients. Additionally, the Faecalibacterium and Blautia genus were higher in psoriasis patients while Bacteroides and Paraprevotella in non-psoriasis controls (p < 0.05, LDA score > 2). Moderate-to-severe psoriasis patients had lower biodiversity than mild psoriatic patients (p = 0.049). No differences for beta-diversity were found. We developed a Psoriasis-Microbiota Index (PMI), which discriminated among psoriasis patients and controls with sensitivity: 0.78 and specificity: 0.79. Furthermore, we performed a meta-analysis with published data to validate this index. We demonstrated gut dysbiosis in psoriasis patients, suggesting a role in psoriasis pathophysiology. Furthermore, we developed a PMI with the potential to discriminate between psoriasis patients and controls across different populations, which could be used as a biomarker in the clinical practice.
Collapse
|
111
|
Mann EA, Bae E, Kostyuchek D, Chung HJ, McGee JS. The Gut Microbiome: Human Health and Inflammatory Skin Diseases. Ann Dermatol 2020; 32:265-272. [PMID: 33911753 PMCID: PMC7992658 DOI: 10.5021/ad.2020.32.4.265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/16/2019] [Accepted: 02/10/2020] [Indexed: 01/01/2023] Open
Abstract
The human microbiome is a rich environment consisting of bacteria, fungi and other commensal microorganisms of the gut, mucosa and skin. The functional role of the gut microbiome includes facilitation in metabolism of macronutrients, maturation of the immune system, and production of pro- or anti-inflammatory signaling molecules and peptides. The identification of these resident organisms has brought about a new understanding of disease processes. Nevertheless, more questions remain regarding the interactions within the microbiome, its interactions with the host, and its contributions to the pathophysiology of disease. The purpose of this review is to examine the existing medical literature to highlight the role of the gut microbiome in human health, also paying attention to its role in several inflammatory skin diseases, namely atopic dermatitis, psoriasis, and rosacea.
Collapse
Affiliation(s)
- Emily A Mann
- Department of Dermatology, Boston University School of Medicine, Boston, MA, USA
| | - Edward Bae
- Department of Dermatology, Boston University School of Medicine, Boston, MA, USA
| | - Darya Kostyuchek
- Department of Dermatology, Boston University School of Medicine, Boston, MA, USA
| | - Hye Jin Chung
- Department of Dermatology, Boston University School of Medicine, Boston, MA, USA
| | - Jean S McGee
- Department of Dermatology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
112
|
Sikora M, Stec A, Chrabaszcz M, Knot A, Waskiel-Burnat A, Rakowska A, Olszewska M, Rudnicka L. Gut Microbiome in Psoriasis: An Updated Review. Pathogens 2020; 9:pathogens9060463. [PMID: 32545459 PMCID: PMC7350295 DOI: 10.3390/pathogens9060463] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/29/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022] Open
Abstract
(1) Background: A growing body of evidence highlights that intestinal dysbiosis is associated with the development of psoriasis. The gut–skin axis is the novel concept of the interaction between skin diseases and microbiome through inflammatory mediators, metabolites and the intestinal barrier. The objective of this study was to synthesize current data on the gut microbial composition in psoriasis. (2) Methods: We conducted a systematic review of studies investigating intestinal microbiome in psoriasis, using the PRISMA checklist. We searched MEDLINE, EMBASE, and Web of Science databases for relevant published articles (2000–2020). (3) Results: All of the 10 retrieved studies reported alterations in the gut microbiome in patients with psoriasis. Eight studies assessed alpha- and beta-diversity. Four of them reported a lack of change in alpha-diversity, but all confirmed significant changes in beta-diversity. At the phylum-level, at least two or more studies reported a lower relative abundance of Bacteroidetes, and higher Firmicutes in psoriasis patients versus healthy controls. (4) Conclusions: There is a significant association between alterations in gut microbial composition and psoriasis; however, there is high heterogeneity between studies. More unified methodological standards in large-scale studies are needed to understand microbiota’s contribution to psoriasis pathogenesis and its modulation as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Mariusz Sikora
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
- Correspondence:
| | - Albert Stec
- Student Research Committee, Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (A.S.); (A.K.)
