1
|
Zhu R, Yao X, Li W. Langerhans cells and skin immune diseases. Eur J Immunol 2024; 54:e2250280. [PMID: 39030782 DOI: 10.1002/eji.202250280] [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: 04/07/2023] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/22/2024]
Abstract
Langerhans cells (LCs) are the key antigen-presenting cells in the epidermis in normal conditions and respond differentially to environmental and/or endogenous stimuli, exerting either proinflammatory or anti-inflammatory effects. Current knowledge about LCs mainly originates from studies utilizing mouse models, whereas with the development of single-cell techniques, there has been significant progress for human LCs, which has updated our understanding of the phenotype, ontogeny, differentiation regulation, and function of LCs. In this review, we delineated the progress of human LCs and summarized LCs' function in inflammatory skin diseases, providing new ideas for precise regulation of LC function in the prevention and treatment of skin diseases.
Collapse
Affiliation(s)
- Ronghui Zhu
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, P. R. China
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Hubei Province & Key Laboratory of Skin Infection and Immunity, Wuhan, P. R. China
| | - Xu Yao
- Department, of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, P. R. China
| | - Wei Li
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, P. R. China
| |
Collapse
|
2
|
Seiser S, Arzani H, Ayub T, Phan-Canh T, Staud C, Worda C, Kuchler K, Elbe-Bürger A. Native human and mouse skin infection models to study Candida auris-host interactions. Microbes Infect 2024; 26:105234. [PMID: 37813159 DOI: 10.1016/j.micinf.2023.105234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/29/2023] [Accepted: 09/30/2023] [Indexed: 10/11/2023]
Abstract
The World Health Organization (WHO) declared certain fungal pathogens as global health threats for the next decade. Candida auris (C. auris) is a newly emerging skin-tropic multidrug-resistant fungal pathogen that can cause life-threatening infections of high mortality in hospitals and healthcare settings. Here, we address an unmet need and present novel native ex vivo skin models, thus extending previous C. auris-host interaction studies. We exploit histology and immunofluorescence analysis of ex vivo skin biopsies of human adult and fetal, as well as mouse origin infected with C. auris via distinct routes. We demonstrate that an intact skin barrier efficiently protects from C. auris penetration and invasion. Although C. auris readily grows on native human skin, it can reach deeper layers only upon physical disruption of the barrier by needling or through otherwise damaged skin. By contrast, a barrier disruption is not necessary for C. auris penetration of native mouse skin. Importantly, we show that C. auris undergoes morphogenetic changes upon skin penetration, as it acquires pseudohyphal growth phenotypes in deeper human and mouse dermis. Taken together, this new human and mouse skin model toolset yields new insights into C. auris colonization, adhesion, growth and invasion properties of native versus damaged human skin. The results form a crucial basis for future studies on skin immune defense to colonizing pathogens, and offer new options for testing the action and efficacy of topical antimicrobial compound formulations.
Collapse
Affiliation(s)
- Saskia Seiser
- Medical University of Vienna, Department of Dermatology, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Hossein Arzani
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria
| | - Tanya Ayub
- Medical University of Vienna, Department of Dermatology, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Trinh Phan-Canh
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria
| | - Clement Staud
- Medical University of Vienna, Department of Plastic and Reconstructive Surgery, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Christof Worda
- Medical University of Vienna, Department of Obstetrics and Gynecology, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Karl Kuchler
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna Biocenter, Dr. Bohr-Gasse 9/2, 1030 Vienna, Austria.
| | - Adelheid Elbe-Bürger
- Medical University of Vienna, Department of Dermatology, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
3
|
Trompette A, Ubags ND. Skin barrier immunology from early life to adulthood. Mucosal Immunol 2023; 16:194-207. [PMID: 36868478 DOI: 10.1016/j.mucimm.2023.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 03/05/2023]
Abstract
Our skin has a unique barrier function, which is imperative for the body's protection against external pathogens and environmental insults. Although interacting closely and sharing many similarities with key mucosal barrier sites, such as the gut and the lung, the skin also provides protection for internal tissues and organs and has a distinct lipid and chemical composition. Skin immunity develops over time and is influenced by a multiplicity of different factors, including lifestyle, genetics, and environmental exposures. Alterations in early life skin immune and structural development may have long-term consequences for skin health. In this review, we summarize the current knowledge on cutaneous barrier and immune development from early life to adulthood, with an overview of skin physiology and immune responses. We specifically highlight the influence of the skin microenvironment and other host intrinsic, host extrinsic (e.g. skin microbiome), and environmental factors on early life cutaneous immunity.
Collapse
Affiliation(s)
- Aurélien Trompette
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Niki D Ubags
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.
| |
Collapse
|
4
|
Visscher MO, Carr AN, Narendran V. Epidermal Immunity and Function: Origin in Neonatal Skin. Front Mol Biosci 2022; 9:894496. [PMID: 35755808 PMCID: PMC9215705 DOI: 10.3389/fmolb.2022.894496] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/05/2022] [Indexed: 11/22/2022] Open
Abstract
The fascinating story of epidermal immunity begins in utero where the epidermal barrier derives from the ectoderm and evolves through carefully orchestrated biological processes, including periderm formation, keratinocyte differentiation, proliferation, cornification, and maturation, to generate a functional epidermis. Vernix caseosa derives from epidermal cells that mix with sebaceous lipids and coat the fetus during late gestation, likely to provide conditions for cornification. At birth, infants dramatically transition from aqueous conditions to a dry gaseous environment. The epidermal barrier begins to change within hours, exhibiting decreased hydration and low stratum corneum (SC) cohesion. The SC varied by gestational age (GA), transformed over the next 2–3 months, and differed considerably versus stable adult skin, as indicated by analysis of specific protein biomarkers. Regardless of gestational age, the increased infant SC proteins at 2–3 months after birth were involved in late differentiation, cornification, and filaggrin processing compared to adult skin. Additionally, the natural moisturizing factor (NMF), the product of filaggrin processing, was higher for infants than adults. This suggests that neonatal skin provides innate immunity and protection from environmental effects and promotes rapid, continued barrier development after birth. Functional genomic analysis showed abundant differences across biological processes for infant skin compared to adult skin. Gene expression for extracellular matrix, development, and fatty acid metabolism was higher for infant skin, while adult skin had increased expression of genes for the maintenance of epidermal homeostasis, antigen processing/presentation of immune function, and others. These findings provide descriptive information about infant epidermal immunity and its ability to support the newborn’s survival and growth, despite an environment laden with microbes, high oxygen tension, and irritants.
Collapse
Affiliation(s)
- Marty O Visscher
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| | - Andrew N Carr
- The Procter and Gamble Company, Cincinnati, OH, United States
| | - Vivek Narendran
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
5
|
Reitermaier R, Ayub T, Staller J, Kienzl P, Fortelny N, Vieyra-Garcia PA, Worda C, Fiala C, Staud C, Eppel W, Scharrer A, Krausgruber T, Elbe-Bürger A. The molecular and phenotypic makeup of fetal human skin T lymphocytes. Development 2022; 149:dev199781. [PMID: 34604909 PMCID: PMC8601710 DOI: 10.1242/dev.199781] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022]
Abstract
The adult human skin contains a vast number of T cells that are essential for skin homeostasis and pathogen defense. T cells are first observed in the skin at the early stages of gestation; however, our understanding of their contribution to early immunity has been limited by their low abundance and lack of comprehensive methodologies for their assessment. Here, we describe a new workflow for isolating and expanding significant amounts of T cells from fetal human skin. Using multiparametric flow cytometry and in situ immunofluorescence, we found a large population with a naive phenotype and small populations with a memory and regulatory phenotype. Their molecular state was characterized using single-cell transcriptomics and TCR repertoire profiling. Importantly, culture of total fetal skin biopsies facilitated T cell expansion without a substantial impact on their phenotype, a major prerequisite for subsequent functional assays. Collectively, our experimental approaches and data advance the understanding of fetal skin immunity and potential use in future therapeutic interventions.
Collapse
Affiliation(s)
- René Reitermaier
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Tanya Ayub
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Julia Staller
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Philip Kienzl
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Nikolaus Fortelny
- Department of Biosciences, University of Salzburg, Salzburg 5020, Austria
| | | | - Christof Worda
- Department of Obstetrics & Gynecology, Medical University of Vienna, Vienna 1090, Austria
| | - Christian Fiala
- Gynmed Clinic, Vienna 1150, Austria
- Department of Women's and Children's Health, Division of Obstetrics and Gynaecology, Karolinska Institute and Karolinska University Hospital, Stockholm 171 77, Sweden
| | - Clement Staud
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna 1090, Austria
| | - Wolfgang Eppel
- Department of Obstetrics & Gynecology, Medical University of Vienna, Vienna 1090, Austria
| | - Anke Scharrer
- Department of Pathology, Medical University of Vienna, Vienna 1090, Austria
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | |
Collapse
|
6
|
Naik S. One Size Does Not Fit All: Diversifying Immune Function in the Skin. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:227-234. [PMID: 35017212 PMCID: PMC8820520 DOI: 10.4049/jimmunol.2100758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/11/2021] [Indexed: 01/17/2023]
Abstract
Our body's most outward facing epithelial barrier, the skin, serves as the frontline defense against myriad environmental assailants. To combat these motley threats, the skin has evolved a sophisticated immunological arsenal. In this article, I provide an overview of the skin's complex architecture and the distinct microniches in which immune cells reside and function. I review burgeoning literature on the synchronized immune, stromal, epithelial, and neuronal cell responses in healthy and inflamed skin. Next, I delve into the distinct requirement and mechanisms of long-term immune surveillance and tissue adaptation at the cutaneous frontier. Finally, by discussing the contributions of immune cells in maintaining and restoring tissue integrity, I underscore the constellation of noncanonical functions undertaken by the skin immune system. Just as our skin's immune system benefits from embracing diverse defense strategies, so, too, must we in the immunology research community support disparate perspectives and people from all walks of life.
