1
|
Traidl-Hoffmann C, Afghani J, Akdis CA, Akdis M, Aydin H, Bärenfaller K, Behrendt H, Bieber T, Bigliardi P, Bigliardi-Qi M, Bonefeld CM, Bösch S, Brüggen MC, Diemert S, Duchna HW, Fähndrich M, Fehr D, Fellmann M, Frei R, Garvey LH, Gharbo R, Gökkaya M, Grando K, Guillet C, Guler E, Gutermuth J, Herrmann N, Hijnen DJ, Hülpüsch C, Irvine AD, Jensen-Jarolim E, Kong HH, Koren H, Lang CCV, Lauener R, Maintz L, Mantel PY, Maverakis E, Möhrenschlager M, Müller S, Nadeau K, Neumann AU, O'Mahony L, Rabenja FR, Renz H, Rhyner C, Rietschel E, Ring J, Roduit C, Sasaki M, Schenk M, Schröder J, Simon D, Simon HU, Sokolowska M, Ständer S, Steinhoff M, Piccirillo DS, Taïeb A, Takaoka R, Tapparo M, Teixeira H, Thyssen JP, Traidl S, Uhlmann M, van de Veen W, van Hage M, Virchow C, Wollenberg A, Yasutaka M, Zink A, Schmid-Grendelmeier P. Navigating the evolving landscape of atopic dermatitis: Challenges and future opportunities: The 4th Davos declaration. Allergy 2024; 79:2605-2624. [PMID: 39099205 DOI: 10.1111/all.16247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/18/2024] [Accepted: 07/04/2024] [Indexed: 08/06/2024]
Abstract
The 4th Davos Declaration was developed during the Global Allergy Forum in Davos which aimed to elevate the care of patients with atopic dermatitis (AD) by uniting experts and stakeholders. The forum addressed the high prevalence of AD, with a strategic focus on advancing research, treatment, and management to meet the evolving challenges in the field. This multidisciplinary forum brought together top leaders from research, clinical practice, policy, and patient advocacy to discuss the critical aspects of AD, including neuroimmunology, environmental factors, comorbidities, and breakthroughs in prevention, diagnosis, and treatment. The discussions were geared towards fostering a collaborative approach to integrate these advancements into practical, patient-centric care. The forum underlined the mounting burden of AD, attributing it to significant environmental and lifestyle changes. It acknowledged the progress in understanding AD and in developing targeted therapies but recognized a gap in translating these innovations into clinical practice. Emphasis was placed on the need for enhanced awareness, education, and stakeholder engagement to address this gap effectively and to consider environmental and lifestyle factors in a comprehensive disease management strategy. The 4th Davos Declaration marks a significant milestone in the journey to improve care for people with AD. By promoting a holistic approach that combines research, education, and clinical application, the Forum sets a roadmap for stakeholders to collaborate to improve patient outcomes in AD, reflecting a commitment to adapt and respond to the dynamic challenges of AD in a changing world.
Collapse
Affiliation(s)
- Claudia Traidl-Hoffmann
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Jamie Afghani
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Cezmi A Akdis
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Mübecel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | | | - Katja Bärenfaller
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Heidrun Behrendt
- Center for Allergy and Environment (ZAUM), Technische Universität München, Germany
| | - Thomas Bieber
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Davos Biosciences, Davos, Switzerland
| | | | | | - Charlotte Menné Bonefeld
- Department of Immunology and Microbiology, The LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Stefanie Bösch
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | - Marie Charlotte Brüggen
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | | | - Hans-Werner Duchna
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Hochgebirgsklinik Davos, Davos, Switzerland
| | | | - Danielle Fehr
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | | | - Remo Frei
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Bern University Hospital, Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Lena H Garvey
- Department of Dermatology and Allergy, Allergy Clinic, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Raschid Gharbo
- Psychosomatic Department, Hochgebirgsklinik, Davos, Switzerland
| | - Mehmet Gökkaya
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
| | - Karin Grando
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | - Carole Guillet
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
- Faculty of Medicine, University of Zürich, Zürich, Switzerland
| | | | | | - Nadine Herrmann
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Dirk Jan Hijnen
- Diakonessenhuis Utrecht Zeist Doorn Locatie Utrecht, Erasmus MC, University Medical Center Utrecht, Utrecht, Netherlands
| | - Claudia Hülpüsch
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Alan D Irvine
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Erika Jensen-Jarolim
- Center of Pathophysiology, Infectiology and Immunology, Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
- The interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Heidi H Kong
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Hillel Koren
- Environmental Health, LLC, Durham, North Carolina, USA
| | - Claudia C V Lang
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Immunology and Microbiology, The LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
| | - Roger Lauener
- Ostschweizer Kinderspital St. Gallen, St.Gallen, Switzerland
| | - Laura Maintz
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Pierre-Yves Mantel
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Emanuel Maverakis
- Department of Dermatology, University of California Davis, Sacramento, California, USA
| | | | - Svenja Müller
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Kari Nadeau
- Stanford University School of Medicine, Stanford, California, USA
| | - Avidan U Neumann
- Institute of Environmental Medicine and Integrative Health, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
| | - Liam O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine and School of Microbiology, University College Cork, Cork, Ireland
| | | | - Harald Renz
- Institute of Laboratory Medicine, Philipps University, Marburg, Germany
| | - Claudio Rhyner
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Ernst Rietschel
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Johannes Ring
- Klinik und Poliklinik für Dermatologie und Allergologie am Biederstein, Technische Universität München, Munich, Germany
| | - Caroline Roduit
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Bern University Hospital, Bern, Switzerland
- Ostschweizer Kinderspital St. Gallen, St.Gallen, Switzerland
| | - Mari Sasaki
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Bern University Hospital, Bern, Switzerland
| | - Mirjam Schenk
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Jens Schröder
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Schleswig-Holstein (UK-SH), Kiel, Germany
