1
|
Ullm F, Renner A, Freudenberg U, Werner C, Pompe T. The Influence of Sulfation Degree of Glycosaminoglycan-Functionalized 3D Collagen I Networks on Cytokine Profiles of In Vitro Macrophage-Fibroblast Cocultures. Gels 2024; 10:450. [PMID: 39057473 PMCID: PMC11276094 DOI: 10.3390/gels10070450] [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: 06/12/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Cell-cell interactions between fibroblasts and immune cells, like macrophages, are influenced by interaction with the surrounding extracellular matrix during wound healing. In vitro hydrogel models that mimic and modulate these interactions, especially of soluble mediators like cytokines, may allow for a more detailed investigation of immunomodulatory processes. In the present study, a biomimetic extracellular matrix model based on fibrillar 3D collagen I networks with a functionalization with heparin or 6-ON-desulfated heparin, as mimics of naturally occurring heparan sulfate, was developed to modulate cytokine binding effects with the hydrogel matrix. The constitution and microstructure of the collagen I network were found to be stable throughout the 7-day culture period. A coculture study of primary human fibroblasts/myofibroblasts and M-CSF-stimulated macrophages was used to show its applicability to simulate processes of progressed wound healing. The quantification of secreted cytokines (IL-8, IL-10, IL-6, FGF-2) in the cell culture supernatant demonstrated the differential impact of glycosaminoglycan functionalization of the collagen I network. Most prominently, IL-6 and FGF-2 were shown to be regulated by the cell culture condition and network constitution, indicating changes in paracrine and autocrine cell-cell communication of the fibroblast-macrophage coculture. From this perspective, we consider our newly established in vitro hydrogel model suitable for mechanistic coculture analyses of primary human cells to unravel the role of extracellular matrix factors in key events of tissue regeneration and beyond.
Collapse
Affiliation(s)
- Franziska Ullm
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103 Leipzig, Germany; (F.U.); (A.R.)
| | - Alexander Renner
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103 Leipzig, Germany; (F.U.); (A.R.)
| | - Uwe Freudenberg
- Max Bergmann Center of Biomaterials, Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Strasse 6, 01069 Dresden, Germany; (U.F.); (C.W.)
| | - Carsten Werner
- Max Bergmann Center of Biomaterials, Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Strasse 6, 01069 Dresden, Germany; (U.F.); (C.W.)
| | - Tilo Pompe
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103 Leipzig, Germany; (F.U.); (A.R.)
- Max Bergmann Center of Biomaterials, Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Strasse 6, 01069 Dresden, Germany; (U.F.); (C.W.)
| |
Collapse
|
2
|
Morrison AI, Mikula AM, Spiekstra SW, de Kok M, Affandi AJ, Roest HP, van der Laan LJW, de Winde CM, Koning JJ, Gibbs S, Mebius RE. An Organotypic Human Lymph Node Model Reveals the Importance of Fibroblastic Reticular Cells for Dendritic Cell Function. Tissue Eng Regen Med 2024; 21:455-471. [PMID: 38114886 PMCID: PMC10987465 DOI: 10.1007/s13770-023-00609-x] [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: 07/14/2023] [Revised: 10/19/2023] [Accepted: 10/22/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Human lymph node (HuLN) models have emerged with invaluable potential for immunological research and therapeutic application given their fundamental role in human health and disease. While fibroblastic reticular cells (FRCs) are instrumental to HuLN functioning, their inclusion and recognition of importance for organotypic in vitro lymphoid models remain limited. METHODS Here, we established an in vitro three-dimensional (3D) model in a collagen-fibrin hydrogel with primary FRCs and a dendritic cell (DC) cell line (MUTZ-3 DC). To study and characterise the cellular interactions seen in this 3D FRC-DC organotypic model compared to the native HuLN; flow cytometry, immunohistochemistry, immunofluorescence and cytokine/chemokine analysis were performed. RESULTS FRCs were pivotal for survival, proliferation and localisation of MUTZ-3 DCs. Additionally, we found that CD1a expression was absent on MUTZ-3 DCs that developed in the presence of FRCs during cytokine-induced MUTZ-3 DC differentiation, which was also seen with primary monocyte-derived DCs (moDCs). This phenotype resembled HuLN-resident DCs, which we detected in primary HuLNs, and these CD1a- MUTZ-3 DCs induced T cell proliferation within a mixed leukocyte reaction (MLR), indicating a functional DC status. FRCs expressed podoplanin (PDPN), CD90 (Thy-1), CD146 (MCAM) and Gremlin-1, thereby resembling the DC supporting stromal cell subset identified in HuLNs. CONCLUSION This 3D FRC-DC organotypic model highlights the influence and importance of FRCs for DC functioning in a more realistic HuLN microenvironment. As such, this work provides a starting point for the development of an in vitro HuLN.