Collapse
Affiliation(s)
- Shruti Naik
- Department of Pathology, Department of Medicine, Ronald O. Perelman Department of Dermatology, and Perlmutter Cancer Center, New York University Langone Health, 550 First Avenue, New York, New York. 10016 USA,Correspondence to:
| |
Collapse
|
7
|
Xu Y, Zhang J, Hu Y, Li X, Sun L, Peng Y, Sun Y, Liu B, Bian Z, Rong Z. Single-cell transcriptome analysis reveals the dynamics of human immune cells during early fetal skin development. Cell Rep 2021; 36:109524. [PMID: 34380039 DOI: 10.1016/j.celrep.2021.109524] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 05/12/2021] [Accepted: 07/22/2021] [Indexed: 01/12/2023] Open
Abstract
The immune system of skin develops in stages in mice. However, the developmental dynamics of immune cells in human skin remains elusive. Here, we perform transcriptome profiling of CD45+ hematopoietic cells in human fetal skin at an estimated gestational age of 10-17 weeks by single-cell RNA sequencing. A total of 13 immune cell types are identified. Skin macrophages show dynamic heterogeneity over the course of skin development. A major shift in lymphoid cell developmental states occurs from the first to the second trimester that implies an in situ differentiation process. Gene expression analysis reveals a typical developmental program in immune cells in accordance with their functional maturation, possibly involving metabolic reprogramming. Finally, we identify transcription factors (TFs) that potentially regulate cellular transitions by comparing TFs and TF target gene networks. These findings provide detailed insight into how the immune system of the human skin is established during development.
Collapse
Affiliation(s)
- Yingping Xu
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China.
| | - Jun Zhang
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Yongfei Hu
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Xuefei Li
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Lihua Sun
- Development of Gynaecology and Obstetrics, Nanhai Hospital, Southern Medical University, Guangzhou 528200, China
| | - Yu Peng
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Yuzhe Sun
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China
| | - Bing Liu
- State Key Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Zhilei Bian
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
| | - Zhili Rong
- Experimental Research Center, Dermatology Hospital, Southern Medical University, Guangzhou 510091, China; Cancer Research Institute, School of Basic Medical Sciences, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Key Laboratory of Organ Failure Research (Ministry of Education), Southern Medical University, Guangzhou 510515, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China.
| |
Collapse
|
8
|
Mishra A, Lai GC, Yao LJ, Aung TT, Shental N, Rotter-Maskowitz A, Shepherdson E, Singh GSN, Pai R, Shanti A, Wong RMM, Lee A, Khyriem C, Dutertre CA, Chakarov S, Srinivasan KG, Shadan NB, Zhang XM, Khalilnezhad S, Cottier F, Tan ASM, Low G, Chen P, Fan Y, Hor PX, Lee AKM, Choolani M, Vermijlen D, Sharma A, Fuks G, Straussman R, Pavelka N, Malleret B, McGovern N, Albani S, Chan JKY, Ginhoux F. Microbial exposure during early human development primes fetal immune cells. Cell 2021; 184:3394-3409.e20. [PMID: 34077752 PMCID: PMC8240556 DOI: 10.1016/j.cell.2021.04.039] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 02/09/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
The human fetal immune system begins to develop early during gestation; however, factors responsible for fetal immune-priming remain elusive. We explored potential exposure to microbial agents in utero and their contribution toward activation of memory T cells in fetal tissues. We profiled microbes across fetal organs using 16S rRNA gene sequencing and detected low but consistent microbial signal in fetal gut, skin, placenta, and lungs in the 2nd trimester of gestation. We identified several live bacterial strains including Staphylococcus and Lactobacillus in fetal tissues, which induced in vitro activation of memory T cells in fetal mesenteric lymph node, supporting the role of microbial exposure in fetal immune-priming. Finally, using SEM and RNA-ISH, we visualized discrete localization of bacteria-like structures and eubacterial-RNA within 14th weeks fetal gut lumen. These findings indicate selective presence of live microbes in fetal organs during the 2nd trimester of gestation and have broader implications toward the establishment of immune competency and priming before birth.
Collapse
Affiliation(s)
- Archita Mishra
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Ghee Chuan Lai
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Leong Jing Yao
- Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the Academia, 20 College Road, Discovery Tower Level 8, Singapore 169856, Singapore
| | - Thet Tun Aung
- Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117597, Singapore
| | - Noam Shental
- Department of Mathematics and Computer Science, Open University of Israel, Ra'anana 4353701, Israel
| | - Aviva Rotter-Maskowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Edwin Shepherdson
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Gurmit Singh Naranjan Singh
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Rhea Pai
- Genome Institute of Singapore (GIS), A(∗)STAR, 60 Biopolis Street, Singapore 138672, Singapore
| | - Adhika Shanti
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Regina Men Men Wong
- Genome Institute of Singapore (GIS), A(∗)STAR, 60 Biopolis Street, Singapore 138672, Singapore
| | - Andrea Lee
- Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the Academia, 20 College Road, Discovery Tower Level 8, Singapore 169856, Singapore
| | - Costerwell Khyriem
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Charles Antoine Dutertre
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore; Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the Academia, 20 College Road, Discovery Tower Level 8, Singapore 169856, Singapore; Program in Emerging Infectious Disease, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Svetoslav Chakarov
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - K G Srinivasan
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Nurhidaya Binte Shadan
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Xiao-Meng Zhang
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Shabnam Khalilnezhad
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Fabien Cottier
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Alrina Shin Min Tan
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Gillian Low
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Phyllis Chen
- Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the Academia, 20 College Road, Discovery Tower Level 8, Singapore 169856, Singapore
| | - Yiping Fan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore; Experimental Fetal Medicine Group, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Pei Xiang Hor
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Avery Khoo May Lee
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Mahesh Choolani
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, 1E Kent Ridge Road, Singapore 119228, Singpore
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Institute for Medical Immunology, ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels 1050, Belgium
| | - Ankur Sharma
- Genome Institute of Singapore (GIS), A(∗)STAR, 60 Biopolis Street, Singapore 138672, Singapore; Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, the University of Western Australia, PO Box 7214, 6 Verdun Street, Nedlands, Perth, WA 6009, Australia; Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Garold Fuks
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ravid Straussman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Norman Pavelka
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Benoit Malleret
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore; Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117597, Singapore
| | - Naomi McGovern
- Department of Pathology and Centre for Trophoblast Research, Tennis Court Road, Cambridge CB2 1QP, UK.
| | - Salvatore Albani
- Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the Academia, 20 College Road, Discovery Tower Level 8, Singapore 169856, Singapore.
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore; Experimental Fetal Medicine Group, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; OBGYN-Academic Clinical Program, Duke-NUS, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore; Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore 119077, Singapore.
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore; Translational Immunology Institute, Singhealth/Duke-NUS Academic Medical Centre, the Academia, 20 College Road, Discovery Tower Level 8, Singapore 169856, Singapore; Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
9
|
Miah M, Goh I, Haniffa M. Prenatal Development and Function of Human Mononuclear Phagocytes. Front Cell Dev Biol 2021; 9:649937. [PMID: 33898444 PMCID: PMC8060508 DOI: 10.3389/fcell.2021.649937] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
The human mononuclear phagocyte (MP) system, which includes dendritic cells, monocytes, and macrophages, is a critical regulator of innate and adaptive immune responses. During embryonic development, MPs derive sequentially in yolk sac progenitors, fetal liver, and bone marrow haematopoietic stem cells. MPs maintain tissue homeostasis and confer protective immunity in post-natal life. Recent evidence - primarily in animal models - highlight their critical role in coordinating the remodeling, maturation, and repair of target organs during embryonic and fetal development. However, the molecular regulation governing chemotaxis, homeostasis, and functional diversification of resident MP cells in their respective organ systems during development remains elusive. In this review, we summarize the current understanding of the development and functional contribution of tissue MPs during human organ development and morphogenesis and its relevance to regenerative medicine. We outline how single-cell multi-omic approaches and next-generation ex-vivo organ-on-chip models provide new experimental platforms to study the role of human MPs during development and disease.
Collapse
Affiliation(s)
- Mohi Miah
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Issac Goh
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.,Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.,Wellcome Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
10
|
Stephenson E, Webb S, Haniffa M. Multiomics uncovers developing immunological lineages in human. Eur J Immunol 2021; 51:764-772. [PMID: 33569778 PMCID: PMC8600952 DOI: 10.1002/eji.202048769] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/19/2020] [Indexed: 11/06/2022]
Abstract
The development of the human immune system during embryonic and fetal life has historically been difficult to research due to limited access to human tissue. Experimental animal models have been widely used to study development but cellular and molecular programmes may not be conserved across species. The advent of multiomic single-cell technologies and an increase in human developmental tissue biobank resources have facilitated single-cell multiomic studies focused on human immune development. A critical question in the near future is "How do we best reconcile scientific findings across multiple omic modalities, developmental time, and organismic space?" In this review, we discuss the application of single-cell multiomic technologies to unravel the major cellular lineages in the prenatal human immune system. We also identify key areas where the combined power of multiomics technologies can be leveraged to address specific immunological gaps in our current knowledge and explore new research horizons in human development.