| | - Dagmar Simon
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Milena Sokolowska
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Sonja Ständer
- Center for Chronic Pruritus and Department of Dermatology, University Hospital Münster, Münster, Germany
| | - Martin Steinhoff
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- School of Medicine, Weill Cornell Medicine-Qatar, Ar-Rayyan, Qatar
- College of Medicine, Qatar University, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Department of Dermatology, Weill Cornell Medicine, New York, New York, USA
| | - Doris Straub Piccirillo
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Alain Taïeb
- INSERM 1312, University of Bordeaux, Bordeaux, France
| | - Roberto Takaoka
- Department of Dermatology, Faculdade de Medicina, Hospital das Clínicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | | | | | - Jacob Pontoppidan Thyssen
- Department of Dermatology and Venerology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Stephan Traidl
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Miriam Uhlmann
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Marianne van Hage
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institute and Karolinska University Hospital Stockholm, Solna, Sweden
| | - Christian Virchow
- Department of Pneumology, Intensive Care Medicine, Center for Internal Medicine, Universitätsmedizin Rostock, Rostock, Germany
| | - Andreas Wollenberg
- Department of Dermatology and Allergy, Ludwig-Maximilian-University, Munich, Germany
- Department of Dermatology and Allergy, University Hospital Augsburg, Augsburg, Germany
- Comprehensive Center of Inflammation Medicine, University Hospital Schleswig Holstein Campus Luebeck, Lubeck, Germany
| | - Mitamura Yasutaka
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Zurich, Switzerland
| | - Alexander Zink
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Medicine Solna, Division of Dermatology and Venereology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Schmid-Grendelmeier
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Medicine Campus, Davos, Switzerland
- Department of Immunology and Microbiology, The LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology, Allergy Unit, University Hospital of Zürich, Zürich, Switzerland
| |
Collapse
|
2
|
Stefanovic N, Irvine AD. Filaggrin and beyond: New insights into the skin barrier in atopic dermatitis and allergic diseases, from genetics to therapeutic perspectives. Ann Allergy Asthma Immunol 2024; 132:187-195. [PMID: 37758055 DOI: 10.1016/j.anai.2023.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/22/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023]
Abstract
Atopic dermatitis (AD) is the most common inflammatory skin disease worldwide, affecting 20% of children and 5% of adults. One critical component in the pathophysiology of AD is the epidermal skin barrier, with its outermost layer, the stratum corneum (SC), conferring biochemical properties that enable resilience against environmental threats and maintain homeostasis. The skin barrier may be conceptualized as a key facilitator of complex interactions between genetics, host immunity, the cutaneous microbiome, and environmental exposures. The key genetic risk factor for AD development and persistence is a loss-of-function mutation in FLG, with recent advances in genomics focusing on rare variant discovery, establishment of pathogenic mechanisms, and exploration of the role of other epidermal differentiation complex gene variants in AD. Aberrant type 2 inflammatory responses down-regulate the transcription of key epidermal barrier genes, alter the composition of SC lipids, and induce further injury through a neurocutaneous feedback loop and the itch-scratch cycle. The dysbiotic epidermis exhibits reduced bacterial diversity and enhanced colonization with Staphylococcus and Malassezia species, which contribute to both direct barrier injury through the action of bacterial toxins and perpetuation of the inflammatory cascades. Enhanced understanding of each of the pathogenic mechanisms underpinning barrier disruption has led to the development of novel topical and systemic molecules, including interleukin (IL)-4Ra, IL-13, PDE4, and Janus-associated kinase inhibitors, whose clinical effectiveness exceeds conventional treatment modalities. In this narrative review, we aim to summarize the current understanding of the above-mentioned pathophysiological and therapeutic mechanisms, with a focus on the genetic, cellular, and molecular mechanisms underpinning AD development.
Collapse
Affiliation(s)
| | - Alan D Irvine
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
3
|
Schröder JM. Discovery of natural bispecific antibodies: Is psoriasis induced by a toxigenic Corynebacterium simulans and maintained by CIDAMPs as autoantigens? Exp Dermatol 2024; 33:e15014. [PMID: 38284202 DOI: 10.1111/exd.15014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024]
Abstract
The high abundance of Corynebacterium simulans in psoriasis skin suggests a contribution to the psoriasis aetiology. This hypothesis was tested in an exploratory study, where western blot (WB) analyses with extracts of heat-treated C. simulans and psoriasis serum-derived IgG exhibited a single 16 kDa-WB-band. Proteomic analyses revealed ribosomal proteins as candidate C. s.-antigens. A peptidomic analysis unexpectedly showed that psoriasis serum-derived IgG already contained 31 immunopeptides of Corynebacteria ssp., suggesting the presence of natural bispecific antibodies (BsAbs). Moreover, peptidomic analyses gave 372 DECOY-peptides with similarity to virus- and phage proteins, including Corynebacterium diphtheriae phage, and similarity to diphtheria toxin. Strikingly, a peptidomic analysis for human peptides revealed 64 epitopes of major psoriasis autoantigens such as the spacer region of filaggrin, hornerin repeats and others. Most identified immunopeptides represent potential cationic intrinsically disordered antimicrobial peptides (CIDAMPs), which are generated within the epidermis. These may form complexes with bacterial disordered protein regions, representing chimeric antigens containing discontinuous epitopes. In addition, among 128 low-abundance immunopeptides, 48 are putatively psoriasis-relevant such as epitope peptides of PGE2-, vitamin D3- and IL-10-receptors. Further, 47 immunopeptides originated from tumour antigens, and the endogenous retrovirus HERV-K. I propose that persistent infection with a toxigenic C. simulans initiates psoriasis, which is exacerbated as an autoimmune disease by CIDAMPs as autoantigens. The discovery of natural BsAbs allows the identification of antigen epitopes from microbes, viruses, autoantigens and tumour-antigens, and may help to develop epitope-specific peptide-vaccines and therapeutic approaches with antigen-specific regulatory T cells to improve immune tolerance in an autoimmune disease-specific-manner.
