1
|
Schmitter-Sánchez AD, Park S. Immune-Epithelial Cell Interactions during Epidermal Regeneration, Repair, and Inflammatory Diseases. Int J Stem Cells 2025; 18:1-11. [PMID: 38191522 PMCID: PMC11867906 DOI: 10.15283/ijsc23107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/09/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
The multiple layers of the skin cover and protect our entire body. Among the skin layers, the epidermis is in direct contact with the outer environment and serves as the first line of defense. The epidermis functions as a physical and immunological barrier. To maintain barrier function, the epidermis continually regenerates and repairs itself when injured. Interactions between tissue-resident immune cells and epithelial cells are essential to sustain epidermal regeneration and repair. In this review, we will dissect the crosstalk between epithelial cells and specific immune cell populations located in the epidermis during homeostasis and wound repair. In addition, we will analyze the contribution of dysregulated immune-epithelial interactions in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Axel D. Schmitter-Sánchez
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA
- Cell and Molecular Biology Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Sangbum Park
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA
- Division of Dermatology, Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
2
|
Wang H, Yu W, Wang T, Fang D, Wang Z, Wang Y. Therapeutic potential and pharmacological insights of total glucosides of paeony in dermatologic diseases: a comprehensive review. Front Pharmacol 2025; 15:1423717. [PMID: 39822741 PMCID: PMC11735457 DOI: 10.3389/fphar.2024.1423717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 11/27/2024] [Indexed: 01/19/2025] Open
Abstract
Total glucosides of paeony (TGP) are a group of monoterpenes extracted from Paeonia lactiflora Pall., primarily including metabolites such as paeoniflorin and oxypaeoniflorin. Modern pharmacological studies have shown that TGP possesses a variety of biological effects, including immunomodulatory, anti-inflammatory, hepatoprotective, nephroprotective, antidepressant, and cell proliferation regulatory activities. In recent years, clinical research has demonstrated favorable therapeutic effects of TGP on disorders of the liver, cardiovascular, nervous, endocrine, and skeletal systems. Particularly in dermatological treatments, TGP has been found to significantly improve clinical symptoms and shorten the course of the disease. However, there are still certain limitations in the scientific rigor of existing studies and in its clinical application. To assess the potential of TGP in treating dermatologic diseases, this article provides a review of its botanical sources, preparation and extraction processes, quality control, and major chemical metabolites, as well as its pharmacological research and clinical applications in dermatology. Additionally, the mechanisms of action, research gaps, and future directions for TGP in the treatment of dermatologic diseases are discussed, offering valuable guidance for future clinical research on TGP in dermatology.
Collapse
Affiliation(s)
- Huige Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wenchao Yu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tong Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Dianwei Fang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zeyun Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yuanhong Wang
- Department of Dermatology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
3
|
Hovav A, Wilensky A. The role of the epithelial sentinels, Langerhans cells and γδT cells, in oral squamous cell carcinoma. Periodontol 2000 2024; 96:221-228. [PMID: 38273461 PMCID: PMC11579810 DOI: 10.1111/prd.12544] [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: 08/22/2023] [Revised: 11/11/2023] [Accepted: 11/20/2023] [Indexed: 01/27/2024]
Abstract
Oral squamous cell carcinoma (OSCC) arises in the oral epithelium, a tissue in which immune surveillance is mediated by its primary resident leukocytes, Langerhans cells (LCs), and γδT cells. Under steady-state conditions, LCs and γδT cells play a critical role in maintaining oral mucosal homeostasis. As antigen-presenting cells of stratified epithelia, LCs respond to various challenges faced by the epithelium, orchestrating innate, and adaptive immune responses in order to resolve them. γδT cells also sense diverse epithelial insults and react rapidly through cytokine production and cytolytic activity. These epithelial sentinels are also considered to be the first leukocytes in the oral epithelium to encounter early carcinogenic events that have the potential of becoming OSCC. As evident in many malignancies, leukocyte populations help prevent cancer development although they also promote tumor progression. OSCC is no exception, as studies have reported both anti- and pro-tumor roles of LCs and γδT cells. In this review, we summarize the ontogeny of LCs and γδT cells in the oral epithelium and discuss their role in OSCC.
