101
|
Rademacher F, Bartels J, Gläser R, Rodewald M, Schubert S, Drücke D, Rohde H, Harder J. Staphylococcus epidermidis-derived protease Esp mediates proteolytic activation of pro-IL-1beta in human keratinocytes. J Invest Dermatol 2022; 142:2756-2765.e8. [PMID: 35490742 DOI: 10.1016/j.jid.2022.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/03/2022] [Accepted: 04/07/2022] [Indexed: 11/16/2022]
Abstract
The gram-positive bacterium Staphylococcus epidermidis (SE) is an abundant skin commensal. It plays an important role in cutaneous defense by activation of IL-1 signaling. In keratinocytes, SE induces the release of mature IL-1beta. IL-1beta serves as an important cytokine of host defense. It contains an N-terminal prodomain that has to be cleaved off to generate active mature IL-1beta. Typically, processing and release of IL-1beta are associated with inflammasome assembly and activation of the protease caspase-1. Here we report that bacterial challenge of keratinocytes with SE induced the release of mature IL-1beta in a caspase-1-independent manner. Instead, the SE -derived serine protease Esp was identified as a pro-IL-1beta processing factor leading to a proteolytic maturation of active IL-1beta. Esp production and secretion by various SE strains correlated with their capacity to induce release of mature IL-1beta in human primary keratinocytes. Reconstitution of Esp-lacking SE strains with Esp enhanced their capacity to induce IL-1beta release in keratinocytes and skin. Intracellular abundance of pro-IL-1beta and cytotoxic effects of SE suggest release of pro-IL-1beta during injury followed by extracellular Esp-mediated processing to mature IL-1beta. These findings provide further insights into how a skin commensal interacts with keratinocytes to activate cutaneous host innate defense.
Collapse
Affiliation(s)
| | | | - Regine Gläser
- Department of Dermatology, Kiel University, Kiel, Germany
| | - Meno Rodewald
- Department of Dermatology, Kiel University, Kiel, Germany
| | - Sabine Schubert
- Institute of Infection Medicine, Kiel University, Kiel, Germany
| | - Daniel Drücke
- Department of Reconstructive Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Holger Rohde
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jürgen Harder
- Department of Dermatology, Kiel University, Kiel, Germany.
| |
Collapse
|
102
|
Infection prevention requirements for the medical care of immunosuppressed patients: recommendations of the Commission for Hospital Hygiene and Infection Prevention (KRINKO) at the Robert Koch Institute. GMS HYGIENE AND INFECTION CONTROL 2022; 17:Doc07. [PMID: 35707229 PMCID: PMC9174886 DOI: 10.3205/dgkh000410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In Germany, guidelines for hygiene in hospitals are given in form of recommendations by the Commission for Hospital Hygiene and Infection Prevention (Kommission für Krankenhaushygiene und Infektionsprävention, "KRINKO"). The KRINKO and its voluntary work are legitimized by the mandate according to § 23 of the Infection Protection Act (Infektionsschutzgesetz, "IfSG"). The original German version of this document was published in February 2021 and has now been made available to the international professional public in English. The guideline provides recommendations on infection prevention and control for immunocompromised individuals in health care facilities. This recommendation addresses not only measures related to direct medical care of immunocompromised patients, but also management aspects such as surveillance, screening, antibiotic stewardship, and technical/structural aspects such as patient rooms, air quality, and special measures during renovations.
Collapse
|
103
|
Fölster-Holst R. The role of the skin microbiome in atopic dermatitis - correlations and consequences. J Dtsch Dermatol Ges 2022; 20:571-577. [PMID: 35384293 DOI: 10.1111/ddg.14709] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 11/29/2021] [Indexed: 12/26/2022]
Abstract
The physical barrier function of the skin is significantly supported by the (epi-)dermal immune system and the skin's own microbiome. Atopic dermatitis is characterized by an imbalance of all these three factors. The skin microbiome establishes itself immediately after birth and plays an important role in the development and maintenance of immune homeostasis. The clinical picture of atopic dermatitis shows, among other things, changes in the skin microbiome. Particularly during an acute phase, a strongly reduced bacterial diversity as well as the dominance of a single pathogen, Staphylococcus aureus, is observed. Staphylococcus aureus exacerbates the inflammatory process; furthermore, the bacteria produce proteases and toxins that further weaken the already severely compromised barrier function of the skin of patients with atopic dermatitis. However, knowledge of dermal dysbiosis also yields new treatment options for the therapy of the disease. In particular, the application of active bacteria represents a direct influence on the skin microbiome. Results of initial clinical studies on various approaches demonstrate promising results; this article provides an overview of work in this area.
Collapse
Affiliation(s)
- Regina Fölster-Holst
- Department of Dermatology, Venereology and Allergology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
104
|
Castillo‐González R, Fernández‐Delgado I, Comberiati P. Bacteriotherapy with human skin commensals in atopic dermatitis. Allergy 2022; 77:1331-1333. [PMID: 34713451 DOI: 10.1111/all.15162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/18/2021] [Accepted: 10/26/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Raquel Castillo‐González
- Vascular Pathophysiology Department Centro Nacional de Investigaciones Cardiovasculares (CNIC) Madrid Spain
- Department of Immunology Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS‐IP) Madrid Spain
| | - Irene Fernández‐Delgado
- Vascular Pathophysiology Department Centro Nacional de Investigaciones Cardiovasculares (CNIC) Madrid Spain
- Department of Immunology Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS‐IP) Madrid Spain
| | - Pasquale Comberiati
- Department of Clinical and Experimental Medicine, Section of Pediatrics University of Pisa Pisa Italy
- Department of Clinical Immunology and Allergology I.M. Sechenov First Moscow State Medical University Moscow Russia
| |
Collapse
|
105
|
Poh SE, Koh WL, Lim SYD, Wang EC, Yew YW, Common JE, Oon HH, Li H. Expression of Staphylococcus aureus virulence factors in atopic dermatitis. JID INNOVATIONS 2022; 2:100130. [PMID: 35860448 PMCID: PMC9289736 DOI: 10.1016/j.xjidi.2022.100130] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 11/29/2022] Open
Abstract
Atopic dermatitis (AD) is a skin inflammatory disease in which the opportunistic pathogen Staphylococcus aureus is prevalent and abundant. S. aureus harbors several secreted virulence factors that have well-studied functions in infection models, but it is unclear whether these extracellular microbial factors are relevant in the context of AD. To address this question, we designed a culture-independent method to detect and quantify S. aureus virulence factors expressed at the skin sites. We utilized RNase-H‒dependent multiplex PCR for preamplification of reverse-transcribed RNA extracted from tape strips of patients with AD sampled at skin sites with differing severity and assessed the expression of a panel of S. aureus virulence factors using qPCR. We observed an increase in viable S. aureus abundance on sites with increased severity of disease, and many virulence factors were expressed at the AD skin sites. Surprisingly, we did not observe any significant upregulation of the virulence factors at the lesional sites compared with those at the nonlesional control. Overall, we utilized a robust assay to directly detect and quantify viable S. aureus and its associated virulence factors at the site of AD skin lesions. This method can be extended to study the expression of skin microbial genes at the sites of various dermatological conditions.
Collapse
Affiliation(s)
- Si En Poh
- Molecular Engineering Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research, Singapore, Singapore
| | - Winston L.C. Koh
- Molecular Engineering Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research, Singapore, Singapore
| | - Shi Yu Derek Lim
- National Skin Centre, National Healthcare Group, Singapore, Singapore
| | - Etienne C.E. Wang
- National Skin Centre, National Healthcare Group, Singapore, Singapore
- Skin Research Institute of Singapore, Singapore
| | - Yik Weng Yew
- National Skin Centre, National Healthcare Group, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - John E.A. Common
- ASTAR Skin Research Labs (ASRL), Agency for Science, Technology and Research, Singapore, Singapore
| | - Hazel H. Oon
- National Skin Centre, National Healthcare Group, Singapore, Singapore
| | - Hao Li
- Molecular Engineering Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research, Singapore, Singapore
- Department of Chemistry, Faculty of Science, National University of Singapore, Singapore, Singapore
- Correspondence: Hao Li, Department of Chemistry, Faculty of Science, National University of Singapore, 4 Science Drive 2, S9 Level 12, Singapore 117544, Singapore.
| |
Collapse
|
106
|
Zheng Y, Hunt RL, Villaruz AE, Fisher EL, Liu R, Liu Q, Cheung GYC, Li M, Otto M. Commensal Staphylococcus epidermidis contributes to skin barrier homeostasis by generating protective ceramides. Cell Host Microbe 2022; 30:301-313.e9. [PMID: 35123653 PMCID: PMC8917079 DOI: 10.1016/j.chom.2022.01.004] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/07/2021] [Accepted: 01/11/2022] [Indexed: 12/19/2022]
Abstract
Previously either regarded as insignificant or feared as potential sources of infection, the bacteria living on our skin are increasingly recognized for their role in benefitting human health. Skin commensals modulate mucosal immune defenses and directly interfere with pathogens; however, their contribution to the skin's physical integrity is less understood. Here, we show that the abundant skin commensal Staphylococcus epidermidis contributes to skin barrier integrity. S. epidermidis secretes a sphingomyelinase that acquires essential nutrients for the bacteria and assists the host in producing ceramides, the main constituent of the epithelial barrier that averts skin dehydration and aging. In mouse models, S. epidermidis significantly increases skin ceramide levels and prevents water loss of damaged skin in a fashion entirely dependent on its sphingomyelinase. Our findings reveal a symbiotic mechanism that demonstrates an important role of the skin microbiota in the maintenance of the skin's protective barrier.
Collapse
Affiliation(s)
- Yue Zheng
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD 20814, USA
| | - Rachelle L Hunt
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD 20814, USA
| | - Amer E Villaruz
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD 20814, USA
| | - Emilie L Fisher
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD 20814, USA
| | - Ryan Liu
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD 20814, USA
| | - Qian Liu
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gordon Y C Cheung
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD 20814, USA
| | - Min Li
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD 20814, USA.
| |
Collapse
|
107
|
Chia M, Naim AN, Tay AS, Lim K, Lee CK, Yow SJ, Chen J, Common JE, Nagarajan N, Tham EH. Shared signatures and divergence in skin microbiomes of children with atopic dermatitis and their caregivers. J Allergy Clin Immunol 2022; 150:894-908. [DOI: 10.1016/j.jaci.2022.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 01/17/2022] [Accepted: 01/25/2022] [Indexed: 10/18/2022]
|
108
|
Miyano T, Irvine AD, Tanaka RJ. Model-based meta-analysis to optimise S. aureus-targeted therapies for atopic dermatitis. JID INNOVATIONS 2022; 2:100110. [PMID: 35757782 PMCID: PMC9214323 DOI: 10.1016/j.xjidi.2022.100110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 11/29/2022] Open
Abstract
Several clinical trials of Staphylococcus aureus (S. aureus)‒targeted therapies for atopic dermatitis (AD) have shown conflicting results about whether they improve AD severity scores. This study performs a model-based meta-analysis to investigate the possible causes of these conflicting results and suggests how to improve the efficacies of S. aureus‒targeted therapies. We developed a mathematical model that describes systems-level AD pathogenesis involving dynamic interactions between S. aureus and coagulase-negative Staphylococcus (CoNS). Our model simulation reproduced the clinically observed detrimental effects of the application of S. hominis A9 and flucloxacillin on AD severity and showed that these effects disappeared if the bactericidal activity against CoNS was removed. A hypothetical (modeled) eradication of S. aureus by 3.0 log10 colony-forming unit per cm2 without killing CoNS achieved Eczema Area and Severity Index 75 comparable with that of dupilumab. This efficacy was potentiated if dupilumab was administered in conjunction with S. aureus eradication (Eczema Area and Severity Index 75 at week 16) (S. aureus eradication: 66.7%, dupilumab 61.6% and combination 87.8%). The improved efficacy was also seen for virtual dupilumab poor responders. Our model simulation suggests that killing CoNS worsens AD severity and that S. aureus‒specific eradication without killing CoNS could be effective for patients with AD, including dupilumab poor responders. This study will contribute to designing promising S. aureus‒targeted therapy.