Collapse
Affiliation(s)
- Andrew I Morrison
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Aleksandra M Mikula
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Sander W Spiekstra
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Michael de Kok
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Alsya J Affandi
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Henk P Roest
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Charlotte M de Winde
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jasper J Koning
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Susan Gibbs
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Department Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands
| | - Reina E Mebius
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Chalise U, Hale TM. Fibroblasts under pressure: cardiac fibroblast responses to hypertension and antihypertensive therapies. Am J Physiol Heart Circ Physiol 2024; 326:H223-H237. [PMID: 37999643 PMCID: PMC11219059 DOI: 10.1152/ajpheart.00401.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
Approximately 50% of Americans have hypertension, which significantly increases the risk of heart failure. In response to increased peripheral resistance in hypertension, intensified mechanical stretch in the myocardium induces cardiomyocyte hypertrophy and fibroblast activation to withstand increased pressure overload. This changes the structure and function of the heart, leading to pathological cardiac remodeling and eventual progression to heart failure. In the presence of hypertensive stimuli, cardiac fibroblasts activate and differentiate to myofibroblast phenotype capable of enhanced extracellular matrix secretion in coordination with other cell types, mainly cardiomyocytes. Both systemic and local renin-angiotensin-aldosterone system activation lead to increased angiotensin II stimulation of fibroblasts. Angiotensin II directly activates fibrotic signaling such as transforming growth factor β/SMAD and mitogen-activated protein kinase (MAPK) signaling to produce extracellular matrix comprised of collagens and matricellular proteins. With the advent of single-cell RNA sequencing techniques, heterogeneity in fibroblast populations has been identified in the left ventricle in models of hypertension and pressure overload. The various clusters of fibroblasts reveal a range of phenotypes and activation states. Select antihypertensive therapies have been shown to be effective in limiting fibrosis, with some having direct actions on cardiac fibroblasts. The present review focuses on the fibroblast-specific changes that occur in response to hypertension and pressure overload, the knowledge gained from single-cell analyses, and the effect of antihypertensive therapies. Understanding the dynamics of hypertensive fibroblast populations and their similarities and differences by sex is crucial for the advent of new targets and personalized medicine.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota, United States
| | - Taben M Hale
- Department of Basic Medical Sciences, University of Arizona, College of Medicine-Phoenix, Phoenix, Arizona, United States
| |
Collapse
|
4
|
Gao D, Fang L, Liu C, Yang M, Yu X, Wang L, Zhang W, Sun C, Zhuang J. Microenvironmental regulation in tumor progression: Interactions between cancer-associated fibroblasts and immune cells. Biomed Pharmacother 2023; 167:115622. [PMID: 37783155 DOI: 10.1016/j.biopha.2023.115622] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/20/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023] Open
Abstract
The tumor microenvironment (TME), the "soil" on which tumor cells grow, has an important role in regulating the proliferation and metastasis of tumor cells as well as their response to treatment. Cancer-associated fibroblasts (CAFs), as the most abundant stromal cells of the TME, can not only directly alter the immunosuppressive effect of the TME through their own metabolism, but also influence the aggregation and function of immune cells by secreting a large number of cytokines and chemokines, reducing the body's immune surveillance of tumor cells and making them more prone to immune escape. Our study provides a comprehensive review of fibroblast chemotaxis, malignant transformation, metabolic characteristics, and interactions with immune cells. In addition, the current small molecule drugs targeting CAFs have been summarized, including both natural small molecules and targeted drugs for current clinical therapeutic applications. A complete review of the role of fibroblasts in TME from an immune perspective is presented, which has important implications in improving the efficiency of immunotherapy by targeting fibroblasts.