Collapse
Affiliation(s)
- Emily Stephenson
- Biosciences InstituteNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | - Simone Webb
- Biosciences InstituteNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | - Muzlifah Haniffa
- Biosciences InstituteNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
- Department of Dermatology and NIHR Newcastle Biomedical Research CentreNewcastle Hospitals NHS Foundation TrustNewcastle Upon TyneUK
| |
Collapse
|
11
|
Dhariwala MO, Karthikeyan D, Vasquez KS, Farhat S, Weckel A, Taravati K, Leitner EG, Clancy S, Pauli M, Piper ML, Cohen JN, Ashouri JF, Lowe MM, Rosenblum MD, Scharschmidt TC. Developing Human Skin Contains Lymphocytes Demonstrating a Memory Signature. CELL REPORTS MEDICINE 2020; 1:100132. [PMID: 33294857 PMCID: PMC7691438 DOI: 10.1016/j.xcrm.2020.100132] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/20/2020] [Accepted: 10/13/2020] [Indexed: 12/17/2022]
Abstract
Lymphocytes in barrier tissues play critical roles in host defense and homeostasis. These cells take up residence in tissues during defined developmental windows, when they may demonstrate distinct phenotypes and functions. Here, we utilized mass and flow cytometry to elucidate early features of human skin immunity. Although most conventional αβ T (Tconv) cells in fetal skin have a naive, proliferative phenotype, a subset of CD4+ Tconv and CD8+ cells demonstrate memory-like features and a propensity for interferon (IFN)γ production. Skin regulatory T cells dynamically accumulate over the second trimester in temporal and regional association with hair follicle development. These fetal skin regulatory T cells (Tregs) demonstrate an effector memory phenotype while differing from their adult counterparts in expression of key effector molecules. Thus, we identify features of prenatal skin lymphocytes that may have key implications for understanding antigen and allergen encounters in utero and in infancy. CyTOF reveals a complex lymphocyte landscape in developing human skin Developing skin contains CD45RO+ conventional T cells with propensity to produce IFNγ Regulatory T cells (Tregs) in skin before birth display effector memory properties Skin Tregs increase in conjunction with initial hair follicle morphogenesis
Collapse
Affiliation(s)
- Miqdad O Dhariwala
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dhuvarakesh Karthikeyan
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kimberly S Vasquez
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sepideh Farhat
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Antonin Weckel
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Keyon Taravati
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Elizabeth G Leitner
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sean Clancy
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mariela Pauli
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Merisa L Piper
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jarish N Cohen
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA.,Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Judith F Ashouri
- Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Margaret M Lowe
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael D Rosenblum
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tiffany C Scharschmidt
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
12
|
Ferrer IR, West HC, Henderson S, Ushakov DS, Santos E Sousa P, Strid J, Chakraverty R, Yates AJ, Bennett CL. A wave of monocytes is recruited to replenish the long-term Langerhans cell network after immune injury. Sci Immunol 2020; 4:4/38/eaax8704. [PMID: 31444235 DOI: 10.1126/sciimmunol.aax8704] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/15/2019] [Indexed: 12/21/2022]
Abstract
A dense population of embryo-derived Langerhans cells (eLCs) is maintained within the sealed epidermis without contribution from circulating cells. When this network is perturbed by transient exposure to ultraviolet light, short-term LCs are temporarily reconstituted from an initial wave of monocytes but thought to be superseded by more permanent repopulation with undefined LC precursors. However, the extent to which this process is relevant to immunopathological processes that damage LC population integrity is not known. Using a model of allogeneic hematopoietic stem cell transplantation, where alloreactive T cells directly target eLCs, we have asked whether and how the original LC network is ultimately restored. We find that donor monocytes, but not dendritic cells, are the precursors of long-term LCs in this context. Destruction of eLCs leads to recruitment of a wave of monocytes that engraft in the epidermis and undergo a sequential pathway of differentiation via transcriptionally distinct EpCAM+ precursors. Monocyte-derived LCs acquire the capacity of self-renewal, and proliferation in the epidermis matched that of steady-state eLCs. However, we identified a bottleneck in the differentiation and survival of epidermal monocytes, which, together with the slow rate of renewal of mature LCs, limits repair of the network. Furthermore, replenishment of the LC network leads to constitutive entry of cells into the epidermal compartment. Thus, immune injury triggers functional adaptation of mechanisms used to maintain tissue-resident macrophages at other sites, but this process is highly inefficient in the skin.
Collapse
Affiliation(s)
- Ivana R Ferrer
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK and Cancer Institute Department of Haematology, Division of Cancer Studies, University College London, London WC1E 6DD, UK
| | - Heather C West
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK and Cancer Institute Department of Haematology, Division of Cancer Studies, University College London, London WC1E 6DD, UK
| | - Stephen Henderson
- Bill Lyons Informatics Centre, Cancer Institute, University College London, London WC1E 6DD, UK
| | - Dmitry S Ushakov
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, New Hunt's House, Newcomen Street, London SE1 1UL, UK
| | - Pedro Santos E Sousa
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK and Cancer Institute Department of Haematology, Division of Cancer Studies, University College London, London WC1E 6DD, UK
| | - Jessica Strid
- Division of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Ronjon Chakraverty
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK and Cancer Institute Department of Haematology, Division of Cancer Studies, University College London, London WC1E 6DD, UK
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Clare L Bennett
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK and Cancer Institute Department of Haematology, Division of Cancer Studies, University College London, London WC1E 6DD, UK.
| |
Collapse
|
13
|
Becerril-García MA, Yam-Puc JC, Maqueda-Alfaro R, Beristain-Covarrubias N, Heras-Chavarría M, Gallegos-Hernández IA, Calderón-Amador J, Munguía-Fuentes R, Donis-Maturano L, Flores-Langarica A, Flores-Romo L. Langerhans Cells From Mice at Birth Express Endocytic- and Pattern Recognition-Receptors, Migrate to Draining Lymph Nodes Ferrying Antigen and Activate Neonatal T Cells in vivo. Front Immunol 2020; 11:744. [PMID: 32395120 PMCID: PMC7197463 DOI: 10.3389/fimmu.2020.00744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 04/01/2020] [Indexed: 11/13/2022] Open
Abstract
Antigen capturing at the periphery is one of the earliest, crucial functions of antigen-presenting cells (APCs) to initiate immune responses. Langerhans cells (LCs), the epidermal APCs migrate to draining lymph nodes (DLNs) upon acquiring antigens. An arsenal of endocytic molecules is available to this end, including lectins and pathogen recognition receptors (PRRs). However, cutaneous LCs are poorly defined in the early neonatal period. We assessed endocytic molecules expression in situ: Mannose (CD206)-, Scavenger (SRA/CD204)-, Complement (CD2l, CDllb)-, and Fc-Receptors (CD16/32, CD23) as well as CD1d, CD14, CD205, Langerin (CD207), MHCII, and TLR4 in unperturbed epidermal LCs from both adult and early neonatal mice. As most of these markers were negative at birth (day 0), LC presence was revealed with the conspicuous, epidermal LC-restricted ADPase (and confirmed with CD45) staining detecting that they were as numerous as adult ones. Unexpectedly, most LCs at day 0 expressed CD14 and CD204 while very few were MHCII+ and TLR4+. In contrast, adult LCs lacked all these markers except Langerin, CD205, CD11b, MHCII and TLR4. Intriguingly, the CD204+ and CD14+ LCs predominant at day 0, apparently disappeared by day 4. Upon cutaneous FITC application, LCs were reduced in the skin and a CD204+MHCII+FITC+ population with high levels of CD86 subsequently appeared in DLNs, with a concomitant increased percentage of CD3+CD69+ T cells, strongly suggesting that neonatal LCs were able both to ferry the cutaneous antigen into DLNs and to activate neonatal T cells in vivo. Cell cycle analysis indicated that neonatal T cells in DLNs responded with proliferation. Our study reveals that epidermal LCs are present at birth, but their repertoire of endocytic molecules and PRRs differs to that of adult ones. We believe this to be the first description of CDl4, CD204 and TLR4 in neonatal epidermal LCs in situ. Newborns' LCs express molecules to detect antigens during early postnatal periods, are able to take up local antigens and to ferry them into DLNs conveying the information to responsive neonatal T cells.
Collapse
Affiliation(s)
- Miguel Angel Becerril-García
- Department of Cell Biology, Center for Advanced Research, The National Polytechnic Institute, Cinvestav-IPN, Mexico City, Mexico
| | - Juan Carlos Yam-Puc
- Department of Cell Biology, Center for Advanced Research, The National Polytechnic Institute, Cinvestav-IPN, Mexico City, Mexico.,Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Raúl Maqueda-Alfaro
- Department of Cell Biology, Center for Advanced Research, The National Polytechnic Institute, Cinvestav-IPN, Mexico City, Mexico
| | - Nonantzin Beristain-Covarrubias
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Monica Heras-Chavarría
- Department of Cell Biology, Center for Advanced Research, The National Polytechnic Institute, Cinvestav-IPN, Mexico City, Mexico
| | - Isis Amara Gallegos-Hernández
- Department of Cell Biology, Center for Advanced Research, The National Polytechnic Institute, Cinvestav-IPN, Mexico City, Mexico
| | - Juana Calderón-Amador
- Department of Cell Biology, Center for Advanced Research, The National Polytechnic Institute, Cinvestav-IPN, Mexico City, Mexico
| | - Rosario Munguía-Fuentes
- Department of Cell Biology, Center for Advanced Research, The National Polytechnic Institute, Cinvestav-IPN, Mexico City, Mexico
| | - Luis Donis-Maturano
- Department of Cell Biology, Center for Advanced Research, The National Polytechnic Institute, Cinvestav-IPN, Mexico City, Mexico
| | - Adriana Flores-Langarica
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Leopoldo Flores-Romo
- Department of Cell Biology, Center for Advanced Research, The National Polytechnic Institute, Cinvestav-IPN, Mexico City, Mexico
| |
Collapse
|
14
|
Botting RA, Haniffa M. The developing immune network in human prenatal skin. Immunology 2020; 160:149-156. [PMID: 32173857 PMCID: PMC7218404 DOI: 10.1111/imm.13192] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023] Open
Abstract
Establishment of a well‐functioning immune network in skin is crucial for its barrier function. This begins in utero alongside the structural differentiation and maturation of skin, and continues to expand and diversify across the human lifespan. The microenvironment of the developing human skin supports immune cell differentiation and has an overall anti‐inflammatory profile. Immunologically inert and skewed immune populations found in developing human skin promote wound healing, and as such may play a crucial role in the structural changes occurring during skin development.
Collapse
Affiliation(s)
- Rachel Anne Botting
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Muzlifah Haniffa
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.,Wellcome Sanger Institute, Hinxton, UK.,Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
15
|
Amon L, Lehmann CHK, Heger L, Heidkamp GF, Dudziak D. The ontogenetic path of human dendritic cells. Mol Immunol 2020; 120:122-129. [PMID: 32114182 DOI: 10.1016/j.molimm.2020.02.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/15/2020] [Indexed: 02/08/2023]
Abstract
Dendritic cells (DCs) orchestrate adaptive immune responses. In healthy individuals, DCs are drivers and fine-tuners of T cell responses directed against invading pathogens or cancer cells. In parallel, DCs control autoreactive T cells, thereby maintaining T cell tolerance. Under various disease conditions, a disruption of this delicate balance can lead to chronic infections, tumor evasion, or autoimmunity. While great efforts have been made to unravel the origin and development of this powerful cell type in mice, only little is known about the ontogeny of human DCs. Here, we summarize the current understanding of the developmental path of DCs from hematopoietic stem cells to fully functional DCs in their local tissue environment and provide a template for the identification of DCs across various tissues.