Collapse
Affiliation(s)
- Takuya Miyano
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Alan D. Irvine
- Pediatric Dermatology, Children’s Health Ireland at Crumlin, Dublin, Ireland
- Clinical Medicine, College of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Reiko J. Tanaka
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Correspondence: Reiko J. Tanaka, Department of Bioengineering, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom.
| |
Collapse
|
109
|
Licht P, Mailänder V. Transcriptional Heterogeneity and the Microbiome of Cutaneous T-Cell Lymphoma. Cells 2022; 11:cells11030328. [PMID: 35159138 PMCID: PMC8834405 DOI: 10.3390/cells11030328] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/31/2021] [Accepted: 01/13/2022] [Indexed: 11/16/2022] Open
Abstract
Cutaneous T-Cell Lymphomas (CTCL) presents with substantial clinical variability and transcriptional heterogeneity. In the recent years, several studies paved the way to elucidate aetiology and pathogenesis of CTCL using sequencing methods. Several T-cell subtypes were suggested as the source of disease thereby explaining clinical and transcriptional heterogeneity of CTCL entities. Several differentially expressed pathways could explain disease progression. However, exogenous triggers in the skin microenvironment also seem to affect CTCL status. Especially Staphylococcus aureus was shown to contribute to disease progression. Only little is known about the complex microbiome patterns involved in CTCL and how microbial shifts might impact this malignancy. Nevertheless, first hints indicate that the microbiome might at least in part explain transcriptional heterogeneity and that microbial approaches could serve in diagnosis and prognosis. Shaping the microbiome could be a treatment option to maintain stable disease. Here, we review current knowledge of transcriptional heterogeneity of and microbial influences on CTCL. We discuss potential benefits of microbial applications and microbial directed therapies to aid patients with CTCL burden.
Collapse
Affiliation(s)
- Philipp Licht
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
| | - Volker Mailänder
- Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
- Correspondence:
| |
Collapse
|
110
|
Zhang Y, Zhou L, Xia J, Dong C, Luo X. Human Microbiome and Its Medical Applications. Front Mol Biosci 2022; 8:703585. [PMID: 35096962 PMCID: PMC8793671 DOI: 10.3389/fmolb.2021.703585] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
The commensal microbiome is essential for human health and is involved in many processes in the human body, such as the metabolism process and immune system activation. Emerging evidence implies that specific changes in the microbiome participate in the development of various diseases, including diabetes, liver diseases, tumors, and pathogen infections. Thus, intervention on the microbiome is becoming a novel and effective method to treat such diseases. Synthetic biology empowers researchers to create strains with unique and complex functions, making the use of engineered microbes for clinical applications attainable. The aim of this review is to summarize recent advances about the roles of the microbiome in certain diseases and the underlying mechanisms, as well as the use of engineered microbes in the prevention, detection, and treatment of various diseases.
Collapse
Affiliation(s)
- Yangming Zhang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Linguang Zhou
- Department of Pharmacy, Peking University International Hospital, Beijing, China
| | - Jialin Xia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Ce Dong
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaozhou Luo
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- *Correspondence: Xiaozhou Luo,
| |
Collapse
|
111
|
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
|
112
|
Ohkubo T, Matsumoto Y, Ogasawara Y, Sugita T. Alkaline stress inhibits the growth of Staphylococcus epidermidis by inducing TCA cycle-triggered ROS production. Biochem Biophys Res Commun 2021; 588:104-110. [PMID: 34953206 DOI: 10.1016/j.bbrc.2021.12.053] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/15/2021] [Indexed: 12/19/2022]
Abstract
Many species of bacteria interact on the human skin to form a certain microbiome. Delftia acidovorans, a bacterium detected from human skin, inhibits the growth of S. epidermidis, a dominant bacterium of the human skin microbiota. Here, we show that ammonia secreted by D. acidovorans inhibits the growth of S. epidermidis by increasing the pH value of the medium. The pH value of D. acidovorans culture supernatant (CS) was higher than that of the medium without culture. The inhibitory activity of the D. acidovorans CS against the growth of S. epidermidis was decreased by neutralization with hydrochloric acid. Genes encoding enzymes related to ammonia production were found in the D. acidovorans genome. Moreover, the D. acidovorans CS contained a high concentration of ammonia. The addition of ammonia to S. epidermidis culture led to an increase in the reactive oxygen species (ROS) production and inhibited S. epidermidis growth. The addition of sodium hydroxide also led to an increase in the ROS production and inhibited S. epidermidis growth. The inhibitory activity of ammonia and sodium hydroxide against S. epidermidis growth was suppressed by malonic acid, an inhibitor of succinate dehydrogenase in the tricarboxylic acid (TCA) cycle, and N-acetyl-l-cysteine, a free radical scavenger. These findings suggest that D. acidovorans secretes ammonia and alkaline stress inhibits the growth of S. epidermidis by inducing TCA cycle-triggered ROS production.
Collapse
Affiliation(s)
- Tomotaka Ohkubo
- Department of Microbiology, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan; Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Yasuhiko Matsumoto
- Department of Microbiology, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan.
| | - Yuki Ogasawara
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Takashi Sugita
- Department of Microbiology, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| |
Collapse
|
113
|
Skin Microbiota and Clinical Associations in Netherton Syndrome. JID INNOVATIONS 2021; 1:100008. [PMID: 34909712 PMCID: PMC8659401 DOI: 10.1016/j.xjidi.2021.100008] [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: 11/11/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 01/06/2023] Open
Abstract
Netherton syndrome (NS) is a rare, life-threatening syndrome caused by serine protease inhibitor Kazal-type 5 gene (SPINK5) mutations, resulting in skin barrier defect, bacterial skin infections, and allergic sensitization in early childhood. Recent data on adult patients with NS suggest that the presence of Staphylococcus aureus further promotes barrier disruption and skin inflammation. We analyzed the skin microbiota by shotgun sequencing in 12 patients with NS from eight Finnish families with healthy family controls as the reference and correlated the findings with allergen-specific IgE prevalence, immune cell phenotype, and infection history of the patients. Compared with healthy family controls, skin microbiome diversity and normal skin site variability were measurably decreased in patients with NS. No correlation was found between allergic sensitization and skin microbiota as such, but low circulating CD57+ and/or CD8+ T cells significantly correlated with lower microbial diversity and less abundance of S. aureus (P < 0.05). S. aureus was the most prevalent species in patients with NS but also Streptococcus agalactiae was abundant in four patients. The genomic DNA relative abundance of S. aureus secreted virulence peptides and proteases PSMα, staphopain A, and staphopain B were increased in most of the samples of patients with NS, and their abundance was significantly (P < 0.05) associated with recurrent childhood skin infections, confirming the clinical relevance of S. aureus dominance in the NS skin microbiome.
Collapse
|
114
|
Cheung GYC, Bae JS, Otto M. Pathogenicity and virulence of Staphylococcus aureus. Virulence 2021; 12:547-569. [PMID: 33522395 PMCID: PMC7872022 DOI: 10.1080/21505594.2021.1878688] [Citation(s) in RCA: 618] [Impact Index Per Article: 154.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Staphylococcus aureus is one of the most frequent worldwide causes of morbidity and mortality due to an infectious agent. This pathogen can cause a wide variety of diseases, ranging from moderately severe skin infections to fatal pneumonia and sepsis. Treatment of S. aureus infections is complicated by antibiotic resistance and a working vaccine is not available. There has been ongoing and increasing interest in the extraordinarily high number of toxins and other virulence determinants that S. aureus produces and how they impact disease. In this review, we will give an overview of how S. aureus initiates and maintains infection and discuss the main determinants involved. A more in-depth understanding of the function and contribution of S. aureus virulence determinants to S. aureus infection will enable us to develop anti-virulence strategies to counteract the lack of an anti-S. aureus vaccine and the ever-increasing shortage of working antibiotics against this important pathogen.
Collapse
Affiliation(s)
- Gordon Y. C. Cheung
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, Maryland, USA
| | - Justin S. Bae
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, Maryland, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
115
|
Dhariwala MO, Scharschmidt TC. Baby's skin bacteria: first impressions are long-lasting. Trends Immunol 2021; 42:1088-1099. [PMID: 34743922 PMCID: PMC9206859 DOI: 10.1016/j.it.2021.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022]
Abstract
Early life is a dynamic period for skin microbial colonization and immune development. We postulate that microbial exposures in this period durably alter the skin immune trajectory and later disease susceptibility. Bacteria contribute to infant skin immune imprinting via interactions with microbes as well as with cutaneous epithelial and immune cells. Excellent research is underway at the skin microbiome-immune interface, both in deciphering basic mechanisms and implementing their therapeutic applications. As emphasized herein, focusing on the unique opportunities and challenges presented by microbial immune modulation in early life will be important. In our view, only through dedicated study of skin-microbe crosstalk in this developmental window can we elucidate the molecular underpinnings of pivotal events that contribute to sustained host-microbe symbiosis.
Collapse
Affiliation(s)
- Miqdad O Dhariwala
- Department of Dermatology, University of California San Francisco (UCSF), San Francisco, CA 94143, USA
| | - Tiffany C Scharschmidt
- Department of Dermatology, University of California San Francisco (UCSF), San Francisco, CA 94143, USA.
| |
Collapse
|
116
|
Damour A, Robin B, Deroche L, Broutin L, Bellin N, Verdon J, Lina G, Leclère FM, Garcia M, Cremniter J, Lévêque N, Bodet C. Phenol-soluble modulins α are major virulence factors of Staphylococcus aureus secretome promoting inflammatory response in human epidermis. Virulence 2021; 12:2474-2492. [PMID: 34516337 PMCID: PMC8451463 DOI: 10.1080/21505594.2021.1975909] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 08/03/2021] [Accepted: 08/25/2021] [Indexed: 11/10/2022] Open
Abstract
Staphylococcus aureus is a skin commensal microorganism commonly colonizing healthy humans. Nevertheless, S. aureus can also be responsible for cutaneous infections and contribute to flare-up of inflammatory skin diseases such as atopic dermatitis (AD), which is characterized by dysbiosis of the skin microbiota with S. aureus as the predominant species. However, the role of major virulence factors of this pathogen such as phenol-soluble modulin (PSM) toxins in epidermal inflammation remains poorly understood. Stimulation of primary human keratinocytes with sublytic concentrations of synthetic and purified PSM α3 resulted in upregulation of a large panel of pro-inflammatory chemokine and cytokine gene expression, including CXCL1, CXCL2, CXCL3, CXCL5, CXCL8, CCL20, IL-1α, IL-1β, IL-6, IL-36γ and TNF-α, while inducing the release of CXCL8, CCL20, TNF-α and IL-6. In addition, using S. aureus culture supernatant from mutants deleted from genes encoding either α-type PSMs or all PSM production, PSMs were shown to be the main factors of S. aureus secretome responsible for pro-inflammatory mediator induction in human keratinocytes. On the other hand, α-type PSM-containing supernatant triggered an intense induction of pro-inflammatory mediator expression and secretion during both topical and basal layer stimulation of an ex vivo model of human skin explants, a physiologically relevant model of pluristratified epidermis. Taken together, the results of this study show that PSMs and more specifically α-type PSMs are major virulence factors of S. aureus inducing a potent inflammatory response during infection of the human epidermis and could thereby contribute to AD flare-up through exacerbation of skin inflammation.