Collapse
Affiliation(s)
- Avi‐Hai Hovav
- Institute of Biomedical and Oral Research, Faculty of Dental MedicineHebrew UniversityJerusalemIsrael
| | - Asaf Wilensky
- Department of Periodontology, Hadassah Medical Center, Faculty of Dental MedicineHebrew University of JerusalemJerusalemIsrael
| |
Collapse
|
4
|
Shah SA, Oakes RS, Jewell CM. Advancing immunotherapy using biomaterials to control tissue, cellular, and molecular level immune signaling in skin. Adv Drug Deliv Rev 2024; 209:115315. [PMID: 38670230 PMCID: PMC11111363 DOI: 10.1016/j.addr.2024.115315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024]
Abstract
Immunotherapies have been transformative in many areas, including cancer treatments, allergies, and autoimmune diseases. However, significant challenges persist in extending the reach of these technologies to new indications and patients. Some of the major hurdles include narrow applicability to patient groups, transient efficacy, high cost burdens, poor immunogenicity, and side effects or off-target toxicity that results from lack of disease-specificity and inefficient delivery. Thus, there is a significant need for strategies that control immune responses generated by immunotherapies while targeting infection, cancer, allergy, and autoimmunity. Being the outermost barrier of the body and the first line of host defense, the skin presents a unique immunological interface to achieve these goals. The skin contains a high concentration of specialized immune cells, such as antigen-presenting cells and tissue-resident memory T cells. These cells feature diverse and potent combinations of immune receptors, providing access to cellular and molecular level control to modulate immune responses. Thus, skin provides accessible tissue, cellular, and molecular level controls that can be harnessed to improve immunotherapies. Biomaterial platforms - microneedles, nano- and micro-particles, scaffolds, and other technologies - are uniquely capable of modulating the specialized immunological niche in skin by targeting these distinct biological levels of control. This review highlights recent pre-clinical and clinical advances in biomaterial-based approaches to target and modulate immune signaling in the skin at the tissue, cellular, and molecular levels for immunotherapeutic applications. We begin by discussing skin cytoarchitecture and resident immune cells to establish the biological rationale for skin-targeting immunotherapies. This is followed by a critical presentation of biomaterial-based pre-clinical and clinical studies aimed at controlling the immune response in the skin for immunotherapy and therapeutic vaccine applications in cancer, allergy, and autoimmunity.
Collapse
Affiliation(s)
- Shrey A Shah
- Fischell Department of Bioengineering, University of Maryland, College Park, 8278 Paint Branch Drive, College Park, MD 20742, USA
| | - Robert S Oakes
- Fischell Department of Bioengineering, University of Maryland, College Park, 8278 Paint Branch Drive, College Park, MD 20742, USA; Department of Veterans Affairs, VA Maryland Health Care System, 10. N Green Street, Baltimore, MD 21201, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, 8278 Paint Branch Drive, College Park, MD 20742, USA; Department of Veterans Affairs, VA Maryland Health Care System, 10. N Green Street, Baltimore, MD 21201, USA; Robert E. Fischell Institute for Biomedical Devices, 8278 Paint Branch Drive, College Park, MD 20742, USA; Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD, 21201, USA; Marlene and Stewart Greenebaum Cancer Center, 22 S. Greene Street, Suite N9E17, Baltimore, MD, 21201, USA.
| |
Collapse
|
5
|
Clausen BE, Amon L, Backer RA, Berod L, Bopp T, Brand A, Burgdorf S, Chen L, Da M, Distler U, Dress RJ, Dudziak D, Dutertre CA, Eich C, Gabele A, Geiger M, Ginhoux F, Giusiano L, Godoy GJ, Hamouda AEI, Hatscher L, Heger L, Heidkamp GF, Hernandez LC, Jacobi L, Kaszubowski T, Kong WT, Lehmann CHK, López-López T, Mahnke K, Nitsche D, Renkawitz J, Reza RA, Sáez PJ, Schlautmann L, Schmitt MT, Seichter A, Sielaff M, Sparwasser T, Stoitzner P, Tchitashvili G, Tenzer S, Tochoedo NR, Vurnek D, Zink F, Hieronymus T. Guidelines for mouse and human DC functional assays. Eur J Immunol 2023; 53:e2249925. [PMID: 36563126 DOI: 10.1002/eji.202249925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022]
Abstract
This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various non-lymphoid tissues. Recent studies have provided evidence for an increasing number of phenotypically distinct conventional DC (cDC) subsets that on one hand exhibit a certain functional plasticity, but on the other hand are characterized by their tissue- and context-dependent functional specialization. Here, we describe a selection of assays for the functional characterization of mouse and human cDC. The first two protocols illustrate analysis of cDC endocytosis and metabolism, followed by guidelines for transcriptomic and proteomic characterization of cDC populations. Then, a larger group of assays describes the characterization of cDC migration in vitro, ex vivo, and in vivo. The final guidelines measure cDC inflammasome and antigen (cross)-presentation activity. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.