| | - Magdalena Chrabaszcz
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Aleksandra Knot
- Student Research Committee, Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (A.S.); (A.K.)
| | - Anna Waskiel-Burnat
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Adriana Rakowska
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Malgorzata Olszewska
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| |
Collapse
|
113
|
Okada K, Matsushima Y, Mizutani K, Yamanaka K. The Role of Gut Microbiome in Psoriasis: Oral Administration of Staphylococcus aureus and Streptococcus danieliae Exacerbates Skin Inflammation of Imiquimod-Induced Psoriasis-Like Dermatitis. Int J Mol Sci 2020; 21:E3303. [PMID: 32392785 PMCID: PMC7246652 DOI: 10.3390/ijms21093303] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 01/11/2023] Open
Abstract
Psoriasis is one of the common chronic inflammatory skin diseases in which inflammatory cytokines such as IL-17 and TNF-α play critical roles. Skin microbiome of psoriasis patients is reported to have elevated Staphylococcus and Streptococcus genus. There are controversial reports about gut microbiome of psoriasis patients, and whether the diversity of bacteria in genus level is decreased or not is still unclear. Moreover, it is not yet known if these gut bacteria would be the cause of the inflammation or the result of the inflammation. We analyzed the gut microbiome of the inflammatory skin model mouse (keratinocyte-specific caspase-1 transgenic (Kcasp1Tg) mouse), by analyzing the 16S rRNA gene. Staphylocuccus aureus and Streptococcus danieliae were abundant in Kcasp1Tg mouse fecal microbiome. These dominant bacteria as well as recessive control bacteria were orally administrated to antibiotic-treated wild type mice, and set up imiquimod-induced psoriasis-like skin inflammation model. The skin inflammation including ear thickness and histopathological findings was analyzed. The exacerbated skin lesions with the elevated levels of TNF-α, IL-17A, IL-17F, and IL-22 were observed in Staphylocuccus aureus and Streptococcus danieliae administrated groups. Our finding suggests that there is affinity between skin inflammation severity and certain gut bacteria leading to a vicious cycle: skin inflammation populates certain gut bacteria which itself worsens the skin inflammation. This is the first report on Staphylocuccus aureus and Streptococcuus danieliae effects in vivo. Not only treating the skin lesion but also treating the gut microbiome could be the future key treatment for inflammatory skin disease such as psoriasis.
Collapse
Affiliation(s)
| | | | | | - Keiichi Yamanaka
- Department of Dermatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan; (K.O.); (Y.M.); (K.M.)
| |
Collapse
|
114
|
Abstract
This review summarizes existing research on the gut microbiome composition and function in psoriasis and psoriatic arthritis, exploring potential roles in disease pathogenesis, progression, and management. A strong relationship between skin, joint, and gastrointestinal inflammation exists, as demonstrated by an increased prevalence of psoriasis, psoriatic arthritis, and inflammatory bowel disease co-occurring together; however, the link between them has not been fully elucidated. Studies analyzing the gut microbiome in psoriasis and psoriatic arthritis reveal a unique pattern of dysbiosis. With regard to the gut microbiome's role in psoriasis and psoriatic arthritis pathogenesis, we discuss several theories including intestinal permeability, altered immune homeostasis, and imbalance of short- and medium-chain fatty acid-producing bacteria. We also discuss how the gut microbiome affects patient risk of psoriatic arthritis and other serious comorbidities, and how fecal microbes could be used clinically as therapeutic targets or markers of disease.