Collapse
Affiliation(s)
- Alexia Damour
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université De Poitiers, Poitiers, France
| | - Brandon Robin
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université De Poitiers, Poitiers, France
| | - Luc Deroche
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université De Poitiers, Poitiers, France
| | - Lauranne Broutin
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université De Poitiers, Poitiers, France
- Laboratoire De Bactériologie, CHU de Poitiers, Poitiers, France
| | - Nicolas Bellin
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université De Poitiers, Poitiers, France
| | - Julien Verdon
- Laboratoire Ecologie et Biologie des Interactions, UMR CNRS 7267, Université De Poitiers, Poitiers, France
| | - Gérard Lina
- CIRI Centre International de Recherche en Infectiologie, Inserm U1111, Université Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des Agent Infectieux, Hôpital de La Croix Rousse, Hospices Civils de Lyon, Lyon, France
| | - Franck Marie Leclère
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université De Poitiers, Poitiers, France
- Département de Chirurgie Plastique, Reconstructive et Esthétique, CHU de Poitiers, Poitiers, France
| | - Magali Garcia
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université De Poitiers, Poitiers, France
- Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, Poitiers, France
| | - Julie Cremniter
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université De Poitiers, Poitiers, France
- Laboratoire De Bactériologie, CHU de Poitiers, Poitiers, France
| | - Nicolas Lévêque
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université De Poitiers, Poitiers, France
- Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, Poitiers, France
| | - Charles Bodet
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université De Poitiers, Poitiers, France
| |
Collapse
|
117
|
Rebuffat S. Ribosomally synthesized peptides, foreground players in microbial interactions: recent developments and unanswered questions. Nat Prod Rep 2021; 39:273-310. [PMID: 34755755 DOI: 10.1039/d1np00052g] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
It is currently well established that multicellular organisms live in tight association with complex communities of microorganisms including a large number of bacteria. These are immersed in complex interaction networks reflecting the relationships established between them and with host organisms; yet, little is known about the molecules and mechanisms involved in these mutual interactions. Ribosomally synthesized peptides, among which bacterial antimicrobial peptides called bacteriocins and microcins have been identified as contributing to host-microbe interplays, are either unmodified or post-translationally modified peptides. This review will unveil current knowledge on these ribosomal peptide-based natural products, their interplay with the host immune system, and their roles in microbial interactions and symbioses. It will include their major structural characteristics and post-translational modifications, the main rules of their maturation pathways, and the principal ecological functions they ensure (communication, signalization, competition), especially in symbiosis, taking select examples in various organisms. Finally, we address unanswered questions and provide a framework for deciphering big issues inspiring future directions in the field.
Collapse
Affiliation(s)
- Sylvie Rebuffat
- Laboratory Molecules of Communication and Adaptation of Microorganisms (MCAM, UMR 7245 CNRS-MNHN), National Museum of Natural History (MNHN), National Centre of Scientific Research (CNRS), CP 54, 57 rue Cuvier 75005, Paris, France.
| |
Collapse
|
118
|
Hülpüsch C, Weins AB, Traidl‐Hoffmann C, Reiger M. A new era of atopic eczema research: Advances and highlights. Allergy 2021; 76:3408-3421. [PMID: 34407212 DOI: 10.1111/all.15058] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 05/03/2021] [Accepted: 08/12/2021] [Indexed: 01/09/2023]
Abstract
Atopic eczema (AE) is an inflammatory skin disease with involvement of genetic, immunological and environmental factors. One hallmark of AE is a skin barrier disruption on multiple, highly interconnected levels: filaggrin mutations, increased skin pH and a microbiome dysbiosis towards Staphylococcus aureus overgrowth are observed in addition to an abnormal type 2 immune response. Extrinsic factors seem to play a major role in the development of AE. As AE is a first step in the atopic march, its prevention and appropriate treatment are essential. Although standard therapy remains topical treatment, powerful systemic treatment options emerged in the last years. However, thorough endotyping of the individual patients is still required for ideal precision medicine approaches in future. Therefore, novel microbial and immunological biomarkers were described recently for the prediction of disease development and treatment response. This review summarizes the current state of the art in AE research.
Collapse
Affiliation(s)
- Claudia Hülpüsch
- Department of Environmental Medicine Faculty of Medicine University of Augsburg Augsburg Germany
- Institute of Environmental Medicine Helmholtz Zentrum München Augsburg Germany
- CK CARE – Christine Kühne Center for Allergy research and Education Davos Switzerland
| | - Andreas B. Weins
- Department of Dermatology Faculty of Medicine University of Augsburg 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
- CK CARE – Christine Kühne Center for Allergy research and Education Davos Switzerland
- ZIEL Technical University of Munich Freising Germany
| | - Matthias Reiger
- Department of Environmental Medicine Faculty of Medicine University of Augsburg Augsburg Germany
- Institute of Environmental Medicine Helmholtz Zentrum München Augsburg Germany
- CK CARE – Christine Kühne Center for Allergy research and Education Davos Switzerland
| |
Collapse
|
119
|
di Lorenzo R, Bernardi A, Grumetto L, Sacchi A, Avagliano C, Coppola S, de Giovanni di Santa Severina AF, Bruno C, Paparo L, Laneri S, Dini I. Phenylalanine Butyramide Is a New Cosmetic Ingredient with Soothing and Anti-Reddening Potential. Molecules 2021; 26:6611. [PMID: 34771020 PMCID: PMC8586959 DOI: 10.3390/molecules26216611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 01/11/2023] Open
Abstract
Human skin is colonized by diverse commensal microbes, making up the skin microbiota (SM), contributing to skin integrity and homeostasis. Many of the beneficial effects aroused by the SM are exerted by microbial metabolites such as short-chain fatty acids (SCFAs), including butyric acid. The SCFAs can be used in cosmetic formulations against skin diseases to protect SM by preserving and/or restoring their natural balance. Unpleasant sensorial properties and unfavorable physico-chemical properties of butyrate strongly limit its cosmetic use. In contrast, some butyrate derivatives, including phenylalanine butyramide (C13H18N2O2, FBA), a solid form of butyric acid, are odorless while retaining the pharmacokinetic properties and safety profile of butyric acid. This study assessed the FBA's permeation across the skin and its soothing and anti-reddening potential to estimate its cosmetic application. The dosage method used to estimate FBA's levels was validated to be sure of analytical results. The FBA diffusion tests were estimated in vitro using a Franz-type vertical diffusion cell. The soothing action was evaluated in vivo by Colorimeter CL400, measuring the erythema index. The results suggest that the FBA represents an innovative way to exploit the benefits of butyric acid in the cosmetic fields since it cannot reach the bloodstream, is odorless, and has a significative soothing action (decrease the erythema index -15.7% after 30', and -17.8% after 60').
Collapse
Affiliation(s)
- Ritamaria di Lorenzo
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy; (R.d.L.); (A.B.); (L.G.); (A.S.); (C.A.)
| | - Antonietta Bernardi
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy; (R.d.L.); (A.B.); (L.G.); (A.S.); (C.A.)
| | - Lucia Grumetto
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy; (R.d.L.); (A.B.); (L.G.); (A.S.); (C.A.)
| | - Antonia Sacchi
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy; (R.d.L.); (A.B.); (L.G.); (A.S.); (C.A.)
| | - Carmen Avagliano
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy; (R.d.L.); (A.B.); (L.G.); (A.S.); (C.A.)
| | - Serena Coppola
- Department of Translational Medical Science, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy; (S.C.); (A.F.d.G.d.S.S.); (C.B.); (L.P.)
- ImmunoNutritionLab at the CEINGE-Biotecnologie Avanzate s.c.ar.l Research Center, University of Naples Federico II, 80131 Naples, Italy
| | - Anna Fiorenza de Giovanni di Santa Severina
- Department of Translational Medical Science, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy; (S.C.); (A.F.d.G.d.S.S.); (C.B.); (L.P.)
- ImmunoNutritionLab at the CEINGE-Biotecnologie Avanzate s.c.ar.l Research Center, University of Naples Federico II, 80131 Naples, Italy
| | - Cristina Bruno
- Department of Translational Medical Science, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy; (S.C.); (A.F.d.G.d.S.S.); (C.B.); (L.P.)
- ImmunoNutritionLab at the CEINGE-Biotecnologie Avanzate s.c.ar.l Research Center, University of Naples Federico II, 80131 Naples, Italy
| | - Lorella Paparo
- Department of Translational Medical Science, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy; (S.C.); (A.F.d.G.d.S.S.); (C.B.); (L.P.)
- ImmunoNutritionLab at the CEINGE-Biotecnologie Avanzate s.c.ar.l Research Center, University of Naples Federico II, 80131 Naples, Italy
| | - Sonia Laneri
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy; (R.d.L.); (A.B.); (L.G.); (A.S.); (C.A.)
| | - Irene Dini
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy; (R.d.L.); (A.B.); (L.G.); (A.S.); (C.A.)
| |
Collapse
|
120
|
O'Neill AM, Worthing KA, Kulkarni N, Li F, Nakatsuji T, McGrosso D, Mills RH, Kalla G, Cheng JY, Norris JM, Pogliano K, Pogliano J, Gonzalez DJ, Gallo RL. Antimicrobials from a feline commensal bacterium inhibit skin infection by drug-resistant S. pseudintermedius. eLife 2021; 10:66793. [PMID: 34664551 PMCID: PMC8592530 DOI: 10.7554/elife.66793] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 10/02/2021] [Indexed: 12/13/2022] Open
Abstract
Methicillin-resistant Staphylococcus pseudintermedius (MRSP) is an important emerging zoonotic pathogen that causes severe skin infections. To combat infections from drug-resistant bacteria, the transplantation of commensal antimicrobial bacteria as a therapeutic has shown clinical promise. We screened a collection of diverse staphylococcus species from domestic dogs and cats for antimicrobial activity against MRSP. A unique strain (S. felis C4) was isolated from feline skin that inhibited MRSP and multiple gram-positive pathogens. Whole genome sequencing and mass spectrometry revealed several secreted antimicrobials including a thiopeptide bacteriocin micrococcin P1 and phenol-soluble modulin beta (PSMβ) peptides that exhibited antimicrobial and anti-inflammatory activity. Fluorescence and electron microscopy revealed that S. felis antimicrobials inhibited translation and disrupted bacterial but not eukaryotic cell membranes. Competition experiments in mice showed that S. felis significantly reduced MRSP skin colonization and an antimicrobial extract from S. felis significantly reduced necrotic skin injury from MRSP infection. These findings indicate a feline commensal bacterium that could be utilized in bacteriotherapy against difficult-to-treat animal and human skin infections.