Collapse
Affiliation(s)
- Björn E Clausen
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Ronald A Backer
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Luciana Berod
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Tobias Bopp
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Anna Brand
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sven Burgdorf
- Laboratory of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Luxia Chen
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Meihong Da
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Ute Distler
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Regine J Dress
- Institute of Systems Immunology, Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Germany
| | - Charles-Antoine Dutertre
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Christina Eich
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Anna Gabele
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Melanie Geiger
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Lucila Giusiano
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Gloria J Godoy
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Ahmed E I Hamouda
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Lukas Hatscher
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Gordon F Heidkamp
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Lola C Hernandez
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas Jacobi
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Tomasz Kaszubowski
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Wan Ting Kong
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Germany
| | - Tamara López-López
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karsten Mahnke
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Dominik Nitsche
- Laboratory of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Jörg Renkawitz
- Biomedical Center (BMC), Walter Brendel Center of Experimental Medicine, Institute of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, LMU Munich, Munich, Germany
| | - Rifat A Reza
- Biomedical Center (BMC), Walter Brendel Center of Experimental Medicine, Institute of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, LMU Munich, Munich, Germany
| | - Pablo J Sáez
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Schlautmann
- Laboratory of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Madeleine T Schmitt
- Biomedical Center (BMC), Walter Brendel Center of Experimental Medicine, Institute of Cardiovascular Physiology and Pathophysiology, Klinikum der Universität, LMU Munich, Munich, Germany
| | - Anna Seichter
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Malte Sielaff
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
| | - Tim Sparwasser
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Patrizia Stoitzner
- Department of Dermatology, Venerology & Allergology, Medical University Innsbruck, Innsbruck, Austria
| | - Giorgi Tchitashvili
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Stefan Tenzer
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Institute of Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes-Gutenberg University Mainz, Mainz, Germany
- Helmholtz Institute for Translational Oncology Mainz (HI-TRON Mainz), Mainz, Germany
| | - Nounagnon R Tochoedo
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Damir Vurnek
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Germany
| | - Fabian Zink
- Laboratory of Cellular Immunology, LIMES Institute, University of Bonn, Bonn, Germany
| | - Thomas Hieronymus
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
- Institute of Cell and Tumor Biology, RWTH Aachen University, Medical Faculty, Germany
| |
Collapse
|
6
|
Taufalele PV, Wang W, Simmons AJ, Southard-Smith AN, Chen B, Greenlee JD, King MR, Lau KS, Hassane DC, Bordeleau F, Reinhart-King CA. Matrix stiffness enhances cancer-macrophage interactions and M2-like macrophage accumulation in the breast tumor microenvironment. Acta Biomater 2023; 163:365-377. [PMID: 35483629 PMCID: PMC9592676 DOI: 10.1016/j.actbio.2022.04.031] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/17/2022] [Accepted: 04/20/2022] [Indexed: 02/07/2023]
Abstract
The role of intratumor heterogeneity is becoming increasingly apparent in part due to expansion in single cell technologies. Clinically, tumor heterogeneity poses several obstacles to effective cancer therapy dealing with biomarker variability and treatment responses. Matrix stiffening is known to occur during tumor progression and contribute to pathogenesis in several cancer hallmarks, including tumor angiogenesis and metastasis. However, the effects of matrix stiffening on intratumor heterogeneity have not been thoroughly studied. In this study, we applied single-cell RNA sequencing to investigate the differences in the transcriptional landscapes between stiff and compliant MMTV-PyMT mouse mammary tumors. We found similar compositions of cancer and stromal subpopulations in compliant and stiff tumors but differential intercellular communication and a significantly higher concentration of tumor-promoting, M2-like macrophages in the stiffer tumor microenvironments. Interestingly, we found that cancer cells seeded on stiffer substrates recruited more macrophages. Furthermore, elevated matrix stiffness increased Colony Stimulating Factor 1 (CSF-1) expression in breast cancer cells and reduction of CSF-1 expression on stiffer substrates reduced macrophage recruitment. Thus, our results demonstrate that tissue phenotypes were conserved between stiff and compliant tumors but matrix stiffening altered cell-cell interactions which may be responsible for shifting the phenotypic balance of macrophages residing in the tumor microenvironment towards a pro-tumor progression M2 phenotype. STATEMENT OF SIGNIFICANCE: Cells within tumors are highly heterogeneous, posing challenges with treatment and recurrence. While increased tissue stiffness can promote several hallmarks of cancer, its effects on tumor heterogeneity are unclear. We used single-cell RNA sequencing to investigate the differences in the transcriptional landscapes between stiff and compliant MMTV-PyMT mouse mammary tumors. We found similar compositions of cancer and stromal subpopulations in compliant and stiff tumors but differential intercellular communication and a significantly higher concentration of tumor-promoting, M2-like macrophages in the stiffer tumor microenvironments. Using a biomaterial-based platform, we found that cancer cells seeded on stiffer substrates recruited more macrophages, supporting our in vivo findings. Together, our results demonstrate a key role of matrix stiffness in affecting cell-cell communication and macrophage recruitment.