Collapse
Affiliation(s)
- Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Hartmannstraße 14, D-91052 Erlangen, Germany
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Hartmannstraße 14, D-91052 Erlangen, Germany; Medical Immunology Campus Erlangen, D-91054 Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Hartmannstraße 14, D-91052 Erlangen, Germany
| | - Gordon F Heidkamp
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Hartmannstraße 14, D-91052 Erlangen, Germany; Roche Innovation Center Munich, Roche Pharmaceutical Research and Early Development, pRED, Munich, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Hartmannstraße 14, D-91052 Erlangen, Germany; Medical Immunology Campus Erlangen, D-91054 Erlangen, Germany.
| |
Collapse
|
16
|
Borek I, Köffel R, Feichtinger J, Spies M, Glitzner-Zeis E, Hochgerner M, Sconocchia T, Krump C, Tam-Amersdorfer C, Passegger C, Benezeder T, Tittes J, Redl A, Painsi C, Thallinger GG, Wolf P, Stary G, Sibilia M, Strobl H. BMP7 aberrantly induced in the psoriatic epidermis instructs inflammation-associated Langerhans cells. J Allergy Clin Immunol 2019; 145:1194-1207.e11. [PMID: 31870764 DOI: 10.1016/j.jaci.2019.12.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 12/08/2019] [Accepted: 12/13/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Epidermal hyperplasia represents a morphologic hallmark of psoriatic skin lesions. Langerhans cells (LCs) in the psoriatic epidermis engage with keratinocytes (KCs) in tight physical interactions; moreover, they induce T-cell-mediated immune responses critical to psoriasis. OBJECTIVE This study sought to improve the understanding of epidermal factors in psoriasis pathogenesis. METHODS BMP7-LCs versus TGF-β1-LCs were phenotypically characterized and their functional properties were analyzed using flow cytometry, cell kinetic studies, co-culture with CD4 T cells, and cytokine measurements. Furthermore, immunohistology of healthy and psoriatic skin was performed. Additionally, in vivo experiments with Junf/fJunBf/fK5cre-ERT mice were carried out to assess the role of bone morphogenetic protein (BMP) signaling in psoriatic skin inflammation. RESULTS This study identified a KC-derived signal (ie, BMP signaling) to promote epidermal changes in psoriasis. Whereas BMP7 is strictly confined to the basal KC layer in the healthy skin, it is expressed at high levels throughout the lesional psoriatic epidermis. BMP7 instructs precursor cells to differentiate into LCs that phenotypically resemble psoriatic LCs. These BMP7-LCs exhibit proliferative activity and increased sensitivity to bacterial stimulation. Moreover, aberrant high BMP signaling in the lesional epidermis is mediated by a KC intrinsic mechanism, as suggested from murine data and clinical outcome after topical antipsoriatic treatment in human patients. CONCLUSIONS These data indicate that available TGF-β family members within the lesional psoriatic epidermis preferentially signal through the canonical BMP signaling cascade to instruct inflammatory-type LCs and to promote psoriatic epidermal changes. Targeting BMP signaling might allow to therapeutically interfere with cutaneous psoriatic manifestations.
Collapse
Affiliation(s)
- Izabela Borek
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - René Köffel
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Julia Feichtinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria
| | - Melanie Spies
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Elisabeth Glitzner-Zeis
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Mathias Hochgerner
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Tommaso Sconocchia
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Corinna Krump
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Carmen Tam-Amersdorfer
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Christina Passegger
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Theresa Benezeder
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Julia Tittes
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Anna Redl
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Clemens Painsi
- Department of Dermatology, State Hospital Klagenfurt, Klagenfurt, Austria
| | - Gerhard G Thallinger
- Institute of Computational Biotechnology, Graz University of Technology, Graz, Austria; Omics Center Graz, BioTechMed Graz, Graz, Austria
| | - Peter Wolf
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Georg Stary
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Maria Sibilia
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Herbert Strobl
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
17
|
Haid B, Reider D, Nägele F, Spinoit AF, Pechriggl E, Romani N, Fritsch H, Oswald J. Langerhans cells in hypospadias: an analysis of Langerin (CD207) and HLA-DR on epidermal sheets and full thickness skin sections. BMC Urol 2019; 19:114. [PMID: 31718599 PMCID: PMC6852928 DOI: 10.1186/s12894-019-0551-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 10/31/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hypospadias are among the most common genital malformations. Langerhans Cells (LCs) play a pivotal role in HIV and HPV infection. The migration of LC precursors to skin coincides with the embryonic period of hypospadias development and genetic alterations leading to the formation of hypospadias impact the development of ectodermally derived tissues. We hypothesized that this might be associated with a difference in frequency or morphology of epidermal and dermal LCs in hypospadias patients. METHODS A total of 43 patients from two centers were prospectively included into this study after parental consent and ethics approval. Epidermal and dermal sheets were prepared from skin samples of 26 patients with hypospadias, 13 patients without penile malformations and 4 patients with penile malformations other than hypospadias. Immunofluorescence staining of sheets was performed with anti-HLA-DR-FITC and anti-CD207/Langerin-A594 antibodies. Skin sections from 11 patients without penile malformation and 11 patients with hypospadias were stained for Langerin. Frequencies as well as morphology and distribution of epidermal and dermal LCs on sheets and sections were microscopically evaluated. Cell counts were compared by unpaired t-tests. RESULTS There was no difference in frequency of epidermal LCs, Neither on sheets (873 ± 61 vs. 940 ± 84LCs/mm2, p = 0.522) nor on sections (32 ± 3 vs. 30 ± 2LCs/mm2, p = 0.697). Likewise, the frequency of dermal LCs (5,9 ± 0,9 vs. 7.5 ± 1.3LCs/mm2, p = 0.329) was comparable between patients with hypospadias and without penile malformation. No differences became apparent in subgroup analyses, comparing distal to proximal hypospadias (p = 0.949), younger and older boys (p = 0.818) or considering topical dihydrotestosterone treatment prior to surgery (p = 0.08). The morphology of the LCs was not different comparing hypospadias patients with boys without penile malformations. CONCLUSIONS LCs are present in similar frequencies and with a comparable morphology and distribution in patients with hypospadias as compared to children without penile malformations. This suggests that patients with hypospadias are not different from patients with normal penile development considering this particular compartment of their skin immunity.
Collapse
Affiliation(s)
- Bernhard Haid
- Department of Pediatric Urology, Hospital of the Sisters of Charity, Ordensklinikum Linz, Seilerstätte 4, 4020, Linz, Austria. .,Department of Urology, Ludwig Maximilians University, Marchioninistraße 15, 81367, Munich, Germany.
| | - Daniela Reider
- Department for Dermatology and Venereology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Felix Nägele
- Section for clinical and functional Anatomy, Medical University Innsbruck, Müllerstraße 59, 6020, Innsbruck, Austria
| | - Anne-Françoise Spinoit
- Department of Urology, University Clinic Gent, Corneel Heymanslaan 10, 9000, Gent, Belgium
| | - Elisabeth Pechriggl
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University Innsbruck, Innerkoflerstraße 1, Innsbruck, Austria
| | - Nikolaus Romani
- Department for Dermatology and Venereology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Helga Fritsch
- Section for clinical and functional Anatomy, Medical University Innsbruck, Müllerstraße 59, 6020, Innsbruck, Austria
| | - Josef Oswald
- Department of Pediatric Urology, Hospital of the Sisters of Charity, Ordensklinikum Linz, Seilerstätte 4, 4020, Linz, Austria
| |
Collapse
|
18
|
Nelson DS, Marano RL, Joo Y, Tian SY, Patel B, Kaplan DH, Shlomchik MJ, Stevenson K, Bronson RT, Rollins BJ. BRAF V600E and Pten deletion in mice produces a histiocytic disorder with features of Langerhans cell histiocytosis. PLoS One 2019; 14:e0222400. [PMID: 31527903 PMCID: PMC6748438 DOI: 10.1371/journal.pone.0222400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/28/2019] [Indexed: 12/28/2022] Open
Abstract
Langerhans cell histiocytosis (LCH) is characterized by the accumulation of Langerin (CD207)-expressing histiocytes. Mutational activation of mitogen-activated protein kinase pathway genes, in particular BRAF, drives most cases. To test whether activated BRAF is sufficient for the development of LCH, we engineered mice to express BRAF V600E under the control of the human Langerin promoter. These mice have shortened survivals, smaller lymphoid organs, absent Leydig cells, and fewer epidermal LCs than controls, but do not accumulate histiocytes. To test whether the absence of histiocyte proliferation could be due to oncogene-induced senescence, we engineered homozygous Pten loss in the same cells that expressed BRAF V600E. Like mice with intact Pten, these mice have shortened survivals, smaller thymi, and absent Leydig cells. However, loss of Pten also leads to the accumulation of CD207+ histiocytes in spleen, thymus, and some lymph nodes. While many CD207+ histiocytes in the thymus are CD8-, reminiscent of LCH cells, the CD207+ histiocytes in the spleen and lymph nodes are CD8+. These mice also accumulate large numbers of CD207- cells in the lamina propria (LP) of the small intestine. Both the lymphoid and LP phenotypes are likely due to human Langerin promoter-driven BRAF V600E expression in resident CD8+ dendritic cells in the former and LP dendritic cells in the latter and confirm that Pten loss is required to overcome inhibitory pathways induced by BRAF V600E expression. The complex phenotype of these mice is a consequence of the multiple murine cell types in which the human Langerin promoter is active.
Collapse
Affiliation(s)
- David S. Nelson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States of America
| | - Ryan L. Marano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States of America
| | - Yechaan Joo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States of America
| | - Sara Y. Tian
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States of America
| | - Bhumi Patel
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States of America
| | - Daniel H. Kaplan
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Mark J. Shlomchik
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Kristen Stevenson
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Roderick T. Bronson
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, United States of America
| | - Barrett J. Rollins
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States of America
- Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
19
|
Kienzl P, Polacek R, Reithofer M, Reitermaier R, Hagenbach P, Tajpara P, Vierhapper M, Gschwandtner M, Mildner M, Jahn-Schmid B, Elbe-Bürger A. The cytokine environment influence on human skin-derived T cells. FASEB J 2019; 33:6514-6525. [PMID: 30807238 PMCID: PMC6463918 DOI: 10.1096/fj.201801416r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Skin resident T cells provide immediate immunologic responses at their specific location and play a role in the pathogenesis of skin diseases such as psoriasis. Recently, IL-9-producing T cells were described as a major T-cell subtype present in the skin, but knowledge on the biology and in situ regulation of this T-cell subtype is scarce. Here, we investigated the cytokine influence on skin T cells with focus on IL-9-producing T cells because a better understanding of their biology may identify novel therapeutic approaches. Healthy human skin biopsies were cultured either in the presence of IL-2, IL-4, and TGF-β [T helper (Th)9-promoting condition (Th9-PC)] or IL-2 and IL-15 [standard condition (SC)]. Paired analysis of enzymatically isolated skin T cells and emigrated T cells after 4 wk of skin culture showed significant alterations of T-cell phenotypes, cytokine production, and IL-9-producing T-cell frequency. RNA sequencing analysis revealed differentially regulated pathways and identified CXCL8 and CXCL13 as top up-regulated genes in Th9-PC compared with SC. Functionally supernatant of stimulated skin-derived T cells, CXCL8 and CXCL13 increased neutrophil survival. We report that the cytokine environment alters skin-derived T-cell phenotype and functional properties.-Kienzl, P., Polacek, R., Reithofer, M., Reitermaier, R., Hagenbach, P., Tajpara, P., Vierhapper, M., Gschwandtner, M., Mildner, M. Jahn-Schmid, B., Elbe-Bürger, A. The cytokine environment influence on human skin-derived T cells.