Collapse
Affiliation(s)
- Alan M O'Neill
- Department of Dermatology, University of California, San Diego, San Diego, United States
| | - Kate A Worthing
- College of Veterinary Medicine, University of Arizona, Oro Valley, United States
| | - Nikhil Kulkarni
- Department of Dermatology, University of California, San Diego, San Diego, United States
| | - Fengwu Li
- Department of Dermatology, University of California, San Diego, San Diego, United States
| | - Teruaki Nakatsuji
- Department of Dermatology, University of California, San Diego, San Diego, United States
| | - Dominic McGrosso
- Department of Pharmacology, University of California, San Diego, San Diego, United States.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, San Diego, United States
| | - Robert H Mills
- Department of Pharmacology, University of California, San Diego, San Diego, United States.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, San Diego, United States
| | - Gayathri Kalla
- Division of Biological Sciences, University of California, San Diego, San Diego, United States
| | - Joyce Y Cheng
- Department of Dermatology, University of California, San Diego, San Diego, United States
| | - Jacqueline M Norris
- Sydney School of Veterinary Science, University of Sydney, Sydney, Australia
| | - Kit Pogliano
- Division of Biological Sciences, University of California, San Diego, San Diego, United States
| | - Joe Pogliano
- Division of Biological Sciences, University of California, San Diego, San Diego, United States
| | - David J Gonzalez
- Department of Pharmacology, University of California, San Diego, San Diego, United States.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, San Diego, United States
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, San Diego, United States
| |
Collapse
|
121
|
Rapid methicillin resistance diversification in Staphylococcus epidermidis colonizing human neonates. Nat Commun 2021; 12:6062. [PMID: 34663826 PMCID: PMC8523572 DOI: 10.1038/s41467-021-26392-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 10/04/2021] [Indexed: 11/08/2022] Open
Abstract
Early in life, infants are colonized with multiple bacterial strains whose differences in gene content can have important health consequences. Metagenomics-based approaches have revealed gene content differences between different strains co-colonizing newborns, but less is known about the rate, mechanism, and phenotypic consequences of gene content diversification within strains. Here, focusing on Staphylococcus epidermidis, we whole-genome sequence and phenotype more than 600 isolates from newborns. Within days of birth, infants are co-colonized with a highly personalized repertoire of S. epidermidis strains, which are spread across the newborn body. Comparing the genomes of multiple isolates of each strain, we find very little evidence of adaptive evolution via single-nucleotide polymorphisms. By contrast, we observe gene content differences even between otherwise genetically identical cells, including variation of the clinically important methicillin resistance gene, mecA, suggesting rapid gene gain and loss events at rates higher than point mutations. Mapping the genomic architecture of structural variants by long-read Nanopore sequencing, we find that deleted regions were always flanked by direct repeats, consistent with site-specific recombination. However, we find that even within a single genetic background, recombination occurs at multiple, often non-canonical repeats, leading to the rapid evolution of patient-specific diverse structural variants in the SCCmec island and to differences in antibiotic resistance.
Collapse
|
122
|
Burian M, Plange J, Schmitt L, Kaschke A, Marquardt Y, Huth L, Baron JM, Hornef MW, Wolz C, Yazdi AS. Adaptation of Staphylococcus aureus to the Human Skin Environment Identified Using an ex vivo Tissue Model. Front Microbiol 2021; 12:728989. [PMID: 34621255 PMCID: PMC8490888 DOI: 10.3389/fmicb.2021.728989] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/01/2021] [Indexed: 11/13/2022] Open
Abstract
The healthy human epidermis provides physical protection and is impenetrable for pathogenic microbes. Nevertheless, commensal and pathogen bacteria such as Staphylococcus aureus are able to colonize the skin surface, which may subsequently lead to infection. To identify and characterize regulatory elements facilitating adaptation of S. aureus to the human skin environment we used ex vivo tissue explants and quantified S. aureus gene transcription during co-culture. This analysis provided evidence for a significant downregulation of the global virulence regulator agr upon initial contact with skin, regardless of the growth phase of S. aureus prior to co-culture. In contrast, the alternative sigma factor B (sigB) and the antimicrobial peptide-sensing system (graRS) were expressed during early colonization. Consistently, sigB target genes such as the clumping factor A (clfA) and fibrinogen and fibronectin binding protein A (fnbA) were strongly upregulated upon skin contact. At later timepoints of the adhesion process, wall teichoic acid (WTA) synthesis was induced. Besides the expression of adhesive molecules, transcription of molecules involved in immune evasion were increased during late colonization (staphylococcal complement inhibitor and staphylokinase). Similar to nasal colonization, enzymes involved in cell wall metabolism (sceD and atlA) were highly transcribed. Finally, we detected a strong expression of proteases from all three catalytic classes during the entire colonization process. Taken together, we here present an ex vivo skin colonization model that allows the detailed characterization of the bacterial adaptation to the skin environment.
Collapse
Affiliation(s)
- Marc Burian
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, Aachen, Germany
| | - Johanna Plange
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, Aachen, Germany
| | - Laurenz Schmitt
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, Aachen, Germany
| | - Anke Kaschke
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, Aachen, Germany
| | - Yvonne Marquardt
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, Aachen, Germany
| | - Laura Huth
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, Aachen, Germany
| | - Jens M Baron
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, Aachen, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital Aachen, Aachen, Germany
| | - Christiane Wolz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, Tuebingen, Germany
| | - Amir S Yazdi
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
123
|
Kohda K, Li X, Soga N, Nagura R, Duerna T, Nakajima S, Nakagawa I, Ito M, Ikeuchi A. An In Vitro Mixed Infection Model With Commensal and Pathogenic Staphylococci for the Exploration of Interspecific Interactions and Their Impacts on Skin Physiology. Front Cell Infect Microbiol 2021; 11:712360. [PMID: 34604106 PMCID: PMC8481888 DOI: 10.3389/fcimb.2021.712360] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022] Open
Abstract
The skin microbiota has been recognized to play an integral role in the physiology and pathology of the skin. The crosstalk between skin and the resident microbes has been extensively investigated using two-dimensional (2D) and three-dimensional (3D) cell cultures in vitro; however, skin colonization by multiple species and the effects of interspecific interactions on the structure and function of skin remains to be elucidated. This study reports the establishment of a mixed infection model, incorporating both commensal (Staphylococcus epidermidis) and pathogenic (Staphylococcus aureus) bacteria, based on a 3D human epidermal model. We observed that co-infecting the 3D epidermal model with S. aureus and S. epidermidis restricted the growth of S. aureus. In addition, S. aureus induced epidermal cytotoxicity, and the release of proinflammatory cytokines was attenuated by the S. aureus-S. epidermidis mixed infection model. S. epidermidis also inhibited the invasion of the deeper epidermis by S. aureus, eliciting protective effects on the integrity of the epidermal barrier. This 3D culture-based mixed infection model would be an effective replacement for existing animal models and 2D cell culture approaches for the evaluation of diverse biotic and abiotic factors involved in maintaining skin health.
Collapse
Affiliation(s)
- Katsunori Kohda
- Frontier Research Center, Toyota Motor Corporation, Toyota, Japan
| | - Xuan Li
- Frontier Research Center, Toyota Motor Corporation, Toyota, Japan
| | - Naoki Soga
- Frontier Research Center, Toyota Motor Corporation, Toyota, Japan
| | - Risa Nagura
- Frontier Research Center, Toyota Motor Corporation, Toyota, Japan
| | - Tie Duerna
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Saeko Nakajima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ichiro Nakagawa
- Department of Microbiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masakazu Ito
- Frontier Research Center, Toyota Motor Corporation, Toyota, Japan
| | - Akinori Ikeuchi
- Frontier Research Center, Toyota Motor Corporation, Toyota, Japan
| |
Collapse
|
124
|
Abstract
PURPOSE OF REVIEW Staphylococcus aureus is the most common invasive bacterial pathogen infecting children in the U.S. and many parts of the world. This major human pathogen continues to evolve, and recognition of recent trends in epidemiology, therapeutics and future horizons is of high importance. RECENT FINDINGS Over the past decade, a relative rise of methicillin-susceptible S. aureus (MSSA) has occurred, such that methicillin-resistant S. aureus (MRSA) no longer dominates the landscape of invasive disease. Antimicrobial resistance continues to develop, however, and novel therapeutics or preventive modalities are urgently needed. Unfortunately, several recent vaccine attempts proved unsuccessful in humans. SUMMARY Recent scientific breakthroughs highlight the opportunity for novel interventions against S. aureus by interfering with virulence rather than by traditional antimicrobial mechanisms. A S. aureus vaccine remains elusive; the reasons for this are multifactorial, and lessons learned from prior unsuccessful attempts may create a path toward an effective preventive. Finally, new diagnostic modalities have the potential to greatly enhance clinical care for invasive S. aureus disease in children.
Collapse
Affiliation(s)
- James E. Cassat
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation (VI4), Nashville, Tennessee, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Isaac Thomsen
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation (VI4), Nashville, Tennessee, USA
| |
Collapse
|
125
|
Mechanisms for control of skin immune function by the microbiome. Curr Opin Immunol 2021; 72:324-330. [PMID: 34537476 DOI: 10.1016/j.coi.2021.09.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022]
Abstract
The skin represents the largest area for direct contact between microbes and host immunocytes and is a site for constant communication between the host and this diverse and essential microbial community. Coagulase-negative staphylococci are an abundant bacterial genus on the human skin and are regulated through various mechanisms that include the epidermal barrier environment and innate and adaptive immune systems within the epidermis and dermis. In turn, some species and strains of these bacteria produce beneficial products that augment host immunity by exerting specifically targeted antimicrobial, anti-inflammatory, or anti-neoplastic activity while also promoting broad innate and adaptive immune responses. The use of selected skin commensals as a therapeutic has shown promise in recent human clinical trials. This emerging concept of bacteriotherapy is defining mechanisms of action and validating the dependence on the microbiome for maintenance of immune homeostasis.
Collapse
|
126
|
Bieber T. Atopic dermatitis: an expanding therapeutic pipeline for a complex disease. Nat Rev Drug Discov 2021; 21:21-40. [PMID: 34417579 PMCID: PMC8377708 DOI: 10.1038/s41573-021-00266-6] [Citation(s) in RCA: 333] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2021] [Indexed: 02/07/2023]
Abstract
Atopic dermatitis (AD) is a common chronic inflammatory skin disease with a complex pathophysiology that underlies a wide spectrum of clinical phenotypes. AD remains challenging to treat owing to the limited response to available therapies. However, recent advances in understanding of disease mechanisms have led to the discovery of novel potential therapeutic targets and drug candidates. In addition to regulatory approval for the IL-4Ra inhibitor dupilumab, the anti-IL-13 inhibitor tralokinumab and the JAK1/2 inhibitor baricitinib in Europe, there are now more than 70 new compounds in development. This Review assesses the various strategies and novel agents currently being investigated for AD and highlights the potential for a precision medicine approach to enable prevention and more effective long-term control of this complex disease. Recent advances in understanding of the complex phenotype and mechanisms underlying atopic dermatitis (AD) have revealed multiple new potential targets for pharmacological intervention. Here, Bieber reviews therapeutic strategies and assesses the expanding pipeline for the therapy of AD, highlighting the potential for a precision medicine approach to the management of this complex disorder.