Collapse
Affiliation(s)
- Paul V Taufalele
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Alan J Simmons
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Austin N Southard-Smith
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bob Chen
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA; Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Joshua D Greenlee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ken S Lau
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA; Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Duane C Hassane
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - François Bordeleau
- Cancer Research Center and Centre de Recherche du CHU de Québec, Université Laval, Canada
| | | |
Collapse
|
7
|
Gao Y, Wang Z, Cui Y, Xu M, Weng L. Emerging Strategies of Engineering and Tracking Dendritic Cells for Cancer Immunotherapy. ACS APPLIED BIO MATERIALS 2023; 6:24-43. [PMID: 36520013 DOI: 10.1021/acsabm.2c00790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs), a kind of specialized immune cells, play key roles in antitumor immune response and promotion of innate and adaptive immune responses. Recently, many strategies have been developed to utilize DCs in cancer therapy, such as delivering antigens and adjuvants to DCs and using scaffold to recruit and activate DCs. Here we outline how different DC subsets influence antitumor immunity, summarize the FDA-approved vaccines and cancer vaccines under clinical trials, discuss the strategies for engineering DCs and noninvasive tracking of DCs to improve antitumor immunotherapy, and reveal the potential of artificial neural networks for the design of DC based vaccines.
Collapse
Affiliation(s)
- Yu Gao
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Zhixuan Wang
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Ying Cui
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Miaomiao Xu
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Lixing Weng
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.,School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| |
Collapse
|
8
|
Chakrabarti S, Hoque M, Jamil NZ, Singh VJ, Pollacksmith D, Meer N, Pezzano MT. Bone Marrow-Derived Cells Contribute to the Maintenance of Thymic Stroma including TECs. J Immunol Res 2022; 2022:6061746. [PMID: 35528618 PMCID: PMC9076333 DOI: 10.1155/2022/6061746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/24/2022] [Indexed: 12/29/2022] Open
Abstract
In paradox to critical functions for T-cell selection and self-tolerance, the thymus undergoes profound age-associated atrophy and loss of T-cell function, further enhanced by cancer therapies. Identifying thymic epithelial progenitor populations capable of forming functional thymic tissue will be critical in understanding thymic epithelial cell (TEC) ontogeny and designing strategies to reverse involution. We identified a new population of progenitor cells, present in both the thymus and bone marrow (BM) of mice, that coexpress the hematopoietic marker CD45 and the definitive thymic epithelial marker EpCAM and maintain the capacity to form functional thymic tissue. Confocal analysis and qRT-PCR of sorted cells from both BM and thymus confirmed coexpression of CD45 and EpCAM. Grafting of C57BL/6 fetal thymi under the kidney capsule of H2BGFP transgenic mice revealed that peripheral CD45+ EpCAM+ GFP-expressing cells migrate into the developing thymus and contribute to both TECs and FSP1-expressing thymic stroma. Sorted BM-derived CD45+ EpCAM+ cells contribute to reaggregate thymic organ cultures (RTOCs) and differentiate into keratin and FoxN1-expressing TECs, demonstrating that BM cells can contribute to the maintenance of TEC microenvironments previously thought to be derived solely from endoderm. BM-derived CD45+ EpCAM+ cells represent a new source of progenitor cells that contribute to thymic homeostasis. Future studies will characterize the contribution of BM-derived CD45+ EpCAM+ TEC progenitors to distinct functional TEC microenvironments in both the steady-state thymus and under conditions of demand. Cell therapies utilizing this population may help counteract thymic involution in cancer patients.