Collapse
Affiliation(s)
- Philip Kienzl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Romana Polacek
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Manuel Reithofer
- Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - René Reitermaier
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Pia Hagenbach
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Pooja Tajpara
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Martin Vierhapper
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Maria Gschwandtner
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Michael Mildner
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Beatrice Jahn-Schmid
- Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
20
|
Hovav AH. Mucosal and Skin Langerhans Cells – Nurture Calls. Trends Immunol 2018; 39:788-800. [DOI: 10.1016/j.it.2018.08.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/15/2018] [Accepted: 08/15/2018] [Indexed: 10/28/2022]
|
21
|
Strobl H, Krump C, Borek I. Micro-environmental signals directing human epidermal Langerhans cell differentiation. Semin Cell Dev Biol 2018; 86:36-43. [PMID: 29448069 DOI: 10.1016/j.semcdb.2018.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/12/2017] [Accepted: 02/10/2018] [Indexed: 01/11/2023]
Abstract
Human Langerhans cells (LC) can be generated ex vivo from hematopoietic precursor cells in response to cytokines and cell-membrane associated ligands. These in vitro differentiation models provided mechanistic insights into the molecular and cellular pathways underlying the development of this unique, epithelia-associated dendritic cell subset. Notably, the human epidermal microenvironment is fully sufficient to induce LC differentiation from hematopoietic progenitors. Hence, dissecting the molecular characteristics of the human epithelial/epidermal LC niche, and testing defined ligands for their capacity to induce LC differentiation, led to a refined molecular model of LC lineage commitment. During epidermal ontogeny, spatially and temporally regulated availability of TGF-β family members cooperate with other keratinocyte-derived signals, such as E-cadherin and Notch ligands, for instructing LC differentiation. In this review, we discuss the signals known to instruct human hematopoietic progenitor cells and myelomonocytic cells to undergo LC lineage commitment. Additionally, the current methods for generation of large numbers of human LC-like cells ex vivo in defined serum-free media are discussed.
Collapse
Affiliation(s)
- Herbert Strobl
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria.
| | - Corinna Krump
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Izabela Borek
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| |
Collapse
|
22
|
The Reticulum-Associated Protein RTN1A Specifically Identifies Human Dendritic Cells. J Invest Dermatol 2018; 138:1318-1327. [PMID: 29369773 DOI: 10.1016/j.jid.2018.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/22/2017] [Accepted: 01/03/2018] [Indexed: 12/21/2022]
Abstract
RTN1 is an endoplasmic reticulum-associated protein that was initially identified in neuronal tissues. Here we show that the main isoform RTN1A is a marker for dendritic cells. In the skin, HLA-DR+CD1ahighCD207+CD11cweak Langerhans cells were the only cells in the epidermis, and HLA-DR+CD11c+ dendritic cells were the main cells in the dermis, expressing this protein. RTN1A+ dendritic cells were also found in gingiva, trachea, tonsil, thymus, and peripheral blood. During differentiation of MUTZ-3 cells into Langerhans cells, expression of RTN1A mRNA and protein preceded established Langerhans cell markers CD1a and CD207, and RTN1A protein partially co-localized with the endoplasmic reticulum marker protein disulfide isomerase. In line with this observation, we found that RTN1A was expressed by around 80% of Langerhans cell precursors in human embryonic skin. Our findings show that RTN1A is a marker for cells of the dendritic lineage, including Langerhans cells and dermal dendritic cells. This unexpected finding will serve as a starting point for the elucidation of the, until now, elusive functional roles of RTN1A in both the immune and the nervous system.
Collapse
|
23
|
In utero development of memory T cells. Semin Immunopathol 2017; 39:585-592. [DOI: 10.1007/s00281-017-0650-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 08/07/2017] [Indexed: 12/24/2022]
|
24
|
Abstract
The maintenance of monocytes, macrophages, and dendritic cells (DCs) involves manifold pathways of ontogeny and homeostasis that have been the subject of intense study in recent years. The concept of a peripheral mononuclear phagocyte system continually renewed by blood-borne monocytes has been modified to include specialized DC pathways of development that do not involve monocytes, and longevity through self-renewal of tissue macrophages. The study of development remains difficult owing to the plasticity of phenotypes and misconceptions about the fundamental structure of hematopoiesis. However, greater clarity has been achieved in distinguishing inflammatory monocyte-derived DCs from DCs arising in the steady state, and new concepts of conjoined lymphomyeloid hematopoiesis more easily accommodate the shared lymphoid and myeloid phenotypes of some DCs. Cross-species comparisons have also yielded coherent systems of nomenclature for all mammalian monocytes, macrophages, and DCs. Finally, the clear relationships between ontogeny and functional specialization offer information about the regulation of immune responses and provide new tools for the therapeutic manipulation of myeloid mononuclear cells in medicine.
Collapse
|
25
|
Georgountzou A, Papadopoulos NG. Postnatal Innate Immune Development: From Birth to Adulthood. Front Immunol 2017; 8:957. [PMID: 28848557 PMCID: PMC5554489 DOI: 10.3389/fimmu.2017.00957] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/26/2017] [Indexed: 12/20/2022] Open
Abstract
It is well established that adaptive immune responses are deficient in early life, contributing to increased mortality and morbidity. The developmental trajectories of different components of innate immunity are only recently being explored. Individual molecules, cells, or pathways of innate recognition and signaling, within different compartments/anatomical sites, demonstrate variable maturation patterns. Despite some discrepancies among published data, valuable information is emerging, showing that the developmental pattern of cytokine responses during early life is age and toll-like receptor specific, and may be modified by genetic and environmental factors. Interestingly, specific environmental exposures have been linked both to innate function modifications and the occurrence of chronic inflammatory disorders, such as respiratory allergies. As these conditions are on the rise, our knowledge on innate immune development and its modulating factors needs to be expanded. Improved understanding of the sequence of events associated with disease onset and persistence will lead toward meaningful interventions. This review describes the state-of-the-art on normal postnatal innate immune ontogeny and highlights research areas that are currently explored or should be further addressed.
Collapse
Affiliation(s)
- Anastasia Georgountzou
- Allergy and Clinical Immunology Unit, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos G Papadopoulos
- Allergy and Clinical Immunology Unit, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece.,Division of Infection, Inflammation and Respiratory Medicine, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
26
|
Chen JC, Chan CC, Wu CJ, Ou LS, Yu HY, Chang HL, Tseng LY, Kuo ML. Fetal Phagocytes Take up Allergens to Initiate T-Helper Cell Type 2 Immunity and Facilitate Allergic Airway Responses. Am J Respir Crit Care Med 2017; 194:934-947. [PMID: 27064309 DOI: 10.1164/rccm.201508-1703oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
RATIONALE Actively acquired tolerance occurs when foreign antigens come into contact with the immature fetal immune system. OBJECTIVES Armed with the knowledge of actively acquired tolerance, we attempted to prenatally abolish or diminish allergic responses. METHODS In utero injection of adjuvant-free ovalbumin (OVA) was conducted in Gestational Day 14 FVB/N mouse fetuses. Postnatally, mice were evaluated for their resistance to intraperitoneal OVA sensitization and oral or aerosolized OVA challenge, and then they were examined for humoral and cellular immunological profiles, airway hyperresponsiveness to bronchospastic stimuli, and lung histology. Fluorescent conjugates of OVA were used for further studies of mechanisms. MEASUREMENTS AND MAIN RESULTS This presumed tolerogenic action turned out to be a sensitization process with the development of anaphylaxis or heightened recall, T-helper cell type 2-skewed responses to postnatal encounter with OVA. Further postnatal aerosolized OVA stress triggered allergic lungs with functional and structural alterations of airways. The unintended consequence resulted from macrophage-like fetal phagocytes that took up OVA and differentiated toward dendritic cells. These fetal dendritic cell progenitors attenuated proteolysis of endocytosed OVA for delayed presentation in postnatal life. This specialty of fetal phagocytes effectively retains the memory of antigens internalized early before full development of the immune system, leading to an event of in utero sensitization. CONCLUSIONS Our results have mechanical implications for prenatal imprinting of atopy and shed light on the importance of fetal phagocytes in shaping the developing immune system and initiating allergic airway diseases.
Collapse
Affiliation(s)
- Jeng-Chang Chen
- 1 Department of Surgery.,2 Graduate Institute of Clinical Medical Sciences and
| | - Cheng-Chi Chan
- 3 Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; and
| | - Chia-Jen Wu
- 3 Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; and
| | - Liang-Shiou Ou
- 4 Division of Allergy, Asthma and Rheumatology, Department of Pediatrics, and
| | - Hsiu-Yueh Yu
- 5 Pediatric Research Center, Chang Gung Children's Hospital, Taoyuan, Taiwan
| | - Hsueh-Ling Chang
- 5 Pediatric Research Center, Chang Gung Children's Hospital, Taoyuan, Taiwan
| | - Li-Yun Tseng
- 5 Pediatric Research Center, Chang Gung Children's Hospital, Taoyuan, Taiwan
| | - Ming-Ling Kuo
- 4 Division of Allergy, Asthma and Rheumatology, Department of Pediatrics, and.,3 Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; and.,6 Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
27
|
Human fetal dendritic cells promote prenatal T-cell immune suppression through arginase-2. Nature 2017; 546:662-666. [PMID: 28614294 DOI: 10.1038/nature22795] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 04/27/2017] [Indexed: 12/24/2022]
Abstract
During gestation the developing human fetus is exposed to a diverse range of potentially immune-stimulatory molecules including semi-allogeneic antigens from maternal cells, substances from ingested amniotic fluid, food antigens, and microbes. Yet the capacity of the fetal immune system, including antigen-presenting cells, to detect and respond to such stimuli remains unclear. In particular, dendritic cells, which are crucial for effective immunity and tolerance, remain poorly characterized in the developing fetus. Here we show that subsets of antigen-presenting cells can be identified in fetal tissues and are related to adult populations of antigen-presenting cells. Similar to adult dendritic cells, fetal dendritic cells migrate to lymph nodes and respond to toll-like receptor ligation; however, they differ markedly in their response to allogeneic antigens, strongly promoting regulatory T-cell induction and inhibiting T-cell tumour-necrosis factor-α production through arginase-2 activity. Our results reveal a previously unappreciated role of dendritic cells within the developing fetus and indicate that they mediate homeostatic immune-suppressive responses during gestation.