Collapse
Affiliation(s)
- Thomas Bieber
- Department of Dermatology and Allergy, University Hospital, Bonn, Germany. .,Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland. .,Davos Biosciences, Davos, Switzerland.
| |
Collapse
|
127
|
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
|
128
|
Blicharz L, Rudnicka L, Czuwara J, Waśkiel-Burnat A, Goldust M, Olszewska M, Samochocki Z. The Influence of Microbiome Dysbiosis and Bacterial Biofilms on Epidermal Barrier Function in Atopic Dermatitis-An Update. Int J Mol Sci 2021; 22:ijms22168403. [PMID: 34445108 PMCID: PMC8395079 DOI: 10.3390/ijms22168403] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
Atopic dermatitis (AD) is a common inflammatory dermatosis affecting up to 30% of children and 10% of adults worldwide. AD is primarily driven by an epidermal barrier defect which triggers immune dysregulation within the skin. According to recent research such phenomena are closely related to the microbial dysbiosis of the skin. There is growing evidence that cutaneous microbiota and bacterial biofilms negatively affect skin barrier function, contributing to the onset and exacerbation of AD. This review summarizes the latest data on the mechanisms leading to microbiome dysbiosis and biofilm formation in AD, and the influence of these phenomena on skin barrier function.
Collapse
Affiliation(s)
- Leszek Blicharz
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
- Correspondence:
| | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| | - Joanna Czuwara
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| | - Anna Waśkiel-Burnat
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| | - Mohamad Goldust
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Małgorzata Olszewska
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| | - Zbigniew Samochocki
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| |
Collapse
|
129
|
Nakamura K, Williams MR, Kwiecinski JM, Horswill AR, Gallo RL. Staphylococcus aureus Enters Hair Follicles Using Triacylglycerol Lipases Preserved through the Genus Staphylococcus. J Invest Dermatol 2021; 141:2094-2097. [PMID: 33705795 PMCID: PMC8316282 DOI: 10.1016/j.jid.2021.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/25/2021] [Accepted: 02/10/2021] [Indexed: 11/16/2022]
Affiliation(s)
- Kouki Nakamura
- Department of Dermatology, University of California San Diego, San Diego, California, USA
| | - Michael R Williams
- Department of Dermatology, University of California San Diego, San Diego, California, USA
| | - Jakub M Kwiecinski
- Department of Immunology & Microbiology, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Alexander R Horswill
- Department of Immunology & Microbiology, University of Colorado School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard L Gallo
- Department of Dermatology, University of California San Diego, San Diego, California, USA.
| |
Collapse
|
130
|
Torres Salazar BO, Heilbronner S, Peschel A, Krismer B. Secondary Metabolites Governing Microbiome Interaction of Staphylococcal Pathogens and Commensals. Microb Physiol 2021; 31:198-216. [PMID: 34325424 DOI: 10.1159/000517082] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/03/2021] [Indexed: 11/19/2022]
Abstract
Various Staphylococcus species colonize skin and upper airways of warm-blooded animals. They compete successfully with many other microorganisms under the hostile and nutrient-poor conditions of these habitats using mechanisms that we are only beginning to appreciate. Small-molecule mediators, whose biosynthesis requires complex enzymatic cascades, so-called secondary metabolites, have emerged as crucial components of staphylococcal microbiome interactions. Such mediators belong to a large variety of compound classes and several of them have attractive properties for future drug development. They include, for instance, bacteriocins such as lanthipeptides, thiopeptides, and fibupeptides that inhibit bacterial competitor species; signaling molecules such as thiolactone peptides that induce or inhibit sensory cascades in other bacteria; or metallophores such as staphyloferrins and staphylopine that scavenge scant transition metal ions. For some secondary metabolites such as the aureusimines, the exact function remains to be elucidated. How secondary metabolites shape the fitness of Staphylococcus species in the complex context of other microbial and host defense factors remains a challenging field of future research. A detailed understanding will help to harness staphylococcal secondary metabolites for excluding the pathogenic species Staphylococcus aureus from the nasal microbiomes of at-risk patients, and it will be instrumental for the development of advanced anti-infective interventions.
Collapse
Affiliation(s)
- Benjamin O Torres Salazar
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany.,Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, Tübingen, Germany.,German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| | - Simon Heilbronner
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany.,Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, Tübingen, Germany.,German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| | - Andreas Peschel
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany.,Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, Tübingen, Germany.,German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| | - Bernhard Krismer
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany.,Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, Tübingen, Germany.,German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| |
Collapse
|
131
|
Hwang J, Thompson A, Jaros J, Blackcloud P, Hsiao J, Shi VY. Updated understanding of Staphylococcus aureus in atopic dermatitis: From virulence factors to commensals and clonal complexes. Exp Dermatol 2021; 30:1532-1545. [PMID: 34293242 DOI: 10.1111/exd.14435] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/18/2021] [Accepted: 07/20/2021] [Indexed: 12/22/2022]
Abstract
Atopic dermatitis (AD) is a common inflammatory dermatosis that has multiple contributing factors including genetic, immunologic and environmental. Staphylococcus aureus (SA) has long been associated with exacerbation of AD. SA produces many virulence factors that interact with the human skin and immune system. These superantigens and toxins have been shown to contribute to adhesion, inflammation and skin barrier destruction. Recent advances in genome sequencing techniques have led to a broadened understanding of the multiple ways SA interacts with the cutaneous environment in AD hosts. For example, temporal shifts in the microbiome, specifically in clonal complexes of SA, have been identified during AD flares and remission. Herein, we review mechanisms of interaction between the cutaneous microbiome and SA and highlight known differences in SA clonal complexes that contribute to AD pathogenesis. Detailed knowledge of the genetic strains of SA and cutaneous dysbiosis is becoming increasingly relevant in paving the way for microbiome-modulating and precision therapies for AD.
Collapse
Affiliation(s)
- Jonwei Hwang
- University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Alyssa Thompson
- College of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Joanna Jaros
- John H. Stroger Hospital Cook County Health Dermatology, Chicago, Illinois, USA
| | - Paul Blackcloud
- Division of Dermatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Jennifer Hsiao
- Division of Dermatology, University of California, Los Angeles, Los Angeles, California, USA
| | - Vivian Y Shi
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
132
|
Chinnappan M, Harris-Tryon TA. Novel mechanisms of microbial crosstalk with skin innate immunity. Exp Dermatol 2021; 30:1484-1495. [PMID: 34252227 DOI: 10.1111/exd.14429] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/28/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022]
Abstract
Skin is an organ with a dynamic ecosystem that harbours pathogenic and commensal microbes, which constantly communicate amongst each other and with the host immune system. Evolutionarily, skin and its microbiota have evolved to remain in homeostasis. However, frequently this homeostatic relationship is disturbed by a variety of factors such as environmental stress, diet, genetic mutations, and the microbiome itself. Commensal microbes also play a major role in the maintenance of microbial homeostasis. In addition to their ability to limit pathogens, many skin commensals such as Staphylococcus epidermidis and Cutibacterium acnes have recently been implicated in disease pathogenesis either by directly modulating the host immune components or by supporting the expansion of other pathogenic microbes. Likewise, opportunistic skin pathogens such as Staphylococcus aureus and Staphylococcus lugdunensis are able to breach the skin and cause disease. Though much has been established about the microbiota's function in skin immunity, we are in a time where newer mechanistic insights rapidly redefine our understanding of the host/microbial interface in the skin. In this review, we provide a concise summary of recent advances in our understanding of the interplay between host defense strategies and the skin microbiota.
Collapse
Affiliation(s)
- Mahendran Chinnappan
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tamia A Harris-Tryon
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
133
|
Schwierzeck V, Hülpüsch C, Reiger M. Microbiome of Barrier Organs in Allergy: Who Runs the World? Germs! Handb Exp Pharmacol 2021; 268:53-65. [PMID: 34228203 DOI: 10.1007/164_2021_478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Over the last few decades, allergic diseases have been steadily increasing worldwide, a phenomenon that is not yet completely understood. Recent evidence, however, suggests that alterations in the microbiome may be a contributing factor. The microbiome refers to all microorganisms in a habitat including bacteria, fungi, and viruses. Using modern sequencing technologies, we are now capable of detecting and analyzing the human microbiome in more detail than ever before. Epidemiological and experimental studies have indicated that a complex intestinal microbiome supports the development of the immune system during childhood, thus providing protection from allergic diseases, including food allergy. The microbiome becomes an important part of human physiology and forms dynamic relationships with our various barrier systems. For example, bacterial dysbiosis is a hallmark of atopic eczema and correlates with disease progression. Similarly, the lung and nasopharyngeal microbiome is altered in patients with asthma and allergic rhinitis. While these results are interesting, the underlying mechanisms are still unclear and need to be investigated with functional studies. This review gives a short overview of the terminology and methods used in microbiome research before highlighting results concerning the lung, skin, and intestinal microbiome in allergic diseases.
Collapse
Affiliation(s)
- Vera Schwierzeck
- Institute of Hygiene, University Hospital Muenster, Munster, Germany
| | - Claudia Hülpüsch
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum Muenchen, Augsburg, Germany.,CK CARE - Christine Kuehne Center for Allergy Research and Education, Davos, Switzerland
| | - Matthias Reiger
- Department of Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany. .,Institute of Environmental Medicine, Helmholtz Zentrum Muenchen, Augsburg, Germany. .,CK CARE - Christine Kuehne Center for Allergy Research and Education, Davos, Switzerland.
| |
Collapse
|
134
|
Elias AE, McBain AJ, O'Neill CA. The role of the skin microbiota in the modulation of cutaneous inflammation-Lessons from the gut. Exp Dermatol 2021; 30:1509-1516. [PMID: 34173265 DOI: 10.1111/exd.14420] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/02/2021] [Accepted: 06/17/2021] [Indexed: 12/16/2022]
Abstract
Inflammation is a vital defense mechanism used to protect the body from invading pathogens, but dysregulation can lead to chronic inflammatory disorders such as psoriasis and atopic dermatitis. Differences in microbiota composition have been observed in patients with inflammatory skin conditions compared with healthy individuals, particularly within lesions. There is also increasing evidence accumulating to support the notion that the microbiome contributes to the onset or modulates the severity of inflammatory diseases. Despite the known protective effects of orally administered lactic acid bacteria against inflammation, few studies have investigated the potential protective effects of topical application of bacteria on skin health and even fewer have looked at the potential anti-inflammatory effects of skin commensals. If lack of diversity and reduction in the abundance of specific commensal strains is observed in inflammatory skin lesions, and it is known that commensal bacteria can produce anti-inflammatory compounds, we suggest that certain members of the skin microbiota have anti-inflammatory properties that can be harnessed for use as topical therapeutics in inflammatory skin disorders.