Collapse
Affiliation(s)
- Shami Chakrabarti
- Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
| | - Mohammed Hoque
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
| | - Nawshin Zara Jamil
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
| | - Varan J. Singh
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
| | - Daniel Pollacksmith
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
| | - Neelab Meer
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
| | - Mark T. Pezzano
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
- Program in Biology, The Graduate Center of the City University of New York, New York, NY 10016, USA
| |
Collapse
|
9
|
Park S. Building vs. Rebuilding Epidermis: Comparison Embryonic Development and Adult Wound Repair. Front Cell Dev Biol 2022; 9:796080. [PMID: 35145968 PMCID: PMC8822150 DOI: 10.3389/fcell.2021.796080] [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: 10/15/2021] [Accepted: 12/31/2021] [Indexed: 01/05/2023] Open
Abstract
Wound repair is essential to restore tissue function through the rebuilding of pre-existing structures. The repair process involves the re-formation of tissue, which was originally generated by embryonic development, with as similar a structure as possible. Therefore, these two processes share many similarities in terms of creating tissue architecture. However, fundamental differences still exist, such as differences in the cellular components, the status of neighboring tissues, and the surrounding environment. Recent advances in single-cell transcriptomics, in vivo lineage tracing, and intravital imaging revealed subpopulations, long-term cell fates, and dynamic cellular behaviors in live animals that were not detectable previously. This review highlights similarities and differences between adult wound repair and embryonic tissue development with a particular emphasis on the epidermis of the skin.
Collapse
Affiliation(s)
- Sangbum Park
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, United States
- Division of Dermatology, Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, United States
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
10
|
Alcántara-Hernández M, Idoyaga J. Mass cytometry profiling of human dendritic cells in blood and tissues. Nat Protoc 2021; 16:4855-4877. [PMID: 34480131 PMCID: PMC10538357 DOI: 10.1038/s41596-021-00599-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 06/29/2021] [Indexed: 12/15/2022]
Abstract
The immune system comprises distinct functionally specialized cell populations, which can be characterized in depth by mass cytometry protein profiling. Unfortunately, the low-throughput nature of mass cytometry has made it challenging to analyze minor cell populations. This is the case for dendritic cells, which represent 0.2-2% of all immune cells in tissues and yet perform the critical task of initiating and modulating immune responses. Here, we provide an optimized step-by-step protocol for the characterization of well-known and emerging human dendritic cell populations in blood and tissues using mass cytometry. We provide detailed instructions for the generation of single-cell suspensions, sample enrichment, staining, acquisition and data analysis. We also include a barcoding option that reduces acquisition variability and allows the analysis of low numbers of dendritic cells, i.e., ~20,000. In contrast to other protocols, we emphasize the use of negative selection approaches to enrich for minor populations of immune cells while avoiding their activation. The entire procedure can be completed in 2-3 d and can be conveniently paused at several stages. The procedure described in this robust and reliable protocol allows the analysis of human dendritic cells in health and disease and during vaccination.
Collapse
Affiliation(s)
- Marcela Alcántara-Hernández
- Microbiology & Immunology Department and Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Juliana Idoyaga
- Microbiology & Immunology Department and Immunology Program, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
11
|
Ghilas S, Ambrosini M, Cancel JC, Brousse C, Massé M, Lelouard H, Dalod M, Crozat K. Natural killer cells and dendritic epidermal γδ T cells orchestrate type 1 conventional DC spatiotemporal repositioning toward CD8 + T cells. iScience 2021; 24:103059. [PMID: 34568787 PMCID: PMC8449251 DOI: 10.1016/j.isci.2021.103059] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/14/2021] [Accepted: 08/25/2021] [Indexed: 02/03/2023] Open
Abstract
Successful immune responses rely on a regulated delivery of the right signals to the right cells at the right time. Here we show that natural killer (NK) and dendritic epidermal γδ T cells (DETCs) use similar mechanisms to spatiotemporally orchestrate conventional type 1 dendritic cell (cDC1) functions in the spleen, skin, and its draining lymph nodes (dLNs). Upon MCMV infection in the spleen, cDC1 clusterize with activated NK cells in marginal zones. This XCR1-dependent repositioning of cDC1 toward NK cells allows contact delivery of IL-12 and IL-15/IL-15Rα by cDC1, which is critical for NK cell responses. NK cells deliver granulocyte-macrophage colony-stimulating factor (GM-CSF) to cDC1, guiding their CCR7-dependent relocalization into the T cell zone. In MCMV-infected skin, XCL1-secreting DETCs promote cDC1 migration from the skin to the dLNs. This XCR1-dependent licensing of cDC1 both in the spleen and skin accelerates antiviral CD8+ T cell responses, revealing an additional mechanism through which cDC1 bridge innate and adaptive immunity. Upon viral infection in the spleen, NK cells clusterize with cDC1 in the marginal zone This XCL1/XCR1-dependent interaction allows mutual delivery of activating signals NK cell GM-CSF directs cDC1 migration to T cell zone boosting CD8+ T cell priming In the skin, DETCs contact cDC1 via XCL1/XCR1 to promote antiviral CD8+ T cell priming