Collapse
Affiliation(s)
- Dandan Gao
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Liguang Fang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Mengrui Yang
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Xiaoyun Yu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China
| | - Longyun Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China
| | - Wenfeng Zhang
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao Special Administrative Region of China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang 261000, China; Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| |
Collapse
|
5
|
Shiri Aghbash P, Shirvaliloo M, Khalo Abass Kasho A, Alinezhad F, Nauwynck H, Bannazadeh Baghi H. Cluster of differentiation frequency on antigen presenting-cells: The next step to cervical cancer prognosis? Int Immunopharmacol 2022; 108:108896. [DOI: 10.1016/j.intimp.2022.108896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/16/2022] [Accepted: 05/23/2022] [Indexed: 11/29/2022]
|
6
|
Hu P, Leyton L, Hagood JS, Barker TH. Thy-1-Integrin Interactions in cis and Trans Mediate Distinctive Signaling. Front Cell Dev Biol 2022; 10:928510. [PMID: 35733855 PMCID: PMC9208718 DOI: 10.3389/fcell.2022.928510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/23/2022] [Indexed: 12/04/2022] Open
Abstract
Thy-1 is a cell surface glycosylphosphatidylinositol (GPI)-anchored glycoprotein that bears a broad mosaic of biological roles across various cell types. Thy-1 displays strong physiological and pathological implications in development, cancer, immunity, and tissue fibrosis. Quite uniquely, Thy-1 is capable of mediating integrin-related signaling through direct trans- and cis-interaction with integrins. Both interaction types have shown distinctive roles, even when interacting with the same type of integrin, where binding in trans or in cis often yields divergent signaling events. In this review, we will revisit recent progress and discoveries of Thy-1–integrin interactions in trans and in cis, highlight their pathophysiological consequences and explore other potential binding partners of Thy-1 within the integrin regulation/signaling paradigm.
Collapse
Affiliation(s)
- Ping Hu
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, United States
| | - Lisette Leyton
- Cellular Communication Laboratory, Program of Cellular and Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile and Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - James S. Hagood
- Department of Pediatrics, Division of Pulmonology, School of Medicine, University of North Carolina, Chapel Hill, NC, United States
- Program for Rare and Interstitial Lung Disease, School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Thomas H. Barker
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, United States
- *Correspondence: Thomas H. Barker,
| |
Collapse
|
7
|
Ito S, Hirobe S, Yamashita R, Sugiyama A, Takeuchi H, Eguchi R, Yoshida J, Oyamada T, Tachibana M, Okada N. Analysis of immune response induction mechanisms implicating the dose-sparing effect of transcutaneous immunization using a self-dissolving microneedle patch. Vaccine 2022; 40:862-872. [PMID: 34998604 DOI: 10.1016/j.vaccine.2021.12.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/19/2021] [Accepted: 12/25/2021] [Indexed: 11/28/2022]
Abstract
Transcutaneous immunization (TCI) is an effective vaccination method that is easier and less painful than the conventional injectable vaccination method. We previously developed self-dissolving microneedle patches (sdMN) and demonstrated that this TCI method has a high vaccination efficacy in mice and humans. To elucidate the mechanism of immune response induction, which is the basis for the efficacy and safety of TCI with sdMN, we examined the local reaction of the skin where sdMN was applied and the kinetics and differentiation status of immune cells in the draining lymph nodes (DLNs). We found that gene expression of the proinflammatory cytokine Il1b and the downstream transcription factor Irf7 was markedly upregulated in skin tissues after sdMN application. Moreover, activation of Langerhans cells and CD207- dermal dendritic cells, which are subsets of antigen-presenting cells (APCs) in the skin, and their migration to the DLNs were promoted. Furthermore, the activated APC subsets promoted CD4+ T cell and B cell differentiation and the formation of germinal centers, which are the sites of high-affinity antibody production. These phenomena associated with sdMN application may contribute to the efficient production of antigen-specific antibodies after TCI using sdMN. These findings provide essential information regarding immune response induction mechanisms for the development and improvement of TCI preparations.