Collapse
Affiliation(s)
- Abigail E Elias
- Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Andrew J McBain
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Catherine A O'Neill
- Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
135
|
Nakatsuji T, Gallo RL, Shafiq F, Tong Y, Chun K, Butcher AM, Cheng JY, Hata TR. Use of Autologous Bacteriotherapy to Treat Staphylococcus aureus in Patients With Atopic Dermatitis: A Randomized Double-blind Clinical Trial. JAMA Dermatol 2021; 157:2781297. [PMID: 34132739 PMCID: PMC8209585 DOI: 10.1001/jamadermatol.2021.1311] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/21/2021] [Indexed: 12/30/2022]
Abstract
IMPORTANCE Atopic dermatitis (AD) can be negatively affected by Staphylococcus aureus. The skin microbiome of AD is deficient in coagulase-negative Staphylococcus (CoNS) that can kill S aureus. OBJECTIVE To evaluate if the antimicrobial-producing CoNS (CoNS-AM+) of a patient with AD can be autologously reintroduced to the same patient to inhibit survival of S aureus and improve clinical outcomes. DESIGN, SETTING, AND PARTICIPANTS This double-blind, vehicle-controlled, single-center randomized clinical trial of 11 adult patients with moderate to severe AD who were randomized to receive either an autologous CoNS-AM+ (n = 5) or the vehicle (n = 6) was conducted between April 2016 and May 2018. The data were analyzed from May 2018 to July 2019. INTERVENTIONS Autologous CoNS-AM+ was isolated from swabs that were obtained from the nonlesional skin of each patient with AD, expanded by culture, and then reapplied topically to the forearms at a concentration of 107 colony-forming units/g. MAIN OUTCOMES AND MEASURES The primary end point of this study was to assess S aureus abundance after 1 week of application of autologous CoNS-AM+ on patients with AD by culture-based and DNA-based methods. The secondary end points were to assess the safety and clinical outcomes. RESULTS Eleven patients (4 men [36.4%] and 7 women [63/6%]) were recruited based on the inclusion criteria. There were no serious adverse events in groups treated with autologous CoNS-AM+ or the vehicle. Staphylococcus aureus colonization on lesional skin at the end of treatment on patients who were treated with autologous CoNS-AM+ (mean of log10 ratio to baseline, -1.702; 95% CI, -2.882 to -0.523) was reduced by 99.2% compared with vehicle treatment (mean of log10 ratio to baseline, 0.671; 95% CI, -0.289 to 1.613; P = .01) and persisted for 4 days after treatment (CoNS-AM+: mean of log10 ratio to baseline, -1.752; 95% CI, -3.051 to -0.453; vehicle: mean of log10 ratio to baseline, -0.003; 95% CI, -1.083 to 1.076; P = .03). Importantly, local Eczema Area And Severity Index scores that were assessed at day 11 on patients who received CoNS-AM+ (mean of percentage change, -48.45; 95% CI, -84.34 to -12.55) were significantly improved compared with vehicle treatment (mean of percentage change, -4.52; 95% CI, -36.25 to 27.22; P = .04). CONCLUSIONS AND RELEVANCE The data from this randomized clinical trial suggest that bacteriotherapy with an autologous strain of skin commensal bacteria can safely decrease S aureus colonization and improve disease severity. Although larger studies will be needed, this personalized approach for S aureus reduction may provide an alternative treatment for patients with AD beyond antibiotics, immunosuppression, and immunomodulation. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT03158012.
Collapse
Affiliation(s)
- Teruaki Nakatsuji
- Department of Dermatology, University of California, San Diego, La Jolla
| | - Richard L. Gallo
- Department of Dermatology, University of California, San Diego, La Jolla
| | - Faiza Shafiq
- Department of Dermatology, University of California, San Diego, La Jolla
| | - Yun Tong
- Department of Dermatology, University of California, San Diego, La Jolla
| | - Kimberly Chun
- Department of Dermatology, University of California, San Diego, La Jolla
| | - Anna M. Butcher
- Department of Dermatology, University of California, San Diego, La Jolla
| | - Joyce Y. Cheng
- Department of Dermatology, University of California, San Diego, La Jolla
| | - Tissa R. Hata
- Department of Dermatology, University of California, San Diego, La Jolla
| |
Collapse
|
136
|
[The skin microbiome-useful for diagnosis and therapy?]. Hautarzt 2021; 72:579-585. [PMID: 34115159 DOI: 10.1007/s00105-021-04830-4] [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: 05/05/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Our skin is a very important and complex organ of the body. The microorganisms of the skin, the so-called microbiome, represent an important part of the healthy skin barrier and are influenced by various external and internal factors. AIM The question to what extent the skin microbiome represents a diagnostic or even therapeutic target in the context of skin diseases is discussed. MATERIALS AND METHODS A literature search was performed. RESULTS Several diseases are associated with negative alterations of the skin microbiome. In atopic dermatitis, a correlation between severity and increased availability of Staphylococcus aureus is known, with a loss of bacterial diversity on the skin. In the future, S. aureus will not only be used as a diagnostic marker in atopic dermatitis, but also represents a promising target as a predictive marker for therapeutic success. The role of the skin microbiome in psoriasis has not yet been researched in depth. However, there is evidence that dysbiosis of the skin microbiome contributes to the course of psoriasis and that there is a disturbance in immune tolerance in patients. In the case of acne, the involvement of Cutibacterium acnes in the clinical picture is well known; however, recent findings show that it is not sufficient to identify the species, but certain characteristics of C. acnes strains are associated. CONCLUSION Microbial biomarkers are currently only established in atopic dermatitis. For other diseases, this might be the case in the future; however combinations of microorganisms, single species and also strains with specific characteristics must be considered.
Collapse
|
137
|
Boxberger M, Cenizo V, Cassir N, La Scola B. Challenges in exploring and manipulating the human skin microbiome. MICROBIOME 2021; 9:125. [PMID: 34053468 PMCID: PMC8166136 DOI: 10.1186/s40168-021-01062-5] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 03/25/2021] [Indexed: 05/08/2023]
Abstract
The skin is the exterior interface of the human body with the environment. Despite its harsh physical landscape, the skin is colonized by diverse commensal microbes. In this review, we discuss recent insights into skin microbial populations, including their composition and role in health and disease and their modulation by intrinsic and extrinsic factors, with a focus on the pathobiological basis of skin aging. We also describe the most recent tools for investigating the skin microbiota composition and microbe-skin relationships and perspectives regarding the challenges of skin microbiome manipulation. Video abstract.
Collapse
Affiliation(s)
- Manon Boxberger
- IRD, AP-HM, MEPHI, Aix Marseille Université, Marseille, France
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13385 Marseille Cedex 05, France
| | - Valérie Cenizo
- Groupe L’Occitane, R&D Department, Zone Industrielle Saint Maurice, 4100 Manosque, Alpes-de Haute-Provence France
| | - Nadim Cassir
- IRD, AP-HM, MEPHI, Aix Marseille Université, Marseille, France
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13385 Marseille Cedex 05, France
| | - Bernard La Scola
- IRD, AP-HM, MEPHI, Aix Marseille Université, Marseille, France
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13385 Marseille Cedex 05, France
- IRD, AP-HM, SSA, VITROME, Aix Marseille Université, Marseille, France
| |
Collapse
|
138
|
Boero E, Mnich ME, Manetti AGO, Soldaini E, Grimaldi L, Bagnoli F. Human Three-Dimensional Models for Studying Skin Pathogens. Curr Top Microbiol Immunol 2021; 430:3-27. [PMID: 32601967 DOI: 10.1007/82_2020_219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Skin is the most exposed surface of the human body, separating the microbe-rich external environment, from the sterile inner part. When skin is breached or its homeostasis is perturbed, bacterial, fungal and viral pathogens can cause local infections or use the skin as an entry site to spread to other organs. In the last decades, it has become clear that skin provides niches for permanent microbial colonization, and it actively interacts with microorganisms. This crosstalk promotes skin homeostasis and immune maturation, preventing expansion of harmful organisms. Skin commensals, however, are often found to be skin most prevalent and dangerous pathogens. Despite the medical interest, mechanisms of colonization and invasion for most skin pathogens are poorly understood. This limitation is due to the lack of reliable skin models. Indeed, animal models do not adequately mimic neither the anatomy nor the immune response of human skin. Human 3D skin models overcome these limitations and can provide new insights into the molecular mechanisms of microbial pathogenesis. Herein, we address the strengths and weaknesses of different types of human skin models and we review the main findings obtained using these models to study skin pathogens.
Collapse
Affiliation(s)
| | | | | | | | - Luca Grimaldi
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | | |
Collapse
|
139
|
Abstract
The skin microbiome is an ecosystem comprised of a multitude of microbial species interacting with their surroundings, including other microbes and host epithelial and immune cells. These interactions are the basis of important roles within the skin microbiome that provide benefit to the host, boosting multiple aspects of barrier function, a critical function of this essential organ. However, with reward always comes risk; resident skin microbes function in a context-dependent manner, set on the backdrop of a dynamic host and microbial milieu. Here, we discuss the reward of hosting a microbial ecosystem on the skin, including protection from pathogens and tuning of the skin microenvironment. We also give consideration to how these skin residents, often termed "commensals" can cause disorder, damage, and promote skin disease.
Collapse
Affiliation(s)
- Laurice Flowers
- Department of Dermatology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Elizabeth A Grice
- Department of Dermatology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
140
|
The Ambivalent Role of Skin Microbiota and Adrenaline in Wound Healing and the Interplay between Them. Int J Mol Sci 2021; 22:ijms22094996. [PMID: 34066786 PMCID: PMC8125934 DOI: 10.3390/ijms22094996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023] Open
Abstract
After skin injury, wound healing sets into motion a dynamic process to repair and replace devitalized tissues. The healing process can be divided into four overlapping phases: hemostasis, inflammation, proliferation, and maturation. Skin microbiota has been reported to participate in orchestrating the wound healing both in negative and positive ways. Many studies reported that skin microbiota can impose negative and positive effects on the wound. Recent findings have shown that many bacterial species on human skin are able to convert aromatic amino acids into so-called trace amines (TAs) and convert corresponding precursors into dopamine and serotonin, which are all released into the environment. As a stress reaction, wounded epithelial cells release the hormone adrenaline (epinephrine), which activates the β2-adrenergic receptor (β2-AR), impairing the migration ability of keratinocytes and thus re-epithelization. This is where TAs come into play, as they act as antagonists of β2-AR and thus attenuate the effects of adrenaline. The result is that not only TAs but also TA-producing skin bacteria accelerate wound healing. Adrenergic receptors (ARs) play a key role in many physiological and disease-related processes and are expressed in numerous cell types. In this review, we describe the role of ARs in relation to wound healing in keratinocytes, immune cells, fibroblasts, and blood vessels and the possible role of the skin microbiota in wound healing.
Collapse
|
141
|
Nakatsuji T, Hata TR, Tong Y, Cheng JY, Shafiq F, Butcher AM, Salem SS, Brinton SL, Rudman Spergel AK, Johnson K, Jepson B, Calatroni A, David G, Ramirez-Gama M, Taylor P, Leung DYM, Gallo RL. Development of a human skin commensal microbe for bacteriotherapy of atopic dermatitis and use in a phase 1 randomized clinical trial. Nat Med 2021; 27:700-709. [PMID: 33619370 PMCID: PMC8052297 DOI: 10.1038/s41591-021-01256-2] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 01/15/2021] [Indexed: 01/31/2023]
Abstract
Staphylococcus aureus colonizes patients with atopic dermatitis (AD) and exacerbates disease by promoting inflammation. The present study investigated the safety and mechanisms of action of Staphylococcus hominis A9 (ShA9), a bacterium isolated from healthy human skin, as a topical therapy for AD. ShA9 killed S. aureus on the skin of mice and inhibited expression of a toxin from S. aureus (psmα) that promotes inflammation. A first-in-human, phase 1, double-blinded, randomized 1-week trial of topical ShA9 or vehicle on the forearm skin of 54 adults with S. aureus-positive AD (NCT03151148) met its primary endpoint of safety, and participants receiving ShA9 had fewer adverse events associated with AD. Eczema severity was not significantly different when evaluated in all participants treated with ShA9 but a significant decrease in S. aureus and increased ShA9 DNA were seen and met secondary endpoints. Some S. aureus strains on participants were not directly killed by ShA9, but expression of mRNA for psmα was inhibited in all strains. Improvement in local eczema severity was suggested by post-hoc analysis of participants with S. aureus directly killed by ShA9. These observations demonstrate the safety and potential benefits of bacteriotherapy for AD.