Collapse
Affiliation(s)
- Sonia Ghilas
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Marc Ambrosini
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Jean-Charles Cancel
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Carine Brousse
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Marion Massé
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Hugues Lelouard
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Marc Dalod
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Karine Crozat
- Aix Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| |
Collapse
|
12
|
Mo L, Luo X, Yang G, Liu J, Yang L, Liu Z, Wang S, Liu D, Liu Z, Yang P. Epithelial cell-derived CD83 restores immune tolerance in the airway mucosa by inducing regulatory T-cell differentiation. Immunology 2021; 163:310-322. [PMID: 33539546 PMCID: PMC8207377 DOI: 10.1111/imm.13317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/11/2022] Open
Abstract
The mechanism of generation of regulatory T cells (Treg) remains incompletely understood. Recent studies show that CD83 has immune regulatory functions. This study aims to investigate the role of epithelial cell-derived CD83 in the restoration of immune tolerance in the airway mucosa by inducing the Treg differentiation. In this study, CD83 and ovalbumin (OVA)-carrying exosomes were generated from airway epithelial cells. An airway allergy mouse model was developed to test the role of CD83/OVA-carrying exosomes in the suppression of airway allergy by inducing Treg generation. We observed that mouse airway epithelial cells expressed CD83 that could be up-regulated by CD40 ligand. The CD83 deficiency in epithelial cells retarded the Treg generation in the airway mucosa. CD83 up-regulated transforming growth factor-β-inducible early gene 1 expression in CD4+ T cells to promote Foxp3 expression. Exposure of primed CD4+ T cells to CD83/OVA-carrying exosomes promoted antigen-specific Treg generation. Administration of CD83/OVA-carrying exosomes inhibited experimental airway allergic response. In summary, airway epithelial cells express CD83 that is required in the Treg differentiation in the airway mucosa. Administration of CD83/OVA-carrying exosomes can inhibit airway allergy that has the translation potential in the treatment of airway allergic disorders.
Collapse
Affiliation(s)
- Li‐Hua Mo
- Research Center of Allergy & ImmunologyShenzhen University School of MedicineShenzhenChina
| | - Xiang‐Qian Luo
- Department of Pediatric OtolaryngologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Gui Yang
- Department of OtolaryngologyLonggang Central HospitalShenzhenChina
| | - Jiang‐Qi Liu
- Longgang ENT Hospital & Shenzhen ENT InstituteShenzhenChina
| | - Li‐Teng Yang
- Department of Respirology & AllergyThird Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Zhi‐Qiang Liu
- Longgang ENT Hospital & Shenzhen ENT InstituteShenzhenChina
| | - Shuai Wang
- Longgang ENT Hospital & Shenzhen ENT InstituteShenzhenChina
| | - Da‐Bo Liu
- Department of Pediatric OtolaryngologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Zhi‐Gang Liu
- Research Center of Allergy & ImmunologyShenzhen University School of MedicineShenzhenChina
| | - Ping‐Chang Yang
- Research Center of Allergy & ImmunologyShenzhen University School of MedicineShenzhenChina
- Guangdong Provincial Key Laboratory of Regional Immunity and DiseasesShenzhenChina
| |
Collapse
|
13
|
Polak ME, Singh H. Tolerogenic and immunogenic states of Langerhans cells are orchestrated by epidermal signals acting on a core maturation gene module. Bioessays 2021; 43:e2000182. [PMID: 33645739 DOI: 10.1002/bies.202000182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 12/11/2022]
Abstract
Langerhans cells (LCs), residing in the epidermis, are able to induce potent immunogenic responses and also to mediate immune tolerance. We propose that tolerogenic and immunogenic responses of LCs are directed by signaling from the epidermis and involve counter-acting gene circuits that are coupled to a core maturation gene module. We base our analysis on recent genetic and genomic findings facilitating the understanding of the molecular mechanisms controlling these divergent immune functions. Comparing gene regulatory network (GRN) analyses of various types of dendritic cells (DCs) including LCs we integrate signaling-dependent (TGFβ, EpCAM, β-Catenin) and transcription factor (IRF4, IRF1, NFκB) regulated gene circuits that appear to orchestrate the distinctive LC functional states. Our model proposes, that while epidermal signaling in the steady-state promotes LC tolerogenic function, the disruption of cell-cell contacts coupled with inflammatory signaling induces LC immunogenic programing. The conceptual framework emphasizes the sensing of discrete epidermal and inflammatory cues by resident LCs in dictating their genomic programing and cell state dynamics.