Collapse
Affiliation(s)
- Sayami Ito
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Sachiko Hirobe
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Molecular Pharmaceutical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Pharmacy, Osaka University Hospital, 2-15 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryo Yamashita
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Arisa Sugiyama
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Honoka Takeuchi
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryosuke Eguchi
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Junya Yoshida
- FUJIFILM Advanced Research Laboratories, Fujifilm Holdings Corporation, 577-1 Ushijima, Kaisei Town, Ashigarakami-gun, Kanagawa Prefecture 258-8577, Japan
| | - Takayoshi Oyamada
- FUJIFILM Advanced Research Laboratories, Fujifilm Holdings Corporation, 577-1 Ushijima, Kaisei Town, Ashigarakami-gun, Kanagawa Prefecture 258-8577, Japan
| | - Masashi Tachibana
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Vaccine and Immune Regulation (BIKEN), Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Naoki Okada
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Vaccine and Immune Regulation (BIKEN), Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
8
|
Riedl P, Pompe T. Functional label-free assessment of fibroblast differentiation in 3D collagen-I-matrices using particle image velocimetry. Biomater Sci 2021; 9:5917-5927. [PMID: 34291253 DOI: 10.1039/d1bm00638j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Fibroblasts are a diverse population of connective tissue cells that are a key component in physiological wound healing. Myofibroblasts are differentiated fibroblasts occurring in various physiological and pathological conditions, like in the healing of wounds or in the tumour microenvironment. They exhibit important functions compared to fibroblasts in terms of proliferation, protein secretion, and contractility. The gold standard to distinguish myofibroblasts is alpha-smooth muscle actin (αSMA) expression and its incorporation in stress fibres, which is only revealed by gene expression analysis and immunostaining. Here, we introduce an approach to functionally determine the myofibroblast status of live fibroblasts directly in in vitro cell culture by analysing their ability to contract the extracellular matrix around them without the need for labelling. It is based on particle image velocimetry algorithms applied to dynamic deformations of the extracellular matrix network structure imaged by phase contrast microscopy. Advanced image analysis allows us to distinguish between various differentiation stages of fibroblasts including the dynamic change over several days. We further apply machine learning classification to automatically evaluate different cell culture conditions. With this new method, we provide a versatile tool to functionally evaluate the dynamic process of fibroblast differentiation. It can be applied for in vitro screening studies in biomimetic 3D cell cultures with options to extend it to other cell systems with contractile phenotypes.
Collapse
Affiliation(s)
- Philipp Riedl
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, 04103 Leipzig, Germany.
| | | |
Collapse
|
9
|
Hirobe S, Susai R, Takeuchi H, Eguchi R, Ito S, Quan YS, Kamiyama F, Okada N. Characteristics of immune induction by transcutaneous vaccination using dissolving microneedle patches in mice. Int J Pharm 2021; 601:120563. [PMID: 33811967 DOI: 10.1016/j.ijpharm.2021.120563] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 12/23/2022]
Abstract
Transcutaneous immunization (TCI) is an appealing vaccination method. Compared with conventional injectable immunization, TCI is easier and less painful. We previously developed a dissolving microneedle (MN) patch and demonstrated that TCI using MN patches demonstrates high vaccination efficacy without adverse events in humans. In this study, we investigated the immune induction mechanism of TCI using our MN patch, focusing on inflammatory responses in the skin and on the dynamics, activation, and differentiation of various immunocompetent cells in draining lymph nodes (dLNs). We demonstrate that inflammatory cytokines such as IL-6 and TNF-α increased in the skin at an early stage after MN patch application, inducing the infiltration of macrophages and neutrophils and promoting the activation and migration of skin-resident antigen-presenting cells (Langerhans and Langerin- dermal dendritic cells) to dLNs. Moreover, the activated antigen-presenting cells reaching the dLNs enhanced the differentiation of T (Teff, Tem, and Tcm) and B (plasma and memory) cells. This may contribute to the efficient antigen-specific antibody production induced by TCI using MN patches. We believe that our findings reveal a part of the immune induction mechanism by TCI and provide useful information for the development and improvement of TCI formulations based on the immune induction mechanism.