Collapse
Affiliation(s)
- Jens-Michael Schröder
- Department of Dermatology, University-Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
4
|
Amar Y, Rogner D, Silva RL, Foesel BU, Ud-Dean M, Lagkouvardos I, Steimle-Grauer SA, Niedermeier S, Kublik S, Jargosch M, Heinig M, Thomas J, Eyerich S, Wikström JD, Schloter M, Eyerich K, Biedermann T, Köberle M. Darier's disease exhibits a unique cutaneous microbial dysbiosis associated with inflammation and body malodour. MICROBIOME 2023; 11:162. [PMID: 37496039 PMCID: PMC10369845 DOI: 10.1186/s40168-023-01587-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 06/01/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Darier's disease (DD) is a genodermatosis caused by mutations of the ATP2A2 gene leading to disrupted keratinocyte adhesion. Recurrent episodes of skin inflammation and infections with a typical malodour in DD indicate a role for microbial dysbiosis. Here, for the first time, we investigated the DD skin microbiome using a metabarcoding approach of 115 skin swabs from 14 patients and 14 healthy volunteers. Furthermore, we analyzed its changes in the context of DD malodour and the cutaneous DD transcriptome. RESULTS We identified a disease-specific cutaneous microbiome with a loss of microbial diversity and of potentially beneficial commensals. Expansion of inflammation-associated microbes such as Staphylococcus aureus and Staphylococcus warneri strongly correlated with disease severity. DD dysbiosis was further characterized by abundant species belonging to Corynebacteria, Staphylococci and Streptococci groups displaying strong associations with malodour intensity. Transcriptome analyses showed marked upregulation of epidermal repair, inflammatory and immune defence pathways reflecting epithelial and immune response mechanisms to DD dysbiotic microbiome. In contrast, barrier genes including claudin-4 and cadherin-4 were downregulated. CONCLUSIONS These findings allow a better understanding of Darier exacerbations, highlighting the role of cutaneous dysbiosis in DD inflammation and associated malodour. Our data also suggest potential biomarkers and targets of intervention for DD. Video Abstract.
Collapse
Affiliation(s)
- Yacine Amar
- Department of Dermatology and Allergy, Technical University of Munich, School of Medicine, Munich, Germany
| | - Danielle Rogner
- Department of Dermatology and Allergy, Technical University of Munich, School of Medicine, Munich, Germany
| | - Rafaela L Silva
- Department of Dermatology and Allergy, Technical University of Munich, School of Medicine, Munich, Germany
| | - Bärbel U Foesel
- Research Unit Comparative Microbiome Analysis, Helmholtz Zentrum München, Deutsches Forschungszentrum Für Gesundheit Und Umwelt (GmbH), 85764, Neuherberg, Germany
| | - Minhaz Ud-Dean
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Department of Informatics, Technical University of Munich, Garching, Germany
| | - Ilias Lagkouvardos
- Core Facility Microbiome, Technical University of Munich, 85354, Freising, Germany
| | - Susanne A Steimle-Grauer
- Department of Dermatology and Allergy, Technical University of Munich, School of Medicine, Munich, Germany
| | - Sebastian Niedermeier
- Department of Dermatology and Allergy, Technical University of Munich, School of Medicine, Munich, Germany
| | - Susanne Kublik
- Research Unit Comparative Microbiome Analysis, Helmholtz Zentrum München, Deutsches Forschungszentrum Für Gesundheit Und Umwelt (GmbH), 85764, Neuherberg, Germany
| | - Manja Jargosch
- Department of Dermatology and Allergy, Technical University of Munich, School of Medicine, Munich, Germany
| | - Matthias Heinig
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Department of Informatics, Technical University of Munich, Garching, Germany
| | - Jenny Thomas
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Zentrum München, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Stefanie Eyerich
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Zentrum München, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Jakob D Wikström
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Michael Schloter
- Research Unit Comparative Microbiome Analysis, Helmholtz Zentrum München, Deutsches Forschungszentrum Für Gesundheit Und Umwelt (GmbH), 85764, Neuherberg, Germany
| | - Kilian Eyerich
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Dermatology and Venereology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy, Technical University of Munich, School of Medicine, Munich, Germany.
| | - Martin Köberle
- Department of Dermatology and Allergy, Technical University of Munich, School of Medicine, Munich, Germany
| |
Collapse
|
5
|
Wertheim-Tysarowska K, Osipowicz K, Gielniewski B, Wojtaś B, Szabelska-Beręsewicz A, Zyprych-Walczak J, Mika A, Tysarowski A, Duk K, Rygiel AM, Niepokój K, Woźniak K, Kowalewski C, Wierzba J, Jezela-Stanek A. The Epidermal Transcriptome Analysis of a Novel c.639_642dup LORICRIN Variant-Delineation of the Loricrin Keratoderma Pathology. Int J Mol Sci 2023; 24:9459. [PMID: 37298411 PMCID: PMC10254037 DOI: 10.3390/ijms24119459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/15/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Loricrin keratoderma (LK) is a rare autosomal dominant genodermatosis caused by LORICRIN gene mutations. The pathogenesis of the disease is not yet fully understood. So far, only 10 pathogenic variants in LORICRIN have been described, with all of them but one being deletions or insertions. The significance of rare nonsense variants remains unclear. Furthermore, no data regarding the RNA expression in affected patients are available. The aim of this study is to describe the two variants in the LORICRIN gene found in two distinct families: the novel pathogenic variant c.639_642dup and a rare c.10C > T (p.Gln4Ter) of unknown significance. We also present the results of the transcriptome analysis of the lesional loricrin keratoderma epidermis of a patient with c.639_642dup. We show that in the LK lesion, the genes associated with epidermis development and keratocyte differentiation are upregulated, while genes engaged in cell adhesion, differentiation developmental processes, ion homeostasis and transport, signaling and cell communication are downregulated. In the context of the p.Gln4Ter clinical significance evaluation, we provide data indicating that LORICRIN haploinsufficiency has no skin consequences. Our results give further insight into the pathogenesis of LK, which may have therapeutic implications in the future and important significance in the context of genetic counseling.