Collapse
|
28
|
Jurkin J, Krump C, Köffel R, Fieber C, Schuster C, Brunner PM, Borek I, Eisenwort G, Lim C, Mages J, Lang R, Bauer W, Mechtcheriakova D, Meshcheryakova A, Elbe-Bürger A, Stingl G, Strobl H. Human skin dendritic cell fate is differentially regulated by the monocyte identity factor Kruppel-like factor 4 during steady state and inflammation. J Allergy Clin Immunol 2017; 139:1873-1884.e10. [PMID: 27742396 PMCID: PMC5538449 DOI: 10.1016/j.jaci.2016.09.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/25/2016] [Accepted: 09/09/2016] [Indexed: 11/01/2022]
Abstract
BACKGROUND Langerhans cell (LC) networks play key roles in immunity and tolerance at body surfaces. LCs are established prenatally and can be replenished from blood monocytes. Unlike skin-resident dermal DCs (dDCs)/interstitial-type DCs and inflammatory dendritic epidermal cells appearing in dermatitis/eczema lesions, LCs lack key monocyte-affiliated markers. Inversely, LCs express various epithelial genes critical for their long-term peripheral tissue residency. OBJECTIVE Dendritic cells (DCs) are functionally involved in inflammatory diseases; however, the mechanisms remained poorly understood. METHODS In vitro differentiation models of human DCs, gene profiling, gene transduction, and immunohistology were used to identify molecules involved in DC subset specification. RESULTS Here we identified the monocyte/macrophage lineage identity transcription factor Kruppel-like factor 4 (KLF4) to be inhibited during LC differentiation from human blood monocytes. Conversely, KLF4 is maintained or induced during dermal DC and monocyte-derived dendritic cell/inflammatory dendritic epidermal cell differentiation. We showed that in monocytic cells KLF4 has to be repressed to allow their differentiation into LCs. Moreover, respective KLF4 levels in DC subsets positively correlate with proinflammatory characteristics. We identified epithelial Notch signaling to repress KLF4 in monocytes undergoing LC commitment. Loss of KLF4 in monocytes transcriptionally derepresses Runt-related transcription factor 3 in response to TGF-β1, thereby allowing LC differentiation marked by a low cytokine expression profile. CONCLUSION Monocyte differentiation into LCs depends on activation of Notch signaling and the concomitant loss of KLF4.
Collapse
Affiliation(s)
- Jennifer Jurkin
- Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Corinna Krump
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - René Köffel
- Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Christina Fieber
- Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Christopher Schuster
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Patrick M Brunner
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Izabela Borek
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - Gregor Eisenwort
- Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Clarice Lim
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria; Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Jörg Mages
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang Bauer
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Diana Mechtcheriakova
- Departments of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Anastasia Meshcheryakova
- Departments of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Adelheid Elbe-Bürger
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Georg Stingl
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Herbert Strobl
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria; Institute of Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
29
|
Picarda G, Chéneau C, Humbert JM, Bériou G, Pilet P, Martin J, Duteille F, Perrot P, Bellier-Waast F, Heslan M, Haspot F, Guillon F, Josien R, Halary FA. Functional Langerinhigh-Expressing Langerhans-like Cells Can Arise from CD14highCD16−Human Blood Monocytes in Serum-Free Condition. THE JOURNAL OF IMMUNOLOGY 2016; 196:3716-28. [DOI: 10.4049/jimmunol.1501304] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 02/23/2016] [Indexed: 12/31/2022]
|
30
|
Walraven M, Talhout W, Beelen RHJ, van Egmond M, Ulrich MMW. Healthy human second-trimester fetal skin is deficient in leukocytes and associated homing chemokines. Wound Repair Regen 2016; 24:533-41. [PMID: 26873861 DOI: 10.1111/wrr.12421] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 02/01/2016] [Indexed: 12/22/2022]
Abstract
The lack of immune cells in mid-gestational fetal skin is often mentioned as a key factor underlying scarless healing. However, the scarless healing ability is conserved until long after the immune system in the fetus is fully developed. Therefore, we studied human second-trimester fetal skin and compared the numbers of immune cells and chemokine levels from fetal skin with adult skin. By using immunohistochemistry, we show that healthy fetal skin contains significant lower numbers of CD68(+) -macrophages, Tryptase(+) -mast cells, Langerin(+) -Langerhans cells, CD1a(+) -dendritic cells, and CD3(+) -T cells compared to adult skin. Staining with an early lineage leukocyte marker, i.e., CD45, verified that the number of CD45(+) -immune cells was indeed significantly lower in fetal skin but that sufficient numbers of immune cells were present in the fetal lymph node. No differences in the vascular network were observed between fetal and adult skin. Moreover, significant lower levels of lymphocyte chemokines CCL17, CCL21, and CCL27 were observed in fetal skin. However, levels of inflammatory interleukins such as IL-6, IL-8, and IL-10 were undetectable and levels of CCL2 were similar in healthy fetal and adult skin. In conclusion, this study shows that second-trimester fetal skin contains low levels of immune cells and leukocyte chemokines compared to adult skin. This immune cell deficiency includes CD45(+) leukocytes, despite the abundant presence of these cells in the lymph node. The immune deficiency in healthy second-trimester fetal skin may result in reduced inflammation during wound healing, and could underlie the scarless healing capacities of the fetal skin.
Collapse
Affiliation(s)
- Mariëlle Walraven
- Department of Molecular Cell Biology & Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Wendy Talhout
- Department of Molecular Cell Biology & Immunology, VU University Medical Center, Amsterdam, The Netherlands.,Association of Dutch Burn Centers, Beverwijk, The Netherlands
| | - Robert H J Beelen
- Department of Molecular Cell Biology & Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW This article summarizes recent research on the ontogeny of Langerhans cells and regulation of their homeostasis in quiescent and inflamed conditions. RECENT FINDINGS Langerhans cells originate prenatally and may endure throughout life, independently of bone marrow-derived precursors. Fate-mapping experiments have recently resolved the relative contribution of primitive yolk sac and fetal liver hematopoiesis to the initial formation of Langerhans cells. In postnatal life, local self-renewal restores Langerhans cell numbers following chronic or low-grade inflammatory insults. However, severe inflammation recruits de-novo bone marrow-derived precursors in two waves; a transient population of classical monocytes followed by uncharacterized myeloid precursors that form a stable self-renewing Langerhans cell network as inflammation subsides. Human CD1c⁺ dendritic cells have Langerhans cell potential in vitro, raising the possibility that dendritic cell progenitors provide the second wave. Langerhans cell development depends upon transforming growth factor beta receptor signaling with distinct pathways active during differentiation and homeostasis. Langerhans cell survival is mediated by multiple pathways including mechanistic target of rapamycin and extracellular signal-regulated kinase signaling, mechanisms that become highly relevant in Langerhans cell neoplasia. SUMMARY The study of Langerhans cells continues to provide novel and unexpected insights into the origin and regulation of myeloid cell populations. The melding of macrophage and dendritic cell biology, shaped by a unique habitat, is a special feature of Langerhans cells.
Collapse
Affiliation(s)
- Matthew Collin
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne, UK
| | | |
Collapse
|
32
|
Abstract
Langerhans cell histiocytosis (LCH) is heterogeneous disease characterized by common histology of inflammatory lesions containing Langerin(+) (CD207) histiocytes. Emerging data support a model in which MAPK activation in self-renewing hematopoietic progenitors may drive disseminated high-risk disease, whereas MAPK activation in more differentiated committed myeloid populations may induce low-risk LCH. The heterogeneous clinical manifestations with shared histology may represent the final common pathway of an acquired defect of differentiation, initiated at more than one point. Implications of this model include re-definition of LCH as a myeloid neoplasia and re-focusing therapeutic strategies on the cells and lineages of origin.
Collapse
Affiliation(s)
- Matthew Collin
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Venetia Bigley
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Kenneth L McClain
- Texas Children's Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Carl E Allen
- Texas Children's Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
33
|
Development of Blood and Lymphatic Endothelial Cells in Embryonic and Fetal Human Skin. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2563-74. [PMID: 26188132 DOI: 10.1016/j.ajpath.2015.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 04/08/2015] [Accepted: 05/13/2015] [Indexed: 12/26/2022]
Abstract
Blood and lymphatic vessels provide nutrients for the skin and fulfill important homeostatic functions, such as the regulation of immunologic processes. In this study, we investigated the development of blood and lymphatic endothelial cells in prenatal human skin in situ using multicolor immunofluorescence and analyzed angiogenic molecules by protein arrays of lysates and cell culture supernatants. We found that at 8 to 10 weeks of estimated gestational age, CD144(+) vessels predominantly express the venous endothelial cell marker PAL-E, whereas CD144(+)PAL-E(-) vessels compatible with arteries only appear at the end of the first trimester. Lymphatic progenitor cells at 8 weeks of estimated gestational age express CD31, CD144, Prox1, and temporary PAL-E. At that developmental stage not all lymphatic progenitor cells express podoplanin or Lyve-1, which are acquired with advancing gestational age in a stepwise fashion. Already in second-trimester human skin, the phenotype of blood and lymphatic vessels roughly resembles the one in adult skin. The expression pattern of angiogenic molecules in lysates and cell culture supernatants of prenatal skin did not reveal the expected bent to proangiogenic molecules, indicating a complex regulation of angiogenesis during ontogeny. In summary, this study provides enticing new insights into the development and phenotypic characteristics of the vascular system in human prenatal skin.