Collapse
Affiliation(s)
- Teruaki Nakatsuji
- Department of Dermatology, University of California, San Diego, La Jolla, CA, USA
| | - Tissa R Hata
- Department of Dermatology, University of California, San Diego, La Jolla, CA, USA
| | - Yun Tong
- Department of Dermatology, University of California, San Diego, La Jolla, CA, USA
| | - Joyce Y Cheng
- Department of Dermatology, University of California, San Diego, La Jolla, CA, USA
| | - Faiza Shafiq
- Department of Dermatology, University of California, San Diego, La Jolla, CA, USA
| | - Anna M Butcher
- Department of Dermatology, University of California, San Diego, La Jolla, CA, USA
| | - Secilia S Salem
- Department of Dermatology, University of California, San Diego, La Jolla, CA, USA
| | - Samantha L Brinton
- Department of Dermatology, University of California, San Diego, La Jolla, CA, USA
| | - Amanda K Rudman Spergel
- Division of Allergy, Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Keli Johnson
- Rho Federal Systems Division, Inc., Durham, NC, USA
| | - Brett Jepson
- Rho Federal Systems Division, Inc., Durham, NC, USA
| | | | - Gloria David
- Rho Federal Systems Division, Inc., Durham, NC, USA
| | - Marco Ramirez-Gama
- Division of Allergy and Immunology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Patricia Taylor
- Division of Allergy and Immunology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Donald Y M Leung
- Division of Allergy and Immunology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
142
|
Williams MR, Cau L, Wang Y, Kaul D, Sanford JA, Zaramela LS, Khalil S, Butcher AM, Zengler K, Horswill AR, Dupont CL, Hovnanian A, Gallo RL. Interplay of Staphylococcal and Host Proteases Promotes Skin Barrier Disruption in Netherton Syndrome. Cell Rep 2021; 30:2923-2933.e7. [PMID: 32130897 PMCID: PMC7183042 DOI: 10.1016/j.celrep.2020.02.021] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/04/2019] [Accepted: 02/05/2020] [Indexed: 01/09/2023] Open
Abstract
Netherton syndrome (NS) is a monogenic skin disease resulting from loss of function of lymphoepithelial Kazal-type-related protease inhibitor (LEKTI-1). In this study we examine if bacteria residing on the skin are influenced by the loss of LEKTI-1 and if interaction between this human gene and resident bacteria contributes to skin disease. Shotgun sequencing of the skin microbiome demonstrates that lesional skin of NS subjects is dominated by Staphylococcus aureus (S. aureus) and Staphylococcus epidermidis (S. epidermidis). Isolates of either species from NS subjects are able to induce skin inflammation and barrier damage on mice. These microbes promote skin inflammation in the setting of LEKTI-1 deficiency due to excess proteolytic activity promoted by S. aureus phenol-soluble modulin α as well as increased bacterial proteases staphopain A and B from S. aureus or EcpA from S. epidermidis. These findings demonstrate the critical need for maintaining homeostasis of host and microbial proteases to prevent a human skin disease.
Collapse
Affiliation(s)
- Michael R Williams
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA.
| | - Laura Cau
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA; SILAB, R&D Department, Brive, France.
| | - Yichen Wang
- INSERM, UMR 1163, Laboratory of Genetic Skin Diseases, Imagine Institute and Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Drishti Kaul
- J. Craig Venter Institute, La Jolla, CA 92093, USA
| | - James A Sanford
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Livia S Zaramela
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Shadi Khalil
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA; University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Anna M Butcher
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Karsten Zengler
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, San Diego, CA 92093, USA; Department of Bioengineering, University of California, San Diego, CA 92093, USA
| | - Alexander R Horswill
- Department of Veterans Affairs Denver Health Care System, Denver, CO, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora 80045, CO, USA
| | | | - Alain Hovnanian
- INSERM, UMR 1163, Laboratory of Genetic Skin Diseases, Imagine Institute and Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA; Center for Microbiome Innovation, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
143
|
He J, Jia Y. Application of omics technologies in dermatological research and skin management. J Cosmet Dermatol 2021; 21:451-460. [PMID: 33759323 DOI: 10.1111/jocd.14100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/19/2021] [Accepted: 03/10/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND "Omics" are usually based on the use of high-throughput analysis methods for global analysis of biological samples and the discovery of biomarkers, and may provide new insights into biological phenomena. Over the last few years, the development of omics technologies has considerably accelerated the pace of dermatological research. AIMS The purpose of this article was to review the development of omics in recent decades and their application in dermatological research. METHODS An extensive literature search was conducted on omics technologies since the first research on omics. RESULTS This article summarizes the history and main research methods of the six omics technologies, including genomics, transcriptomics, proteomics, metabolomics, lipidomics, and microbiomics. Their application in certain skin diseases and cosmetics research and development are also summarized. CONCLUSIONS This information will help to understand the mechanism of some skin diseases and the discovery of potential biomarkers, and provide new insights for skin health management and cosmetics research and development.
Collapse
Affiliation(s)
- Jianbiao He
- Beijing Key Laboratory of Plant Resources Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, China.,Key Laboratory of Cosmetic of China National Light Industry, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, China.,Institute of cosmetic regulatory science, Beijing Technology and Business University, Beijing, China
| | - Yan Jia
- Beijing Key Laboratory of Plant Resources Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, China.,Key Laboratory of Cosmetic of China National Light Industry, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, China.,Institute of cosmetic regulatory science, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
144
|
Liu J, Ting JP, Al-Azzam S, Ding Y, Afshar S. Therapeutic Advances in Diabetes, Autoimmune, and Neurological Diseases. Int J Mol Sci 2021; 22:ijms22062805. [PMID: 33802091 PMCID: PMC8001105 DOI: 10.3390/ijms22062805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 02/08/2023] Open
Abstract
Since 2015, 170 small molecules, 60 antibody-based entities, 12 peptides, and 15 gene- or cell-therapies have been approved by FDA for diverse disease indications. Recent advancement in medicine is facilitated by identification of new targets and mechanisms of actions, advancement in discovery and development platforms, and the emergence of novel technologies. Early disease detection, precision intervention, and personalized treatments have revolutionized patient care in the last decade. In this review, we provide a comprehensive overview of current and emerging therapeutic modalities developed in the recent years. We focus on nine diseases in three major therapeutics areas, diabetes, autoimmune, and neurological disorders. The pathogenesis of each disease at physiological and molecular levels is discussed and recently approved drugs as well as drugs in the clinic are presented.
Collapse
Affiliation(s)
- Jinsha Liu
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Joey Paolo Ting
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Shams Al-Azzam
- Professional Scientific Services, Eurofins Lancaster Laboratories, Lancaster, PA 17605, USA;
| | - Yun Ding
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Sepideh Afshar
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
- Correspondence:
| |
Collapse
|
145
|
Cau L, Williams MR, Butcher AM, Nakatsuji T, Kavanaugh JS, Cheng JY, Shafiq F, Higbee K, Hata TR, Horswill AR, Gallo RL. Staphylococcus epidermidis protease EcpA can be a deleterious component of the skin microbiome in atopic dermatitis. J Allergy Clin Immunol 2021; 147:955-966.e16. [PMID: 32634452 PMCID: PMC8058862 DOI: 10.1016/j.jaci.2020.06.024] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 05/19/2020] [Accepted: 06/09/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Staphylococcus aureus and Staphylococcus epidermidis are the most abundant bacteria found on the skin of patients with atopic dermatitis (AD). S aureus is known to exacerbate AD, whereas S epidermidis has been considered a beneficial commensal organism. OBJECTIVE In this study, we hypothesized that S epidermidis could promote skin damage in AD by the production of a protease that damages the epidermal barrier. METHODS The protease activity of S epidermidis isolates was compared with that of other staphylococcal species. The capacity of S epidermidis to degrade the barrier and induce inflammation was examined by using human keratinocyte tissue culture and mouse models. Skin swabs from atopic and healthy adult subjects were analyzed for the presence of S epidermidis genomic DNA and mRNA. RESULTS S epidermidis strains were observed to produce strong cysteine protease activity when grown at high density. The enzyme responsible for this activity was identified as EcpA, a cysteine protease under quorum sensing control. EcpA was shown to degrade desmoglein-1 and LL-37 in vitro, disrupt the physical barrier, and induce skin inflammation in mice. The abundance of S epidermidis and expression of ecpA mRNA were increased on the skin of some patients with AD, and this correlated with disease severity. Another commensal skin bacterial species, Staphylococcus hominis, can inhibit EcpA production by S epidermidis. CONCLUSION S epidermidis has commonly been regarded as a beneficial skin microbe, whereas S aureus has been considered deleterious. This study suggests that the overabundance of S epidermidis found on some atopic patients can act similarly to S aureus and damage the skin by expression of a cysteine protease.
Collapse
Affiliation(s)
- Laura Cau
- Department of Dermatology, University of California San Diego, San Diego, Calif; R&D Department, SILAB, Brive, France
| | - Michael R Williams
- Department of Dermatology, University of California San Diego, San Diego, Calif
| | - Anna M Butcher
- Department of Dermatology, University of California San Diego, San Diego, Calif
| | - Teruaki Nakatsuji
- Department of Dermatology, University of California San Diego, San Diego, Calif
| | - Jeffrey S Kavanaugh
- Department of Immunology and Microbiology, University of Colorado Anschutz, Medical Campus, Aurora, Colo
| | - Joyce Y Cheng
- Department of Dermatology, University of California San Diego, San Diego, Calif
| | - Faiza Shafiq
- Department of Dermatology, University of California San Diego, San Diego, Calif
| | - Kyle Higbee
- Department of Dermatology, University of California San Diego, San Diego, Calif
| | - Tissa R Hata
- Department of Dermatology, University of California San Diego, San Diego, Calif
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado Anschutz, Medical Campus, Aurora, Colo; Department of Veterans Affairs Eastern Colorado Health Care System, Aurora, Colo
| | - Richard L Gallo
- Department of Dermatology, University of California San Diego, San Diego, Calif; Center for Microbiome Innovation, University of California San Diego, San Diego, Calif.
| |
Collapse
|
146
|
Edslev SM, Olesen CM, Nørreslet LB, Ingham AC, Iversen S, Lilje B, Clausen ML, Jensen JS, Stegger M, Agner T, Andersen PS. Staphylococcal Communities on Skin Are Associated with Atopic Dermatitis and Disease Severity. Microorganisms 2021; 9:microorganisms9020432. [PMID: 33669791 PMCID: PMC7921937 DOI: 10.3390/microorganisms9020432] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/16/2022] Open
Abstract
The skin microbiota of atopic dermatitis (AD) patients is characterized by increased Staphylococcus aureus colonization, which exacerbates disease symptoms and has been linked to reduced bacterial diversity. Skin bacterial communities in AD patients have mostly been described at family and genus levels, while species-level characterization has been limited. In this study, we investigated the role of the bacteria belonging to the Staphylococcus genus using targeted sequencing of the tuf gene with genus-specific primers. We compared staphylococcal communities on lesional and non-lesional skin of AD patients, as well as AD patients with healthy controls, and determined the absolute abundance of bacteria present at each site. We observed that the staphylococcal community, bacterial alpha diversity, and bacterial densities were similar on lesional and non-lesional skin, whereas AD severity was associated with significant changes in staphylococcal composition. Increased S. aureus, Staphylococcus capitis, and Staphylococcus lugdunensis abundances were correlated with increased severity. Conversely, Staphylococcus hominis abundance was negatively correlated with severity. Furthermore, S. hominis relative abundance was reduced on AD skin compared to healthy skin. In conclusion, various staphylococcal species appear to be important for skin health.