Collapse
Affiliation(s)
| | - Katarzyna Osipowicz
- Department of Dermatology, Immunodermatology and Venereology, Medical University of Warsaw, 02-008 Warsaw, Poland
| | - Bartłomiej Gielniewski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Bartosz Wojtaś
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Alicja Szabelska-Beręsewicz
- Department of Mathematical and Statistical Methods, Poznań University of Life Sciences, 60-637 Poznań, Poland
| | - Joanna Zyprych-Walczak
- Department of Mathematical and Statistical Methods, Poznań University of Life Sciences, 60-637 Poznań, Poland
| | - Adriana Mika
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 80-211 Gdansk, Poland
| | - Andrzej Tysarowski
- Molecular and Translational Oncology Department and Cancer Molecular and Genetic Diagnostics Department, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Katarzyna Duk
- Department of Medical Genetics, Institute of Mother and Child, 01-211 Warsaw, Poland
| | | | - Katarzyna Niepokój
- Department of Medical Genetics, Institute of Mother and Child, 01-211 Warsaw, Poland
| | - Katarzyna Woźniak
- Department of Dermatology, Immunodermatology and Venereology, Medical University of Warsaw, 02-008 Warsaw, Poland
| | - Cezary Kowalewski
- Department of Dermatology, Immunodermatology and Venereology, Medical University of Warsaw, 02-008 Warsaw, Poland
| | - Jolanta Wierzba
- Department of Paediatrics, Haematology and Oncology, Department of General Nursery, Medical University of Gdansk, 80-211 Gdansk, Poland
| | - Aleksandra Jezela-Stanek
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 01-138 Warsaw, Poland
| |
Collapse
|
6
|
Jonca N, Simon M. The Cornified Envelope: A Versatile Contributor to the Epidermal Barrier. J Invest Dermatol 2023:S0022-202X(23)00101-X. [PMID: 37149811 DOI: 10.1016/j.jid.2023.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 05/08/2023]
Affiliation(s)
- Nathalie Jonca
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, French National Center for Scientific Research (CNRS), French National Institute of Health and Medical Research (INSERM), University Paul Sabatier, Toulouse, France; Department of Cell Biology and Cytology, Federative Institute of Biology, Purpan University Hospital, Toulouse, France
| | - Michel Simon
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, French National Center for Scientific Research (CNRS), French National Institute of Health and Medical Research (INSERM), University Paul Sabatier, Toulouse, France.
| |
Collapse
|
7
|
Jangir PK, Ogunlana L, Szili P, Czikkely M, Shaw LP, Stevens EJ, Yu Y, Yang Q, Wang Y, Pál C, Walsh TR, MacLean CR. The evolution of colistin resistance increases bacterial resistance to host antimicrobial peptides and virulence. eLife 2023; 12:e84395. [PMID: 37094804 PMCID: PMC10129329 DOI: 10.7554/elife.84395] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 04/05/2023] [Indexed: 04/26/2023] Open
Abstract
Antimicrobial peptides (AMPs) offer a promising solution to the antibiotic resistance crisis. However, an unresolved serious concern is that the evolution of resistance to therapeutic AMPs may generate cross-resistance to host AMPs, compromising a cornerstone of the innate immune response. We systematically tested this hypothesis using globally disseminated mobile colistin resistance (MCR) that has been selected by the use of colistin in agriculture and medicine. Here, we show that MCR provides a selective advantage to Escherichia coli in the presence of key AMPs from humans and agricultural animals by increasing AMP resistance. Moreover, MCR promotes bacterial growth in human serum and increases virulence in a Galleria mellonella infection model. Our study shows how the anthropogenic use of AMPs can drive the accidental evolution of resistance to the innate immune system of humans and animals. These findings have major implications for the design and use of therapeutic AMPs and suggest that MCR may be difficult to eradicate, even if colistin use is withdrawn.
Collapse
Affiliation(s)
- Pramod K Jangir
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Lois Ogunlana
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Petra Szili
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research NetworkSzegedHungary
- Doctoral School of Multidisciplinary Medical Sciences, University of SzegedSzegedHungary
| | - Marton Czikkely
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research NetworkSzegedHungary
| | - Liam P Shaw
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Emily J Stevens
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Yang Yu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural UniversityGuangzhouChina
| | - Qiue Yang
- Fujian Provincial Key Laboratory of Soil Environmental Health and RegulaWon, College of Resources and Environment, Fujian Agriculture and Forestry UniversityFuzhouChina
| | - Yang Wang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural UniversityBeijingChina
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research NetworkSzegedHungary
| | - Timothy R Walsh
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Craig R MacLean
- Department of Biology, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
8
|
CYSRT1: an antimicrobial epidermal protein that can interact with late cornified envelope (LCE) proteins. J Invest Dermatol 2023:S0022-202X(23)00085-4. [PMID: 36804407 DOI: 10.1016/j.jid.2023.01.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 02/17/2023]
Abstract
Late cornified envelope (LCE) proteins are small cationic epidermal proteins with antimicrobial properties, and the combined deletion of LCE3B and LCE3C genes is a risk factor for psoriasis that affects skin microbiome composition. In a yeast two-hybrid screen we identified cysteine-rich tail 1 protein (CYSRT1) as an interacting partner of members of all LCE groups except LCE6. These interactions were confirmed in a mammalian cell system by co-immunoprecipitation. CYSRT1 is a protein of unknown function that is specifically expressed in cutaneous and oral epithelia and spatially colocalizes with LCE proteins in the upper layers of the suprabasal epidermis. Constitutive CYSRT1 expression is present in fully differentiated epidermis and can be further induced in vivo by disruption of the skin barrier upon stratum corneum removal. Transcriptional regulation correlates to keratinocyte terminal differentiation but not to skin bacteria exposure. Similar to LCEs, CYSRT1 was found to have antibacterial activity against Pseudomonas aeruginosa. Comparative gene sequence analysis and protein amino acid alignment indicates that CYSRT1 is highly conserved among vertebrates and has putative antimicrobial activity. To summarize, we identified CYSRT1 in the outer skin layer, where it colocalizes with LCE proteins and contributes to the constitutive epidermal antimicrobial host defense repertoire.
Collapse
|
9
|
Choudhary MI, Römling U, Nadeem F, Bilal HM, Zafar M, Jahan H, ur-Rahman A. Innovative Strategies to Overcome Antimicrobial Resistance and Tolerance. Microorganisms 2022; 11:microorganisms11010016. [PMID: 36677308 PMCID: PMC9863313 DOI: 10.3390/microorganisms11010016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial resistance and tolerance are natural phenomena that arose due to evolutionary adaptation of microorganisms against various xenobiotic agents. These adaptation mechanisms make the current treatment options challenging as it is increasingly difficult to treat a broad range of infections, associated biofilm formation, intracellular and host adapted microbes, as well as persister cells and microbes in protected niches. Therefore, novel strategies are needed to identify the most promising drug targets to overcome the existing hurdles in the treatment of infectious diseases. Furthermore, discovery of novel drug candidates is also much needed, as few novel antimicrobial drugs have been introduced in the last two decades. In this review, we focus on the strategies that may help in the development of innovative small molecules which can interfere with microbial resistance mechanisms. We also highlight the recent advances in optimization of growth media which mimic host conditions and genome scale molecular analyses of microbial response against antimicrobial agents. Furthermore, we discuss the identification of antibiofilm molecules and their mechanisms of action in the light of the distinct physiology and metabolism of biofilm cells. This review thus provides the most recent advances in host mimicking growth media for effective drug discovery and development of antimicrobial and antibiofilm agents.