Collapse
Affiliation(s)
- Marta E Polak
- Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, UK.,Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Harinder Singh
- Center for Systems Immunology, Departments of Immunology and Computational and Systems Biology, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
14
|
Novel Concepts: Langerhans Cells in the Tumour Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1273:147-158. [PMID: 33119880 DOI: 10.1007/978-3-030-49270-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Langerhans cells (LCs) are immune cells that reside in the stratified epithelium of the skin and mucosal membranes. They play a range of roles in the skin, including antigen presentation and maintenance of peripheral tolerance. Reports of LC numbers have been variable in different cancer types, with the majority of studies indicating a reduction in their number. Changes in the cytokine profile and other secreted molecules, downregulation of surface molecules on cells and hypoxia all contribute to the regulation of LCs in the tumour microenvironment. Functionally, LCs have been reported to regulate immunity and carcinogenesis in different cancer types. An improved understanding of the function and biology of LCs in tumours is essential knowledge that underpins the development of new cancer immunotherapies.
Collapse
|
15
|
Selective AhR knockout in langerin-expressing cells abates Langerhans cells and polarizes Th2/Tr1 in epicutaneous protein sensitization. Proc Natl Acad Sci U S A 2020; 117:12980-12990. [PMID: 32461368 DOI: 10.1073/pnas.1917479117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) represents an environmental sensor regulating immune responses. In the skin, AhR is expressed in several cell types, including keratinocytes, epidermal Langerhans cells (LC), and dermal dendritic cells (DC). The mechanisms how AhR activates or inhibits cutaneous immune responses remain controversial, owing to differences in the cell-specific functions of AhR and the different activating ligands. Therefore, we sought to investigate the role of AhR in LC and langerin+ and negative DC in the skin. To this aim, we generated Langerin-specific and CD11c-specific knockout (-/-) mice lacking AhR, respectively, in LC and Langerin+ dermal DC and in all CD11c+ cells. These were then tested in an epicutaneous protein (ovalbumin, Ova) sensitization model. Immunofluorescence microscopy and flow cytometry revealed that Langerin-AhR-/- but not CD11c-AhR-/- mice harbored a decreased number of LC with fewer and stunted dendrites in the epidermis as well as a decreased number of LC in skin-draining lymph nodes (LN). Moreover, in the absence of AhR, we detected an enhanced T helper type-2 (Th2) [increased interleukin 5 (IL-5) and interleukin 13 (IL-13)] and T regulatory type-1 (Tr1) (IL-10) response when LN cells were challenged with Ova in vitro, though the number of regulatory T cells (Treg) in the LN remained comparable. Langerin-AhR-/- mice also exhibited increased blood levels of Ova-specific immunoglobulin E (IgE). In conclusion, deletion of AhR in langerin-expressing cells diminishes the number and activation of LC, while enhancing Th2 and Tr1 responses upon epicutaneous protein sensitization.
Collapse
|
16
|
Xue D, Tabib T, Morse C, Lafyatis R. Transcriptome landscape of myeloid cells in human skin reveals diversity, rare populations and putative DC progenitors. J Dermatol Sci 2019; 97:41-49. [PMID: 31836271 DOI: 10.1016/j.jdermsci.2019.11.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 11/08/2019] [Accepted: 11/25/2019] [Indexed: 01/21/2023]
Abstract
BACKGROUND The heterogeneous functions of dermal myeloid cells in antigen presentation, and scavenging pathogens and cell debris places them centrally in cutaneous inflammation. Single cell transcriptomics can provide new understanding of the heterogeneity and function of yet incompletely understood human dermal myeloid cell subsets. OBJECTIVE Investigate the transcriptome landscape of myeloid cells in healthy human skin. METHODS Single cell RNA-sequencing was performed on skin biopsies from ten healthy donors and analyzed to identify myeloid cell populations. RESULTS One LIN- HLA-DR+ cluster with expression of myeloid-specific genes was identified as a cluster of myeloid cells. Upon reanalysis of this cluster, we identified three macrophage subsets, marked by high expression of CCR1, MARCO or TREM2; and six dendritic cell subsets, marked by high expression of CLEC9A, CXorf21, MCOLN2, LAMP3, KIAA0101 and Langerin, representing respectively cDC1, two subsets of cDC2, a novel DC type, a cluster of proliferating DC, and a Langerhans cell subset. GO term analysis indicated specialized functions for the discrete rare populations of myeloid cells: TREM2 Mφ in lipid metabolism and LAMP3 DC as a mature cDC. Proliferating DCs appeared to represent cDC2 progenitors. CONCLUSION The transcriptional landscape of myeloid cell populations in human skin indicates several, novel populations with specialized functions, as well as a rare proliferating DC population that likely accounts for local regeneration or expansion of dermal DCs. We provide robust gene expression markers for each of these populations that should permit better understandings of their roles in various homeostatic and pathologic immune processes in the skin.