Collapse
|
115
|
Recent Advances in Psoriasis Research; the Clue to Mysterious Relation to Gut Microbiome. Int J Mol Sci 2020; 21:ijms21072582. [PMID: 32276410 PMCID: PMC7177330 DOI: 10.3390/ijms21072582] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 12/13/2022] Open
Abstract
Psoriasis is a chronic inflammatory cutaneous disease, characterized by activated plasmacytoid dendritic cells, myeloid dendritic cells, Th17 cells, and hyperproliferating keratinocytes. Recent studies revealed skin-resident cells have pivotal roles in developing psoriatic skin lesions. The balance in effector T cells and regulatory T cells is disturbed, leading Foxp3-positive regulatory T cells to produce proinflammatory IL-17. Not only acquired but also innate immunity is important in psoriasis pathogenesis, especially in triggering the disease. Group 3 innate lymphoid cell are considered one of IL-17-producing cells in psoriasis. Short chain fatty acids produced by gut microbiota stabilize expression of Foxp3 in regulatory T cells, thereby stabilizing their function. The composition of gut microbiota influences the systemic inflammatory status, and associations been shown with diabetes mellitus, cardiovascular diseases, psychomotor diseases, and other systemic inflammatory disorders. Psoriasis has been shown to frequently comorbid with diabetes mellitus, cardiovascular diseases, psychomotor disease and obesity, and recent report suggested the similar abnormality in gut microbiota as the above comorbid diseases. However, the precise mechanism and relation between psoriasis pathogenesis and gut microbiota needs further investigation. This review introduces the recent advances in psoriasis research and tries to provide clues to solve the mysterious relation of psoriasis and gut microbiota.
Collapse
|
116
|
Abstract
Spondyloarthritis (SpA) is a group of chronic, inflammatory rheumatic diseases mainly affecting the axial skeleton. Although the pathogenesis of the disease remains elusive, alterations of intestinal microbial composition have been demonstrated in patients with SpA and associated with intestinal and systemic immune alterations. Substantial data have been published in recent years in ethnically different patient populations, demonstrating in a consolidated way the presence of alterations in the composition of the microbial flora in patients with SpA. It is not currently possible to establish whether these alterations are intrinsically inherent in the disease, for example, the effect of particular genes that confer susceptibility to the disease itself, or are a consequence of a more systemic inflammatory process that also involves the intestine. However, data deriving from animal models and studies on relatives of patients with SpA strongly suggest that these alterations might precede the onset of the disease. In this review, we will try to critically analyze studies on dysbiosis in SpA and animal models of SpA, analyzing their functional consequences and the impact of biotechnological therapies on intestinal bacterial composition.
Collapse
Affiliation(s)
- Daniele Mauro
- Department of Precision Medicine, Università Degli Studi Della Campania L. Vanvitelli, Naples, Italy
| | - Francesco Ciccia
- Department of Precision Medicine, Università Degli Studi Della Campania L. Vanvitelli, Naples, Italy.
| |
Collapse
|
117
|
Alesa DI, Alshamrani HM, Alzahrani YA, Alamssi DN, Alzahrani NS, Almohammadi ME. The role of gut microbiome in the pathogenesis of psoriasis and the therapeutic effects of probiotics. J Family Med Prim Care 2019; 8:3496-3503. [PMID: 31803643 PMCID: PMC6881942 DOI: 10.4103/jfmpc.jfmpc_709_19] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/17/2019] [Accepted: 10/04/2019] [Indexed: 12/17/2022] Open
Abstract
The adult intestine hosts a huge number of diverse bacterial species, collectively referred to as the microbiome, that reside mainly in the lower gut, where they maintain a symbiotic relationship with their host. Recent research points to a central role of the microbiome in many biological processes. These microbial communities are influenced by multiple environmental and dietary factors and can modulate immune responses. In addition to local effects on the gastrointestinal tract, the microbiota is associated with effects on other organs and tissues, such as the skin. Indeed, an altered microbiome has been associated with skin disorders in several instances. Thus, in this review, we describe the recent advances regarding the interplay between gut microbiota and the skin. We explore how this potential link affects skin homeostasis and its influence on modulating the cutaneous immune response, focusing on psoriasis disorder. Finally, we discuss how to take advantage of this interplay to manage this disorder, particularly through probiotics administration. In the gastrointestinal tract, the microbiome has been proven to be important in the maintenance of the balance between effector T cells and regulatory T cells, and the induction of immunoglobulin A. Moreover, gut bacterial dysbiosis is associated with chronic inflammatory disorders of the skin, such as psoriasis. Thus, the microbiome can be considered an effective therapeutical target for treating this disorder. Despite some limitations, interventions with probiotics seem promising for the development of a preventive therapy by restoring altered microbiome functionality or as an adjuvant in specific immunotherapy.