Collapse
Affiliation(s)
- M. Iqbal Choudhary
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Ute Römling
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 65 Stockholm, Sweden
- Correspondence: (U.R.); (H.J.); Tel.: +46-8-5248-7319 (U.R.); +92-21-111-232-292 (ext. 301) (H.J.)
| | - Faiza Nadeem
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Hafiz Muhammad Bilal
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Munirah Zafar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Humera Jahan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- Correspondence: (U.R.); (H.J.); Tel.: +46-8-5248-7319 (U.R.); +92-21-111-232-292 (ext. 301) (H.J.)
| | - Atta ur-Rahman
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
10
|
Niehues H, van der Krieken DA, Ederveen THA, Jansen PAM, van Niftrik L, Mesman R, Netea MG, Smits JPH, Schalkwijk J, van den Bogaard EH, Zeeuwen PLJM. Antimicrobial late cornified envelope (LCE) proteins: the psoriasis risk factor LCE3B/C-del affects microbiota composition. J Invest Dermatol 2021; 142:1947-1955.e6. [PMID: 34942199 DOI: 10.1016/j.jid.2021.11.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 11/03/2021] [Accepted: 11/22/2021] [Indexed: 12/20/2022]
Abstract
Late cornified envelope (LCE) proteins are predominantly expressed in the skin and other cornified epithelia. Based on sequence similarity, this eighteen-member homologous gene family has been subdivided into six groups. The LCE3 proteins have been the focus of dermatological research, as the combined deletion of LCE3B and LCE3C genes (LCE3B/C-del) is a risk factor for psoriasis. We previously reported that LCE3B/C-del increases expression of the LCE3A gene and that LCE3 proteins exert antibacterial activity. In the current study we analyzed the antimicrobial properties of other family members and the role of LCE3B/C-del in modulation of microbiota composition of the skin and oral cavity. Differences in killing efficiency and specificity between the LCE proteins and their target microbes were found, and the amino acid content, rather than the order, of the well-conserved central domain of the LCE3A protein was found responsible for its antibacterial activity. In vivo, LCE3B/C-del correlated with a higher beta-diversity in the skin and oral microbiota. From these results we conclude that all LCE proteins possess antimicrobial activity. Tissue-specific and genotype-dependent antimicrobial protein profiles impact skin and oral microbiota composition, which could direct towards LCE3B/C-del associated dysbiosis and a possible role for microbiota in the pathophysiology of psoriasis.
Collapse
Affiliation(s)
- Hanna Niehues
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Danique A van der Krieken
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Thomas H A Ederveen
- Center for Molecular and Biomolecular Informatics, RIMLS, Radboudumc, Nijmegen, The Netherlands
| | - Patrick A M Jansen
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Laura van Niftrik
- Department of Microbiology, Institute for Water and Wetland Research, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Rob Mesman
- Department of Microbiology, Institute for Water and Wetland Research, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, RIMLS, Radboudumc, Nijmegen, The Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Jos P H Smits
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Joost Schalkwijk
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Ellen H van den Bogaard
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Patrick L J M Zeeuwen
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen Medical Center (Radboudumc), Nijmegen, The Netherlands.
| |
Collapse
|
11
|
Christophers E, Schröder JM. Evolution of innate defense in human skin. Exp Dermatol 2021; 31:304-311. [PMID: 34694661 DOI: 10.1111/exd.14482] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 10/01/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022]
Abstract
More often as compared to other barrier systems (gastrointestinal, urogenital, and respiratory linings) human skin over millions of years has been subject to fundamental changes in structure and function. When life on land started, the first changes consisted in the formation of a coherent impermeable stratum corneum. Two-legged locomotion was followed by loss of body hair and formation of sweat glands. Major changes took place after the agricultural revolution, investigating settlements with domestication of animals and plants. Living together after giving up nomadic life, hairless skin became a battlefield for pathogens, members of the skin microbiome, and arthropod visits. Human skin became exceptional in showing a boosted, highly developed immune system which is much more complex as compared to the "skins" of other species. A recently found skin disinfection system ("Cationic Intrinsically Disordered Antimicrobial Peptides, CIDAMPs") dates back to the origins of life and still is active in present-day integuments. As a skin-restricted and effective principle, keratinocyte- myeloid synergy (KMS) is recognized. As a consequence of such highly developed immune defense, the basic contributions of KMS - cells (keratinocytes, neutrophils, macrophages) in regulating innate immunity is emphasized. Antimicrobial peptides and chemokines became major keratinocyte products. The formation of impermeable str. corneum membrane has enabled KMS - cells to accumulate within upper skin levels and cause a special group of human skin diseases, pustular dermatoses.
Collapse
Affiliation(s)
- Enno Christophers
- Department of Dermatology, University-Hospital Schleswig-Holstein, Kiel, Germany
| | | |
Collapse
|
12
|
Rademacher F, Gläser R, Harder J. Antimicrobial peptides and proteins: Interaction with the skin microbiota. Exp Dermatol 2021; 30:1496-1508. [PMID: 34310774 DOI: 10.1111/exd.14433] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/25/2021] [Accepted: 07/14/2021] [Indexed: 12/18/2022]
Abstract
The cutaneous microbiota comprises all living skin microorganisms. There is increasing evidence that the microbiota plays a crucial role in skin homeostasis. Accordingly, a dysbiosis of the microbiota may trigger cutaneous inflammation. The need for a balanced microbiota requires specific regulatory mechanisms that control and shape the microbiota. In this review, we highlight the present knowledge suggesting that antimicrobial peptides (AMPs) may exert a substantial influence on the microbiota by controlling their growth. This is supported by own data showing the differential influence of principal skin-derived AMPs on commensal staphylococci. Vice versa, we also illuminate how the cutaneous microbiota interacts with skin-derived AMPs by modulating AMP expression and how microbiota members protect themselves from the antimicrobial activity of AMPs. Taken together, the current picture suggests that a fine-tuned and well-balanced AMP-microbiota interplay on the skin surface may be crucial for skin health.