Collapse
Affiliation(s)
- Dan Xue
- Division of Rheumatology and Clinical Rheumatology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tracy Tabib
- Division of Rheumatology and Clinical Rheumatology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christina Morse
- Division of Rheumatology and Clinical Rheumatology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Rheumatology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
17
|
Amon L, Lehmann CHK, Baranska A, Schoen J, Heger L, Dudziak D. Transcriptional control of dendritic cell development and functions. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:55-151. [PMID: 31759434 DOI: 10.1016/bs.ircmb.2019.10.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs) are major regulators of adaptive immunity, as they are not only capable to induce efficient immune responses, but are also crucial to maintain peripheral tolerance and thereby inhibit autoimmune reactions. DCs bridge the innate and the adaptive immune system by presenting peptides of self and foreign antigens as peptide MHC complexes to T cells. These properties render DCs as interesting target cells for immunomodulatory therapies in cancer, but also autoimmune diseases. Several subsets of DCs with special properties and functions have been described. Recent achievements in understanding transcriptional programs on single cell level, together with the generation of new murine models targeting specific DC subsets, advanced our current understanding of DC development and function. Thus, DCs arise from precursor cells in the bone marrow with distinct progenitor cell populations splitting the monocyte populations and macrophage populations from the DC lineage, which upon lineage commitment can be separated into conventional cDC1, cDC2, and plasmacytoid DCs (pDCs). The DC populations harbor intrinsic programs enabling them to react for specific pathogens in dependency on the DC subset, and thereby orchestrate T cell immune responses. Similarities, but also varieties, between human and murine DC subpopulations are challenging, and will require further investigation of human specimens under consideration of the influence of the tissue micromilieu and DC subset localization in the future.
Collapse
Affiliation(s)
- Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Baranska
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Janina Schoen
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
18
|
Sumpter TL, Balmert SC, Kaplan DH. Cutaneous immune responses mediated by dendritic cells and mast cells. JCI Insight 2019; 4:123947. [PMID: 30626752 DOI: 10.1172/jci.insight.123947] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
In the skin, complex cellular networks maintain barrier function and immune homeostasis. Tightly regulated multicellular cascades are required to initiate innate and adaptive immune responses. Innate immune cells, particularly DCs and mast cells, are central to these networks. Early studies evaluated the function of these cells in isolation, but recent studies clearly demonstrate that cutaneous DCs (dermal DCs and Langerhans cells) physically interact with neighboring cells and are receptive to activation signals from surrounding cells, such as mast cells. These interactions amplify immune activation. In this review, we discuss the known functions of cutaneous DC populations and mast cells and recent studies highlighting their roles within cellular networks that determine cutaneous immune responses.
Collapse
Affiliation(s)
| | | | - Daniel H Kaplan
- Department of Dermatology and.,Department of Immunology, University of Pittsburgh School of Medicine,Pittsburgh, Pennsylvania, USA
| |
Collapse
|
19
|
Zinc in Keratinocytes and Langerhans Cells: Relevance to the Epidermal Homeostasis. J Immunol Res 2018; 2018:5404093. [PMID: 30622978 PMCID: PMC6304883 DOI: 10.1155/2018/5404093] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/10/2018] [Indexed: 02/07/2023] Open
Abstract
In the skin, the epidermis is continuously exposed to various kinds of external substances and stimuli. Therefore, epidermal barriers are crucial for providing protection, safeguarding health, and regulating water balance by maintaining skin homeostasis. Disruption of the epidermal barrier allows external substances and stimuli to invade or stimulate the epidermal cells, leading to the elicitation of skin inflammation. The major components of the epidermal barrier are the stratum corneum (SC) and tight junctions (TJs). The presence of zinc in the epidermis promotes epidermal homeostasis; hence, this study reviewed the role of zinc in the formation and function of the SC and TJs. Langerhans cells (LCs) are one of the antigen-presenting cells found in the epidermis. They form TJs with adjacent keratinocytes (KCs), capture external antigens, and induce antigen-specific immune reactions. Thus, the function of zinc in LCs was examined in this review. We also summarized the general knowledge of zinc and zinc transporters in the epidermis with updated findings.
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: 2.6] [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]
|