Collapse
Affiliation(s)
- Sofie Marie Edslev
- Bacteria, Parasites, and Fungi, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark; (A.C.I.); (S.I.); (B.L.); (J.S.J.); (M.S.); (P.S.A.)
- Correspondence:
| | - Caroline Meyer Olesen
- Department of Dermatology, Bispebjerg Hospital, Bispebjerg bakke 23, 2400 Copenhagen, Denmark; (C.M.O.); (L.B.N.); (M.-L.C.); (T.A.)
| | - Line Brok Nørreslet
- Department of Dermatology, Bispebjerg Hospital, Bispebjerg bakke 23, 2400 Copenhagen, Denmark; (C.M.O.); (L.B.N.); (M.-L.C.); (T.A.)
| | - Anna Cäcilia Ingham
- Bacteria, Parasites, and Fungi, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark; (A.C.I.); (S.I.); (B.L.); (J.S.J.); (M.S.); (P.S.A.)
| | - Søren Iversen
- Bacteria, Parasites, and Fungi, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark; (A.C.I.); (S.I.); (B.L.); (J.S.J.); (M.S.); (P.S.A.)
| | - Berit Lilje
- Bacteria, Parasites, and Fungi, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark; (A.C.I.); (S.I.); (B.L.); (J.S.J.); (M.S.); (P.S.A.)
| | - Maja-Lisa Clausen
- Department of Dermatology, Bispebjerg Hospital, Bispebjerg bakke 23, 2400 Copenhagen, Denmark; (C.M.O.); (L.B.N.); (M.-L.C.); (T.A.)
| | - Jørgen Skov Jensen
- Bacteria, Parasites, and Fungi, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark; (A.C.I.); (S.I.); (B.L.); (J.S.J.); (M.S.); (P.S.A.)
| | - Marc Stegger
- Bacteria, Parasites, and Fungi, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark; (A.C.I.); (S.I.); (B.L.); (J.S.J.); (M.S.); (P.S.A.)
| | - Tove Agner
- Department of Dermatology, Bispebjerg Hospital, Bispebjerg bakke 23, 2400 Copenhagen, Denmark; (C.M.O.); (L.B.N.); (M.-L.C.); (T.A.)
| | - Paal Skytt Andersen
- Bacteria, Parasites, and Fungi, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark; (A.C.I.); (S.I.); (B.L.); (J.S.J.); (M.S.); (P.S.A.)
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 15, 1870 Frederiksberg, Denmark
| |
Collapse
|
147
|
Sotiropoulou G, Zingkou E, Pampalakis G. Redirecting drug repositioning to discover innovative cosmeceuticals. Exp Dermatol 2021; 30:628-644. [PMID: 33544970 DOI: 10.1111/exd.14299] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/23/2021] [Accepted: 02/01/2021] [Indexed: 12/18/2022]
Abstract
Skin appearance is essential for self-esteem and quality of life; consequently, skin care products represent a huge market. In particular, cosmeceuticals constitute a hybrid category of skin care formulations, at the interphase of cosmetics and pharmaceuticals, rationally designed to target (patho) physiological mechanisms aiming to enhance skin health and appearance. Cosmeceuticals are marketed as anti-ageing, anti-wrinkle, hair regrowth, skin whitening and wound healing agents with special emphasis on scar-free healing. An overview on recent cutting-edge advances concerning the discovery and development of enhanced performance cosmeceuticals by drug repositioning approaches is presented here. In this context, we propose "target repositioning," a new term, to highlight that druggable protein targets implicated in multiple diseases (hubs in the diseasome) can be exploited to accelerate the discovery of molecularly targeted cosmeceuticals that can promote skin health as an added benefit, which is a novel concept not described before. In this direction, emphasis is placed on the role of mouse models, for often untreatable skin diseases, as well as recent breakthroughs on monogenic rare skin syndromes, in promoting compound repositioning to innovative cosmeceuticals.
Collapse
Affiliation(s)
- Georgia Sotiropoulou
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, Greece
| | - Eleni Zingkou
- Department of Pharmacy, School of Health Sciences, University of Patras, Rion-Patras, Greece
| | - Georgios Pampalakis
- Department of Pharmacognosy-Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
148
|
Preda M, Mihai MM, Popa LI, Dițu LM, Holban AM, Manolescu LSC, Popa GL, Muntean AA, Gheorghe I, Chifiriuc CM, Popa MI. Phenotypic and genotypic virulence features of staphylococcal strains isolated from difficult-to-treat skin and soft tissue infections. PLoS One 2021; 16:e0246478. [PMID: 33529240 PMCID: PMC7853507 DOI: 10.1371/journal.pone.0246478] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/19/2021] [Indexed: 12/04/2022] Open
Abstract
Chronic infections represent an important burden on the healthcare system and have a significant impact on the patients’ quality of life. While Staphylococcus spp. are commensal bacteria, they can become pathogenic, leading to various types of infections. In this study we aimed to characterize the virulence profiles of staphylococcal strains involved in difficult-to-treat skin and soft tissue infections, from both phenotypic and genotypic points of view. Phenotypic ability of the strains to secrete soluble virulence factors was assessed by a culturing dependent assay and their capacity to develop biofilms on inert substrate was screened by an adapted crystal violet microtiter method. We also tested the presence of several virulence genes by PCR. Most of the studied strains were isolated from purulent secretions of acne lesions and frequently secreted two or three soluble virulence factors. Most frequently secreted soluble virulence factors were caseinase (89%), lipase (71%) and lecithinase (67%). Almost half of the strains produced a well-represented biofilm. The molecular characterization showed the presence of the genes cna, hlg, clfA, and clfB. Staphylococcal strains that produce difficult-to-treat skin and soft tissue infections seem to be characterized by an enhanced ability to produce different soluble virulence factors and to develop biofilms in vitro. Further studies need to be developed in other Staphylococcus spp. infections in order to confirm this hypothesis.
Collapse
Affiliation(s)
- Mădălina Preda
- Department of Microbiology, Parasitology and Virology, Faculty of Midwives and Nursing, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
- ‘Cantacuzino’ National Medico-Military Research and Development Institute, Bucharest, Romania
| | - Mara Mădălina Mihai
- Department of Oncologic Dermatology, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
- Department of Dermatology, ‘Elias’ University Emergency Hospital, Bucharest, Romania
- * E-mail: (MMM); (LIP)
| | - Laura Ioana Popa
- Department of Bioinformatics, The National Institute of Research and Development for Biological Sciences, Bucharest, Romania
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- * E-mail: (MMM); (LIP)
| | - Lia-Mara Dițu
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Alina Maria Holban
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Loredana Sabina Cornelia Manolescu
- Department of Microbiology, Parasitology and Virology, Faculty of Midwives and Nursing, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
| | - Gabriela-Loredana Popa
- Department of Microbiology, Faculty of Dental Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Irina Gheorghe
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Carmen Mariana Chifiriuc
- Research Institute of the University of Bucharest (ICUB), Bucharest, Romania
- Department of Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Mircea-Ioan Popa
- ‘Cantacuzino’ National Medico-Military Research and Development Institute, Bucharest, Romania
- Department of Microbiology, Faculty of Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
149
|
Zhao Y, Han Y, Sun Y, Wei Z, Chen J, Niu X, An Q, Zhang L, Qi R, Gao X. Comprehensive Succinylome Profiling Reveals the Pivotal Role of Lysine Succinylation in Energy Metabolism and Quorum Sensing of Staphylococcus epidermidis. Front Microbiol 2021; 11:632367. [PMID: 33597936 PMCID: PMC7882547 DOI: 10.3389/fmicb.2020.632367] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
Background Lysine succinylation is a newly identified posttranslational modification (PTM), which exists widely from prokaryotes to eukaryotes and participates in various cellular processes, especially in the metabolic processes. Staphylococcus epidermidis is a commensal bacterium in the skin, which attracts more attention as a pathogen, especially in immunocompromised patients and neonates by attaching to medical devices and forming biofilms. However, the significance of lysine succinylation in S. epidermidis proteins has not been investigated. Objectives The purpose of this study was to investigate the physiological and pathological processes of S. epidermidis at the level of PTM. Moreover, by analyzing previous succinylome datasets in various organisms, we tried to provide an in-depth understanding of lysine succinylation. Methods Using antibody affinity enrichment followed by LC-MS/MS analysis, we examined the succinylome of S. epidermidis (ATCC 12228). Then, bioinformatics analysis was performed, including Gene Ontology (GO), KEGG enrichment, motif characterization, secondary structure, protein–protein interaction, and BLAST analysis. Results A total of 1557 succinylated lysine sites in 649 proteins were identified in S. epidermidis (ATCC 12228). Among these succinylation proteins, GO annotation showed that proteins related to metabolic processes accounted for the most. KEGG pathway characterization indicated that proteins associated with the glycolysis/gluconeogenesis and citrate cycle (TCA cycle) pathway were more likely to be succinylated. Moreover, 13 conserved motifs were identified. The specific motif KsuD was conserved in model prokaryotes and eukaryotes. Succinylated proteins with this motif were highly enriched in the glycolysis/gluconeogenesis pathway. One succinylation site (K144) was identified in S-ribosylhomocysteine lyase, a key enzyme in the quorum sensing system, indicating the regulatory role succinylation may play in bacterial processes. Furthermore, 15 succinyltransferases and 18 desuccinylases (erasers) were predicted in S. epidermidis by BLAST analysis. Conclusion We performed the first comprehensive profile of succinylation in S. epidermidis and illustrated the significant role succinylation may play in energy metabolism, QS system, and other bacterial behaviors. This study may be a fundamental basis to investigate the underlying mechanisms of colonization, virulence, and infection of S. epidermidis, as well as provide a new insight into regulatory effects succinylation may lay on metabolic processes (Data are available via ProteomeXchange with identifier PXD022866).
Collapse
Affiliation(s)
- Yiping Zhao
- Key Laboratory of Immunodermatology, Department of Dermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Health and Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yang Han
- Department of Dermatology, Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| | - Yuzhe Sun
- Department of Dermatology, Dermatological Hospital, Southern Medical University, Guangzhou, China
| | - Zhendong Wei
- Department of Dermatology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jialong Chen
- Key Laboratory of Immunodermatology, Department of Dermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Health and Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xueli Niu
- Key Laboratory of Immunodermatology, Department of Dermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Health and Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Qian An
- Key Laboratory of Immunodermatology, Department of Dermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Health and Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Li Zhang
- Key Laboratory of Immunodermatology, Department of Dermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Health and Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ruiqun Qi
- Key Laboratory of Immunodermatology, Department of Dermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Health and Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xinghua Gao
- Key Laboratory of Immunodermatology, Department of Dermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Health and Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
150
|
Anforderungen an die Infektionsprävention bei der medizinischen Versorgung von immunsupprimierten Patienten. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2021; 64:232-264. [PMID: 33394069 PMCID: PMC7780910 DOI: 10.1007/s00103-020-03265-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|