Collapse
Affiliation(s)
- Dalal I Alesa
- Dermatology Resident, Alnoor Specialist Hospital, Makkah, Saudi Arabia
| | | | - Yahya A Alzahrani
- Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Dania N Alamssi
- General Practitioner, Consultant Center for Dermatology and Venereology Clinics, Makkah, Saudi Arabia
| | - Nada S Alzahrani
- Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | | |
Collapse
|
118
|
Alexeyev O. Psoriasis, gut and microbiome. Br J Dermatol 2019; 181:1126. [PMID: 31571198 DOI: 10.1111/bjd.18467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- O Alexeyev
- Department of Pathology, Umeå University, 90185, Umeå, Sweden
| |
Collapse
|
119
|
Sikora M, Stec A, Chrabaszcz M, Waskiel-Burnat A, Zaremba M, Olszewska M, Rudnicka L. Intestinal Fatty Acid Binding Protein, a Biomarker of Intestinal Barrier, is Associated with Severity of Psoriasis. J Clin Med 2019; 8:jcm8071021. [PMID: 31336842 PMCID: PMC6678629 DOI: 10.3390/jcm8071021] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/04/2019] [Accepted: 07/11/2019] [Indexed: 12/18/2022] Open
Abstract
Alterations of intestinal microbiota play a significant role in the pathogenesis of psoriasis. Dysbiosis may cause disruption of the intestinal barrier, which contributes to immune activation by translocation of microbial antigens and metabolites. Intestinal fatty acid binding protein (I-FABP) serves as a biomarker of enterocyte damage. The aim of this study was to investigate clinical and metabolic factors affecting plasma concentration of I-FABP in patients with psoriasis. Eighty patients with psoriasis and 40 control subjects were enrolled in the study. Serum I-FABP (243.00 (108.88–787.10) vs. 114.38 (51.60–241.60) pg/ml, p < 0.001) and neutrophil to lymphocyte ratio (NLR; 2.59 (1.96–3.09) vs. 1.72 (1.36–47 2.11), p < 0.01) were significantly increased in patients with psoriasis compared to controls. A significant positive correlation was found between I-FABP and body mass index (BMI) (r = 0.82, p < 0.001), Psoriasis Area Severity Index (PASI) (r = 0.78, p < 0.001) and neutrophil to lymphocyte ratio (NLR) (r = 0.24, p < 0.001). Rising quartiles of I-FABP were associated with increasing values of BMI, PASI and NLR. The results of the logistic regression model confirmed an increased risk of higher disease severity with I-FABP concentration – odds ratio 3.34 per 100 pg/mL I-FABP increase. In conclusion, intestinal integrity in patients with psoriasis is affected by obesity, severity of the disease and systemic inflammation. The modulation of gut barrier may represent a new therapeutic approach for psoriasis.
Collapse
Affiliation(s)
- Mariusz Sikora
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82a, 02-008 Warsaw, Poland.
| | - Albert Stec
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82a, 02-008 Warsaw, Poland
| | - Magdalena Chrabaszcz
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82a, 02-008 Warsaw, Poland
| | - Anna Waskiel-Burnat
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82a, 02-008 Warsaw, Poland
| | - Michal Zaremba
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82a, 02-008 Warsaw, Poland
| | - Malgorzata Olszewska
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82a, 02-008 Warsaw, Poland
| | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, Koszykowa 82a, 02-008 Warsaw, Poland
| |
Collapse
|