Collapse
|
34
|
Truong A, Ahmad RC, Jennings C, Benjamin L, Kim J. Congenital cutaneous Langerhans histiocytosis with mixed cell populations. J Cutan Pathol 2015; 42:1031-1033. [DOI: 10.1111/cup.12536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 11/30/2022]
Affiliation(s)
- Allison Truong
- Stanford School of Medicine; Stanford University Medical Center; Redwood City CA USA
| | - Regina-Celeste Ahmad
- Department of Dermatology; Stanford University Medical Center; Redwood City CA USA
| | - Charay Jennings
- Department of Pathology; Stanford University Medical Center; Redwood City CA USA
| | - Latanya Benjamin
- Department of Dermatology; Stanford University Medical Center; Redwood City CA USA
- Department of Pediatrics; Lucile Packard Children's Hospital; Stanford CA USA
| | - Jinah Kim
- Department of Dermatology; Stanford University Medical Center; Redwood City CA USA
- Department of Pathology; Stanford University Medical Center; Redwood City CA USA
| |
Collapse
|
35
|
The TGF-β superfamily in dendritic cell biology. Cytokine Growth Factor Rev 2015; 26:647-57. [PMID: 26115564 DOI: 10.1016/j.cytogfr.2015.06.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 06/15/2015] [Indexed: 12/18/2022]
Abstract
The TGF-β superfamily consists of a large group of pleiotropic cytokines that are involved in the regulation of many developmental, physiological and pathological processes. Dendritic cells are antigen-presenting cells that play a key role in innate and adaptive immune responses. Dendritic cells have a complex relationship with the TGF-β cytokine superfamily being both source and targets for many of these cytokines. Some TGF-β family members are expressed by dendritic cells and modulate immune responses, for instance through the induction of T cell polarization. Others play a crucial role in the development and function of the different dendritic cell subsets. This review summarizes the current knowledge on the role of TGF-β family cytokines in dendritic cell biology, focusing on TGF-β as well as on other, less characterized, members of these important immune mediators.
Collapse
|
36
|
Schöppl A, Botta A, Prior M, Akgün J, Schuster C, Elbe-Bürger A. Langerhans cell precursors acquire RANK/CD265 in prenatal human skin. Acta Histochem 2015; 117:425-30. [PMID: 25722033 PMCID: PMC4516852 DOI: 10.1016/j.acthis.2015.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 01/20/2015] [Accepted: 01/29/2015] [Indexed: 02/01/2023]
Abstract
The skin is the first barrier against foreign pathogens and the prenatal formation of a strong network of various innate and adaptive cells is required to protect the newborn from perinatal infections. While many studies about the immune system in healthy and diseased adult human skin exist, our knowledge about the cutaneous prenatal/developing immune system and especially about the phenotype and function of antigen-presenting cells such as epidermal Langerhans cells (LCs) in human skin is still scarce. It has been shown previously that LCs in healthy adult human skin express receptor activator of NF-κB (RANK), an important molecule prolonging their survival. In this study, we investigated at which developmental stage LCs acquire this important molecule. Immunofluorescence double-labeling of cryostat sections revealed that LC precursors in prenatal human skin either do not yet [10–11 weeks of estimated gestational age (EGA)] or only faintly (13–15 weeks EGA) express RANK. LCs express RANK at levels comparable to adult LCs by the end of the second trimester. Comparable with adult skin, dermal antigen-presenting cells at no gestational age express this marker. These findings indicate that epidermal leukocytes gradually acquire RANK during gestation – a phenomenon previously observed also for other markers on LCs in prenatal human skin.
Collapse
Affiliation(s)
- Alice Schöppl
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases (DIAID), Laboratory of Cellular and Molecular Immunobiology of the Skin, Medical University of Vienna, Vienna, Austria
| | - Albert Botta
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases (DIAID), Laboratory of Cellular and Molecular Immunobiology of the Skin, Medical University of Vienna, Vienna, Austria
| | - Marion Prior
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases (DIAID), Laboratory of Cellular and Molecular Immunobiology of the Skin, Medical University of Vienna, Vienna, Austria
| | - Johnnie Akgün
- Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Christopher Schuster
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases (DIAID), Laboratory of Cellular and Molecular Immunobiology of the Skin, Medical University of Vienna, Vienna, Austria
| | - Adelheid Elbe-Bürger
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases (DIAID), Laboratory of Cellular and Molecular Immunobiology of the Skin, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
37
|
|
38
|
Pfisterer K, Lipnik KM, Hofer E, Elbe-Bürger A. CD90 + Human Dermal Stromal Cells Are Potent Inducers of FoxP3 + Regulatory T Cells. J Invest Dermatol 2015; 135:130-141. [DOI: 10.1038/jid.2014.309] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Revised: 06/15/2014] [Accepted: 06/30/2014] [Indexed: 01/14/2023]
|
39
|
McGovern N, Chan JKY, Ginhoux F. Dendritic cells in humans--from fetus to adult. Int Immunol 2014; 27:65-72. [PMID: 25323843 DOI: 10.1093/intimm/dxu091] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The human immune system evolves continuously during development from the embryo into the adult, reflecting the ever-changing environment and demands of our body. This ability of our immune system to sense external cues and adapt as we develop is just as important in the early tolerogenic environment of the fetus, as it is in the constantly pathogen-challenged adult. Dendritic cells (DCs), the professional antigen-sensing and antigen-presenting components of the immune system, play a crucial role in this process where they act as sentinels, both initiating and regulating immune responses. Here, we provide an overview of the human immune system in the developing fetus and the adult, with a focus on DC ontogeny and function during these discrete but intimately linked life stages.
Collapse
Affiliation(s)
- Naomi McGovern
- Singapore Immunology Network, Agency for Science, Technology and Research, 8A Biomedical Grove, IMMUNOS Building #3-4, BIOPOLIS, Singapore 138648, Singapore
| | | | - Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology and Research, 8A Biomedical Grove, IMMUNOS Building #3-4, BIOPOLIS, Singapore 138648, Singapore Department of Reproductive Medicine, Division of Obstetrics and Gynaecology, KK Women's and Children's Hospital, Singapore 229899, Singapore
| |
Collapse
|
40
|
Schuster C, Mildner M, Mairhofer M, Bauer W, Fiala C, Prior M, Eppel W, Kolbus A, Tschachler E, Stingl G, Elbe-Bürger A. Human embryonic epidermis contains a diverse Langerhans cell precursor pool. Development 2014; 141:807-15. [PMID: 24496618 DOI: 10.1242/dev.102699] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Despite intense efforts, the exact phenotype of the epidermal Langerhans cell (LC) precursors during human ontogeny has not been determined yet. These elusive precursors are believed to migrate into the embryonic skin and to express primitive surface markers, including CD36, but not typical LC markers such as CD1a, CD1c and CD207. The aim of this study was to further characterize the phenotype of LC precursors in human embryonic epidermis and to compare it with that of LCs in healthy adult skin. We found that epidermal leukocytes in first trimester human skin are negative for CD34 and heterogeneous with regard to the expression of CD1c, CD14 and CD36, thus contrasting the phenotypic uniformity of epidermal LCs in adult skin. These data indicate that LC precursors colonize the developing epidermis in an undifferentiated state, where they acquire the definitive LC marker profile with time. Using a human three-dimensional full-thickness skin model to mimic in vivo LC development, we found that FACS-sorted, CD207(-) cord blood-derived haematopoietic precursor cells resembling foetal LC precursors but not CD14(+)CD16(-) blood monocytes integrate into skin equivalents, and without additional exogenous cytokines give rise to cells that morphologically and phenotypically resemble LCs. Overall, it appears that CD14(-) haematopoietic precursors possess a much higher differentiation potential than CD14(+) precursor cells.
Collapse
Affiliation(s)
- Christopher Schuster
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases (DIAID), Laboratory of Cellular and Molecular Immunobiology of the Skin, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Visscher M, Narendran V. The Ontogeny of Skin. Adv Wound Care (New Rochelle) 2014; 3:291-303. [PMID: 24761361 DOI: 10.1089/wound.2013.0467] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 06/21/2013] [Indexed: 12/25/2022] Open
Abstract
Significance: During gestation, fetal skin progresses from a single layer derived from ectoderm to a complex, multi-layer tissue with the stratum corneum (SC) as the outermost layer. Innate immunity is a conferred complex process involving a balance of pro- and anti-inflammatory cytokines, structural proteins, and specific antigen-presenting cells. The SC is a part of the innate immune system as an impermeable physical barrier containing anti-microbial lipids and host defense proteins. Postnatally, the epidermis continually replenishes itself, provides a protective barrier, and repairs injuries. Recent Advances: Vernix caseosa protects the fetus during gestation and facilitates development of the SC in the aqueous uterine environment. The anti-infective, hydrating, acidification, and wound-healing properties post birth provide insights for the development of strategies that facilitate SC maturation and repair in the premature infant. Critical Issues: Reduction of infant mortality is a global health priority. Premature infants have an incompetent skin barrier putting them at risk for irritant exposure, skin compromise and life-threatening infections. Effective interventions to accelerate skin barrier maturation are compelling. Future Directions: Investigations to determine the ontogeny of barrier maturation, that is, SC structure, composition, cohesiveness, permeability, susceptibility to injury, and microflora, as a function of gestational age are essential. Clinicians need to know when the premature skin barrier becomes fully competent and comparable to healthy newborn skin. This will guide the development of innovative strategies for optimizing skin barrier development.
Collapse
Affiliation(s)
- Marty Visscher
- Skin Sciences Program, Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Surgery, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Vivek Narendran
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
42
|
Hieronymus T, Zenke M, Baek JH, Seré K. The clash of Langerhans cell homeostasis in skin: Should I stay or should I go? Semin Cell Dev Biol 2014; 41:30-8. [PMID: 24613914 DOI: 10.1016/j.semcdb.2014.02.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 02/14/2014] [Accepted: 02/19/2014] [Indexed: 12/26/2022]
Abstract
Langerhans cells (LC), the skin epidermal contingent of dendritic cells (DC), possess an exceptional life cycle and developmental origin. LC, like all mature blood cells, develop from haematopoietic stem cells (HSC) through successive steps of lineage commitment and differentiation. However, LC development is different to that of other DC subsets and not yet fully understood. Haematopoietic cell fate decisions are instructed by specific growth factors and cytokines produced in specialized microenvironments or niches. Upon ligand binding the cognate surface receptors on HSC and further restricted progenitor cells regulate the signalling pathways that eventually leads to the execution of lineage-determining genetic programs. In this review we focus on a specific set of surface receptor kinases that have been identified as critical regulators of LC development using genetically modified mice. Recent studies suggest for some of these kinases to impact on LC/LC progenitor interaction with the local niche by regulating adhesion and/or migration. During embryonic development, in wound healing and aberrantly in tumour invasion the same kinase receptors control a genetic program known as epithelial-to-mesenchymal-transition (EMT). We will discuss how EMT and its reverse program of mesenchymal-to-epithelial-transition (MET) can serve as universal concepts operating also in LC development.