Collapse
Affiliation(s)
| | - Regine Gläser
- Department of Dermatology, Kiel University, Kiel, Germany
| | - Jürgen Harder
- Department of Dermatology, Kiel University, Kiel, Germany
| |
Collapse
|
13
|
Moretta A, Scieuzo C, Petrone AM, Salvia R, Manniello MD, Franco A, Lucchetti D, Vassallo A, Vogel H, Sgambato A, Falabella P. Antimicrobial Peptides: A New Hope in Biomedical and Pharmaceutical Fields. Front Cell Infect Microbiol 2021; 11:668632. [PMID: 34195099 PMCID: PMC8238046 DOI: 10.3389/fcimb.2021.668632] [Citation(s) in RCA: 207] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/10/2021] [Indexed: 12/14/2022] Open
Abstract
Antibiotics are essential drugs used to treat pathogenic bacteria, but their prolonged use contributes to the development and spread of drug-resistant microorganisms. Antibiotic resistance is a serious challenge and has led to the need for new alternative molecules less prone to bacterial resistance. Antimicrobial peptides (AMPs) have aroused great interest as potential next-generation antibiotics, since they are bioactive small proteins, naturally produced by all living organisms, and representing the first line of defense against fungi, viruses and bacteria. AMPs are commonly classified according to their sources, which are represented by microorganisms, plants and animals, as well as to their secondary structure, their biosynthesis and their mechanism of action. They find application in different fields such as agriculture, food industry and medicine, on which we focused our attention in this review. Particularly, we examined AMP potential applicability in wound healing, skin infections and metabolic syndrome, considering their ability to act as potential Angiotensin-Converting Enzyme I and pancreatic lipase inhibitory peptides as well as antioxidant peptides. Moreover, we argued about the pharmacokinetic and pharmacodynamic approaches to develop new antibiotics, the drug development strategies and the formulation approaches which need to be taken into account in developing clinically suitable AMP applications.
Collapse
Affiliation(s)
- Antonio Moretta
- Department of Sciences, University of Basilicata, Potenza, Italy
| | - Carmen Scieuzo
- Department of Sciences, University of Basilicata, Potenza, Italy
- Spinoff XFlies s.r.l, University of Basilicata, Potenza, Italy
| | | | - Rosanna Salvia
- Department of Sciences, University of Basilicata, Potenza, Italy
- Spinoff XFlies s.r.l, University of Basilicata, Potenza, Italy
| | | | - Antonio Franco
- Department of Sciences, University of Basilicata, Potenza, Italy
- Spinoff XFlies s.r.l, University of Basilicata, Potenza, Italy
| | - Donatella Lucchetti
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Vassallo
- Department of Sciences, University of Basilicata, Potenza, Italy
| | - Heiko Vogel
- Department of Entomology, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Alessandro Sgambato
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | - Patrizia Falabella
- Department of Sciences, University of Basilicata, Potenza, Italy
- Spinoff XFlies s.r.l, University of Basilicata, Potenza, Italy
| |
Collapse
|
14
|
Gene duplications and gene loss in the epidermal differentiation complex during the evolutionary land-to-water transition of cetaceans. Sci Rep 2021; 11:12334. [PMID: 34112911 PMCID: PMC8192740 DOI: 10.1038/s41598-021-91863-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/26/2021] [Indexed: 01/03/2023] Open
Abstract
Major protein components of the mammalian skin barrier are encoded by genes clustered in the Epidermal Differentiation Complex (EDC). The skin of cetaceans, i.e. whales, porpoises and dolphins, differs histologically from that of terrestrial mammals. However, the genetic regulation of their epidermal barrier is only incompletely known. Here, we investigated the EDC of cetaceans by comparative genomics. We found that important epidermal cornification proteins, such as loricrin and involucrin are conserved and subtypes of small proline-rich proteins (SPRRs) are even expanded in numbers in cetaceans. By contrast, keratinocyte proline rich protein (KPRP), skin-specific protein 32 (XP32) and late-cornified envelope (LCE) genes with the notable exception of LCE7A have been lost in cetaceans. Genes encoding proline rich 9 (PRR9) and late cornified envelope like proline rich 1 (LELP1) have degenerated in subgroups of cetaceans. These data suggest that the evolution of an aquatic lifestyle was accompanied by amplification of SPRR genes and loss of specific other epidermal differentiation genes in the phylogenetic lineage leading to cetaceans.
Collapse
|
15
|
Kumar A, Doan VM, Kunkli B, Csősz É. Construction of Unified Human Antimicrobial and Immunomodulatory Peptide Database and Examination of Antimicrobial and Immunomodulatory Peptides in Alzheimer's Disease Using Network Analysis of Proteomics Datasets. Front Genet 2021; 12:633050. [PMID: 33995478 PMCID: PMC8113759 DOI: 10.3389/fgene.2021.633050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/17/2021] [Indexed: 12/26/2022] Open
Abstract
The reanalysis of genomics and proteomics datasets by bioinformatics approaches is an appealing way to examine large amounts of reliable data. This can be especially true in cases such as Alzheimer's disease, where the access to biological samples, along with well-defined patient information can be challenging. Considering the inflammatory part of Alzheimer's disease, our aim was to examine the presence of antimicrobial and immunomodulatory peptides in human proteomic datasets deposited in the publicly available proteomics database ProteomeXchange (http://www.proteomexchange.org/). First, a unified, comprehensive human antimicrobial and immunomodulatory peptide database, containing all known human antimicrobial and immunomodulatory peptides was constructed and used along with the datasets containing high-quality proteomics data originating from the examination of Alzheimer's disease and control groups. A throughout network analysis was carried out, and the enriched GO functions were examined. Less than 1% of all identified proteins in the brain were antimicrobial and immunomodulatory peptides, but the alterations characteristic of Alzheimer's disease could be recapitulated with their analysis. Our data emphasize the key role of the innate immune system and blood clotting in the development of Alzheimer's disease. The central role of antimicrobial and immunomodulatory peptides suggests their utilization as potential targets for mechanistic studies and future therapies.
Collapse
Affiliation(s)
- Ajneesh Kumar
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Biomarker Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Vo Minh Doan
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Biomarker Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs Kunkli
- Biomarker Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Éva Csősz
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Biomarker Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
16
|
Grahl MVC, Lopes FC, Martinelli AHS, Carlini CR, Fruttero LL. Structure-Function Insights of Jaburetox and Soyuretox: Novel Intrinsically Disordered Polypeptides Derived from Plant Ureases. Molecules 2020; 25:molecules25225338. [PMID: 33207637 PMCID: PMC7696265 DOI: 10.3390/molecules25225338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/24/2022] Open
Abstract
Intrinsically disordered proteins (IDPs) and intrinsically disordered regions (IDRs) do not have a stable 3D structure but still have important biological activities. Jaburetox is a recombinant peptide derived from the jack bean (Canavalia ensiformis) urease and presents entomotoxic and antimicrobial actions. The structure of Jaburetox was elucidated using nuclear magnetic resonance which reveals it is an IDP with small amounts of secondary structure. Different approaches have demonstrated that Jaburetox acquires certain folding upon interaction with lipid membranes, a characteristic commonly found in other IDPs and usually important for their biological functions. Soyuretox, a recombinant peptide derived from the soybean (Glycine max) ubiquitous urease and homologous to Jaburetox, was also characterized for its biological activities and structural properties. Soyuretox is also an IDP, presenting more secondary structure in comparison with Jaburetox and similar entomotoxic and fungitoxic effects. Moreover, Soyuretox was found to be nontoxic to zebra fish, while Jaburetox was innocuous to mice and rats. This profile of toxicity affecting detrimental species without damaging mammals or the environment qualified them to be used in biotechnological applications. Both peptides were employed to develop transgenic crops and these plants were active against insects and nematodes, unveiling their immense potentiality for field applications.
Collapse
Affiliation(s)
- Matheus V. Coste Grahl
- Graduate Program in Medicine and Health Sciences, Brain Institute of Rio Grande do Sul (InsCer), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre CEP 90610-000, Brazil;
| | - Fernanda Cortez Lopes
- Graduate Program in Cellular and Molecular Biology, Center of Biotechnology, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves 9500, Building 43431, Porto Alegre CEP 91501-970, RS, Brazil;
| | - Anne H. Souza Martinelli
- Department of Biophysics & Deparment of Molecular Biology and Biotechnology-Biosciences Institute (IB), Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre 91501-970, RS, Brazil;
| | - Celia R. Carlini
- Graduate Program in Medicine and Health Sciences, Brain Institute of Rio Grande do Sul (InsCer), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre CEP 90610-000, Brazil;
- Brain Institute and School of Medicine, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil
- Correspondence: (C.R.C.); (L.L.F.); Tel.: +55-51-3320-3485 (C.R.C.); +54-351-535-3850 (L.L.F.)
| | - Leonardo L. Fruttero
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba CP 5000, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba CP 5000, Argentina
- Correspondence: (C.R.C.); (L.L.F.); Tel.: +55-51-3320-3485 (C.R.C.); +54-351-535-3850 (L.L.F.)
| |
Collapse
|
17
|
Prinz I, Sandrock I, Mrowietz U. Interleukin-17 cytokines: Effectors and targets in psoriasis-A breakthrough in understanding and treatment. J Exp Med 2020; 217:jem.20191397. [PMID: 31727784 PMCID: PMC7037256 DOI: 10.1084/jem.20191397] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/25/2019] [Accepted: 10/30/2019] [Indexed: 12/14/2022] Open
Abstract
This review summarizes the steps from basic research on IL-17 family cytokines to understanding their role in psoriasis pathogenesis to the approval of a number of monoclonal antibodies targeting IL-17 pathways as first line treatment of psoriasis and psoriatic arthritis. The IL-17 cytokine family comprising IL-17A to IL-17F and receptor subunits IL-17RA to IL-17RE represents a genetically ancient intercellular network regulating local tissue homeostasis. Its pivotal role in antifungal defense and its central position in the pathogenesis of inflammatory diseases including psoriasis were discovered only relatively late in the early 2000s. Since the connection of dysregulated IL-17 and psoriasis pathogenesis turned out to be particularly evident, a number of monoclonal antibodies targeting IL-17 pathways have been approved and are used as first line treatment of moderate-to-severe plaque psoriasis and psoriatic arthritis, and further agents are currently in clinical development.
Collapse
Affiliation(s)
- Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany.,Centre for Individualized Infection Medicine, Hannover, Germany.,Cluster of Excellence RESIST - Resolving Infection Susceptibility (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Inga Sandrock
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Ulrich Mrowietz
- Psoriasis Center at the Department of Dermatology and Comprehensive Center for Inflammation Medicine, University Medical Center Schleswig-Holstein, Campus Kiel, Germany
| |
Collapse
|
18
|
Smits JP, Ederveen TH, Rikken G, van den Brink NJ, van Vlijmen-Willems IM, Boekhorst J, Kamsteeg M, Schalkwijk J, van Hijum SA, Zeeuwen PL, van den Bogaard EH. Targeting the Cutaneous Microbiota in Atopic Dermatitis by Coal Tar via AHR-Dependent Induction of Antimicrobial Peptides. J Invest Dermatol 2020; 140:415-424.e10. [DOI: 10.1016/j.jid.2019.06.142] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 06/22/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022]
|
19
|
Oxidative Stress Alters Angiogenic and Antimicrobial Content of Extracellular Vesicles and Improves Flap Survival. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2019; 7:e2588. [PMID: 32537316 PMCID: PMC7288884 DOI: 10.1097/gox.0000000000002588] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/23/2019] [Indexed: 01/08/2023]
Abstract
Extracellular vesicles (EVs) secreted from adipose-derived mesenchymal stem cells (ADSCs) (ADSC-EVs) improve flap survival after ischemia-reperfusion injury. Exposure of parent ADSCs to oxidative stress has been shown to enhance this effect, but mechanisms are unclear. We aimed to determine whether angiogenesis-promoting protein and microRNA (miRNA) content is altered in EVs after preconditioning with hydrogen peroxide (H2O2 ADSC-EVs) and whether H2O2 ADSC-EVs can increase viability of random pattern skin flaps. Methods EVs secreted by human ADSCs were isolated after culture in EV-depleted medium ± H2O2. Nanoparticle tracking analysis determined size and concentration of purified EVs. Mass spectrometry and small RNA next-generation sequencing were performed to compare proteomic and miRNA profiles. ADSC-EVs, H2O2 ADSC-EVs, or vehicle were injected into random pattern skin flaps of BALB/c mice (4-5 mice per group). Viable and necrotic areas were measured on day 7, and tissues underwent histologic analysis. Results Angiogenic and antimicrobial protein content of EVs was altered with H2O2 preconditioning. Functional enrichment analysis identified constitutive photomorphogenesis 9 signalosome (known to direct vascular endothelial growth factor production) as the major enriched Gene Ontology term unique to H2O2 ADSC-EVs. Two miRNAs were increased, and 12 (including 10 antiangiogenic miRNAs) were reduced in H2O2 ADSC-EVs. Enhanced viability (P < 0.05) of flaps treated with H2O2 ADSC-EVs compared with vehicle corresponded to increased capillary density in the H2O2 group (P < 0.001). Conclusion Altered protein and miRNA content in ADSC-EVs after H2O2 pretreatment likely contributes to enhanced therapeutic effects on flap survival observed in preclinical models.