Collapse
Affiliation(s)
- Thomas Hieronymus
- Institute for Biomedical Engineering, Department of Cell Biology, Medical Faculty, RWTH Aachen University, Aachen, Germany; Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany.
| | - Martin Zenke
- Institute for Biomedical Engineering, Department of Cell Biology, Medical Faculty, RWTH Aachen University, Aachen, Germany; Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Jea-Hyun Baek
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kristin Seré
- Institute for Biomedical Engineering, Department of Cell Biology, Medical Faculty, RWTH Aachen University, Aachen, Germany; Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
43
|
Yasmin N, Bauer T, Modak M, Wagner K, Schuster C, Köffel R, Seyerl M, Stöckl J, Elbe-Bürger A, Graf D, Strobl H. Identification of bone morphogenetic protein 7 (BMP7) as an instructive factor for human epidermal Langerhans cell differentiation. ACTA ACUST UNITED AC 2013; 210:2597-610. [PMID: 24190429 PMCID: PMC3832935 DOI: 10.1084/jem.20130275] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Bone morphogenetic protein 7 (BMP7) promotes the differentiation of Langerhans cells in the epidermis during prenatal development. Human Langerhans cell (LC) precursors populate the epidermis early during prenatal development and thereafter undergo massive proliferation. The prototypic antiproliferative cytokine TGF-β1 is required for LC differentiation from human CD34+ hematopoietic progenitor cells and blood monocytes in vitro. Similarly, TGF-β1 deficiency results in LC loss in vivo. However, immunohistology studies revealed that human LC niches in early prenatal epidermis and adult basal (germinal) keratinocyte layers lack detectable TGF-β1. Here we demonstrated that these LC niches express high levels of bone morphogenetic protein 7 (BMP7) and that Bmp7-deficient mice exhibit substantially diminished LC numbers, with the remaining cells appearing less dendritic. BMP7 induces LC differentiation and proliferation by activating the BMP type-I receptor ALK3 in the absence of canonical TGF-β1–ALK5 signaling. Conversely, TGF-β1–induced in vitro LC differentiation is mediated via ALK3; however, co-induction of ALK5 diminished TGF-β1–driven LC generation. Therefore, selective ALK3 signaling by BMP7 promotes high LC yields. Within epidermis, BMP7 shows an inverse expression pattern relative to TGF-β1, the latter induced in suprabasal layers and up-regulated in outer layers. We observed that TGF-β1 inhibits microbial activation of BMP7-generated LCs. Therefore, TGF-β1 in suprabasal/outer epidermal layers might inhibit LC activation, resulting in LC network maintenance.
Collapse
Affiliation(s)
- Nighat Yasmin
- Institute of Pathophysiology and Immunology, Center for Molecular Medicine and 2 Center for Medical Research, Medical University Graz, A-8036 Graz, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Schuster C, Gläser R, Fiala C, Eppel W, Harder J, Schröder JM, Elbe-Bürger A. Prenatal human skin expresses the antimicrobial peptide RNase 7. Arch Dermatol Res 2013; 305:545-9. [PMID: 23545750 PMCID: PMC3717166 DOI: 10.1007/s00403-013-1340-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 02/28/2013] [Accepted: 03/18/2013] [Indexed: 11/30/2022]
Abstract
Antimicrobial peptides and proteins (AMPs) play important roles in skin immune defense due to their capacity to inhibit growth of microbes. During intrauterine life, the skin immune system has to acquire the prerequisites to protect the newborn from infection in the hostile environment after birth, which includes the production of skin AMPs. The aim of this study was to analyze the expression of RNase 7, HBD-2/3 and psoriasin during human skin development, thus, providing a deeper insight about the maturity of a fundamental component of the innate immune system. We found low RNase 7 expression levels in the periderm but no expression of HBD-2/3 and psoriasin in first trimester human skin using immunohistochemistry. At the end of the second trimester, RNase 7 is expressed weakly in all epidermal layers with a marked signal in the stratum corneum. HBD-3 and psoriasin are focally expressed while HBD-2 is not detectable. Analysis of supernatants from cultured prenatal skin cells showed that in contrast to adult control, RNase 7 and psoriasin are not found in prenatal skin, suggesting that AMPs are detectable but are not secreted. This study shows the differential expression of AMPs in developing, non-perturbed human prenatal skin. It is conceivable that the combined expression of RNase 7, HBD-3 and psoriasin in fetal skin constitutes a developmental program to exert a broad spectrum of antimicrobial activity to maintain sterility in the amniotic cavity.
Collapse
Affiliation(s)
- Christopher Schuster
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Laboratory of Cellular and Molecular Immunobiology of the Skin, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
It is unclear how the Langerhans cell (LC) network is maintained in adult epidermis. In this issue of Immunity, Seré et al. (2012) show that LCs are replenished in two waves. Monocyte-derived, short-lived LCs come first. A second wave follows, and these LCs of nonmonocytic origin are long-lived.
Collapse
Affiliation(s)
- Nikolaus Romani
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria.
| | | | | |
Collapse
|
46
|
Seré K, Baek JH, Ober-Blöbaum J, Müller-Newen G, Tacke F, Yokota Y, Zenke M, Hieronymus T. Two distinct types of Langerhans cells populate the skin during steady state and inflammation. Immunity 2013; 37:905-16. [PMID: 23159228 DOI: 10.1016/j.immuni.2012.07.019] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 07/30/2012] [Indexed: 11/20/2022]
Abstract
Langerhans cells (LCs), the dendritic cells (DCs) in skin epidermis, possess an exceptional life cycle and developmental origin. Here we identified two types of LCs, short-term and long-term LCs, which transiently or stably reconstitute the LC compartment, respectively. Short-term LCs developed from Gr-1(hi) monocytes under inflammatory conditions and occurred independently of the transcription factor Id2. Long-term LCs arose from bone marrow in steady state and were critically dependent on Id2. Surface marker and gene expression analysis positioned short-term LCs close to Gr-1(hi) monocytes, which is indicative of their monocytic origin. We also show that LC reconstitution after UV light exposure occurs in two waves: an initial fast and transient wave of Gr-1(hi) monocyte-derived short-term LCs is followed by a second wave of steady-state precursor-derived long-term LCs. Our data demonstrate the presence of two types of LCs that develop through different pathways in inflammation and steady state.
Collapse
Affiliation(s)
- Kristin Seré
- Institute for Biomedical Engineering, Department of Cell Biology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Maturing of the fetal and neonatal immune system. Clin Immunol 2013. [DOI: 10.1016/b978-0-7234-3691-1.00050-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
49
|
Bauer T, Zagórska A, Jurkin J, Yasmin N, Köffel R, Richter S, Gesslbauer B, Lemke G, Strobl H. Identification of Axl as a downstream effector of TGF-β1 during Langerhans cell differentiation and epidermal homeostasis. ACTA ACUST UNITED AC 2012; 209:2033-47. [PMID: 23071254 PMCID: PMC3478937 DOI: 10.1084/jem.20120493] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Transforming growth factor-β1 (TGF-β1) is a fundamental regulator of immune cell development and function. In this study, we investigated the effects of TGF-β1 on the differentiation of human Langerhans cells (LCs) and identified Axl as a key TGF-β1 effector. Axl belongs to the TAM (Tyro3, Axl, and Mer) receptor tyrosine kinase family, whose members function as inhibitors of innate inflammatory responses in dendritic cells and are essential to the prevention of lupus-like autoimmunity. We found that Axl expression is induced by TGF-β1 during LC differentiation and that LC precursors acquire Axl early during differentiation. We also describe prominent steady-state expression as well as inflammation-induced activation of Axl in human epidermal keratinocytes and LCs. TGF-β1-induced Axl enhances apoptotic cell (AC) uptake and blocks proinflammatory cytokine production. The antiinflammatory role of Axl in the skin is reflected in a marked impairment of the LC network preceding spontaneous skin inflammation in mutant mice that lack all three TAM receptors. Our findings highlight the importance of constitutive Axl expression to tolerogenic barrier immunity in the epidermis and define a mechanism by which TGF-β1 enables silent homeostatic clearing of ACs to maintain long-term self-tolerance.
Collapse
Affiliation(s)
- Thomas Bauer
- Institute of Immunology, Center of Pathophysiology, Infectiology, and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Iram N, Mildner M, Prior M, Petzelbauer P, Fiala C, Hacker S, Schöppl A, Tschachler E, Elbe-Bürger A. Age-related changes in expression and function of Toll-like receptors in human skin. Development 2012; 139:4210-9. [PMID: 23034637 PMCID: PMC3912866 DOI: 10.1242/dev.083477] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Toll-like receptors (TLRs) initiate innate immune responses and direct subsequent adaptive immunity. They play a major role in cutaneous host defense against micro-organisms and in the pathophysiology of several inflammatory skin diseases. To understand the role of TLRs in the acquisition of immunological competence, we conducted a comprehensive study to evaluate TLR expression and function in the developing human skin before and after birth and compared it with adults. We found that prenatal skin already expresses the same spectrum of TLRs as adult skin. Strikingly, many TLRs were significantly higher expressed in prenatal (TLRs 1-5) and infant and child (TLRs 1 and 3) skin than in adult skin. Surprisingly, neither dendritic cell precursors in prenatal skin nor epidermal Langerhans cells and dermal dendritic cells in adult skin expressed TLRs 3 and 6, whereas the staining pattern and intensity of both TLRs in fetal basal keratinocytes was almost comparable to those of adults. Stimulation of primary human keratinocytes from fetal, neonatal and adult donors with selected TLR agonists revealed that the synthetic TLR3 ligand poly (I:C) specifically, mimicking viral double-stranded RNA, induced a significantly enhanced secretion of CXCL8/IL8, CXCL10/IP-10 and TNFα in fetal and neonatal keratinocytes compared with adult keratinocytes. This study demonstrates quantitative age-specific modifications in TLR expression and innate skin immune reactivity in response to TLR activation. Thus, antiviral innate immunity already in prenatal skin may contribute to protect the developing human body from viral infections in utero in a scenario where the adaptive immune system is not yet fully functional.
Collapse
Affiliation(s)
- Nousheen Iram
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Laboratory of Cellular and Molecular Immunobiology of the Skin, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|