Collapse
|
20
|
Kintses B, Jangir PK, Fekete G, Számel M, Méhi O, Spohn R, Daruka L, Martins A, Hosseinnia A, Gagarinova A, Kim S, Phanse S, Csörgő B, Györkei Á, Ari E, Lázár V, Nagy I, Babu M, Pál C, Papp B. Chemical-genetic profiling reveals limited cross-resistance between antimicrobial peptides with different modes of action. Nat Commun 2019; 10:5731. [PMID: 31844052 PMCID: PMC6915728 DOI: 10.1038/s41467-019-13618-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/14/2019] [Indexed: 11/09/2022] Open
Abstract
Antimicrobial peptides (AMPs) are key effectors of the innate immune system and promising therapeutic agents. Yet, knowledge on how to design AMPs with minimal cross-resistance to human host-defense peptides remains limited. Here, we systematically assess the resistance determinants of Escherichia coli against 15 different AMPs using chemical-genetics and compare to the cross-resistance spectra of laboratory-evolved AMP-resistant strains. Although generalizations about AMP resistance are common in the literature, we find that AMPs with different physicochemical properties and cellular targets vary considerably in their resistance determinants. As a consequence, cross-resistance is prevalent only between AMPs with similar modes of action. Finally, our screen reveals several genes that shape susceptibility to membrane- and intracellular-targeting AMPs in an antagonistic manner. We anticipate that chemical-genetic approaches could inform future efforts to minimize cross-resistance between therapeutic and human host AMPs.
Collapse
Affiliation(s)
- Bálint Kintses
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary.
- HCEMM-BRC Translational Microbiology Lab, Szeged, Hungary.
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary.
| | - Pramod K Jangir
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Gergely Fekete
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
| | - Mónika Számel
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Orsolya Méhi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Réka Spohn
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Lejla Daruka
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ana Martins
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Ali Hosseinnia
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Alla Gagarinova
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Sunyoung Kim
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Sadhna Phanse
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Bálint Csörgő
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Department of Microbiology and Immunology, University of California, San Francisco, USA
| | - Ádám Györkei
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
| | - Eszter Ari
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Viktória Lázár
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - István Nagy
- Sequencing Platform, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Mohan Babu
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary.
| | - Balázs Papp
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary.
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary.
| |
Collapse
|
21
|
Schröder JM. Seeing Is Believing: Vitamin A Promotes Skin Health through a Host-Derived Antibiotic. Cell Host Microbe 2019; 25:769-770. [PMID: 31194936 DOI: 10.1016/j.chom.2019.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In this issue of Cell Host & Microbe, Harris et al. (2019) show that members of the resistin-like molecule (RELM) family are bactericidal, antimicrobial peptides that promote skin defenses in a vitamin-A-dependent manner. Administration of vitamin A analogs triggers RELM, which improves host resistance against skin pathogens in mice.
Collapse
Affiliation(s)
- Jens-M Schröder
- Department of Dermatology, University-Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
| |
Collapse
|
22
|
Latendorf T, Gerstel U, Wu Z, Bartels J, Becker A, Tholey A, Schröder JM. Cationic Intrinsically Disordered Antimicrobial Peptides (CIDAMPs) Represent a New Paradigm of Innate Defense with a Potential for Novel Anti-Infectives. Sci Rep 2019; 9:3331. [PMID: 30833614 PMCID: PMC6399351 DOI: 10.1038/s41598-019-39219-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/19/2018] [Indexed: 12/22/2022] Open
Abstract
In the search for potential mechanisms underlying the remarkable resistance of healthy skin against infection by soil bacteria like Pseudomonas (P.) aeruginosa we identified fragments of the intrinsically disordered protein hornerin as potent microbicidal agents in the stratum corneum. We found that, independent of the amino acid (AA)-sequence, any tested linear cationic peptide containing a high percentage of disorder-promoting AA and a low percentage of order-promoting AA is a potent microbicidal antimicrobial. We further show that the antimicrobial activity of these cationic intrinsically disordered antimicrobial peptides (CIDAMPs) depends on the peptide chain length, its net charge, lipidation and environmental conditions. The ubiquitous presence of latent CIDAMP sources in nature suggests a common and yet overlooked adapted innate disinfection system of body surfaces. The simple structure and virtually any imaginable sequence or composition of disorder-promoting AA allow the generation of a plethora of CIDAMPs. These are potential novel microbicidal anti-infectives for various bacterial pathogens, including P. aeruginosa, methicillin-resistant Staphylococcus aureus (MRSA) and fungal pathogens like Candida albicans and Cryptococcus neoformans.
Collapse
Affiliation(s)
- Ties Latendorf
- Department of Dermatology, University-Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ulrich Gerstel
- Department of Dermatology, University-Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Zhihong Wu
- Department of Dermatology, University-Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
- Institute of Biochemistry and Cell Biology, Zhejiang University of Science and Technology, 310023, Hangzhou, China
| | - Joachim Bartels
- Department of Dermatology, University-Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Alexander Becker
- Institute for Experimental Medicine-AG Systematic Proteomics & Bioanalytics, Kiel University (CAU), Kiel, Germany
| | - Andreas Tholey
- Institute for Experimental Medicine-AG Systematic Proteomics & Bioanalytics, Kiel University (CAU), Kiel, Germany
| | - Jens-Michael Schröder
- Department of Dermatology, University-Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
| |
Collapse
|