Collapse
Affiliation(s)
- Sachiko Hirobe
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Molecular Pharmaceutical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Pharmacy, Osaka University Hospital, 2-15 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Ryo Susai
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Honoka Takeuchi
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryosuke Eguchi
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Sayami Ito
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ying-Shu Quan
- CosMED Pharmaceutical Co. Ltd., 32 Higashikujokawanishi-cho, Minami-ku, Kyoto 601-8014, Japan
| | - Fumio Kamiyama
- CosMED Pharmaceutical Co. Ltd., 32 Higashikujokawanishi-cho, Minami-ku, Kyoto 601-8014, Japan
| | - Naoki Okada
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Laboratory of Vaccine and Immune Regulation (BIKEN), Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
10
|
Montes-Gómez AE, García-Cordero J, Marcial-Juárez E, Shrivastava G, Visoso-Carvajal G, Juárez-Delgado FJ, Flores-Romo L, Sanchez-Torres MC, Santos-Argumedo L, Bustos-Arriaga J, Cedillo-Barrón L. Crosstalk Between Dermal Fibroblasts and Dendritic Cells During Dengue Virus Infection. Front Immunol 2020; 11:538240. [PMID: 33193307 PMCID: PMC7645109 DOI: 10.3389/fimmu.2020.538240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 09/01/2020] [Indexed: 12/29/2022] Open
Abstract
Dengue virus infection (DENV-2) is transmitted by infected mosquitoes via the skin, where many dermal and epidermal cells are potentially susceptible to infection. Most of the cells in an area of infection will establish an antiviral microenvironment to control viral replication. Although cumulative studies report permissive DENV-2 infection in dendritic cells, keratinocytes, and fibroblasts, among other cells also infected, little information is available regarding cell-to-cell crosstalk and the effect of this on the outcome of the infection. Therefore, our study focused on understanding the contribution of fibroblast and dendritic cell crosstalk to the control or promotion of dengue. Our results suggest that dendritic cells promote an antiviral state over fibroblasts by enhancing the production of type I interferon, but not proinflammatory cytokines. Infected and non-infected fibroblasts promoted partial dendritic cell maturation, and the fibroblast-matured cells were less permissive to infection and showed enhanced type I interferon production. We also observed that the soluble mediators produced by non-infected or Poly (I:C) transfected fibroblasts induced allogenic T cell proliferation, but mediators produced by DENV-2 infected fibroblasts inhibited this phenomenon. Additionally, the effects of fibroblast soluble mediators on CD14+ monocytes were analyzed to assess whether they affected the differentiation of monocyte derived dendritic cells (moDC). Our data showed that mediators produced by infected fibroblasts induced variable levels of monocyte differentiation into dendritic cells, even in the presence of recombinant GM-CSF and IL-4. Cells with dendritic cell-like morphology appeared in the culture; however, flow cytometry analysis showed that the mediators did not fully downregulate CD14 nor did they upregulate CD1a. Our data revealed that fibroblast-dendritic cell crosstalk promoted an antiviral response mediated manly by type I interferons over fibroblasts. Furthermore, the maturation of dendritic cells and T cell proliferation were promoted, which was inhibited by DENV-2-induced mediators. Together, our results suggest that activation of the adaptive immune response is influenced by the crosstalk of skin resident cells and the intensity of innate immune responses established in the microenvironment of the infected skin.
Collapse
Affiliation(s)
- Alfredo E. Montes-Gómez
- Departamento de Biomedicina Molecular Centro de Investigación y de Estudios Avanzados, del Instituto Politécnico Nacional, Ciudad de México, México
| | - Julio García-Cordero
- Departamento de Biomedicina Molecular Centro de Investigación y de Estudios Avanzados, del Instituto Politécnico Nacional, Ciudad de México, México
| | - Edith Marcial-Juárez
- Departamento de Biología Celular Centro de Investigación y de Estudios Avanzados, del Instituto Politécnico Nacional, Ciudad de México, México
| | - Gaurav Shrivastava
- Departamento de Biomedicina Molecular Centro de Investigación y de Estudios Avanzados, del Instituto Politécnico Nacional, Ciudad de México, México
| | - Giovani Visoso-Carvajal
- Departamento de Biomedicina Molecular Centro de Investigación y de Estudios Avanzados, del Instituto Politécnico Nacional, Ciudad de México, México
| | | | - Leopoldo Flores-Romo
- Departamento de Biología Celular Centro de Investigación y de Estudios Avanzados, del Instituto Politécnico Nacional, Ciudad de México, México
| | - Ma. Carmen Sanchez-Torres
- Departamento de Biomedicina Molecular Centro de Investigación y de Estudios Avanzados, del Instituto Politécnico Nacional, Ciudad de México, México
| | - Leopoldo Santos-Argumedo
- Departamento de Biomedicina Molecular Centro de Investigación y de Estudios Avanzados, del Instituto Politécnico Nacional, Ciudad de México, México
| | - José Bustos-Arriaga
- Unidad de Biomedicina, Laboratorio de Biología Molecular e Inmunología de arbovirus, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, México
| | - Leticia Cedillo-Barrón
- Departamento de Biomedicina Molecular Centro de Investigación y de Estudios Avanzados, del Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
11
|
Sapudom J, Müller CD, Nguyen KT, Martin S, Anderegg U, Pompe T. Matrix Remodeling and Hyaluronan Production by Myofibroblasts and Cancer-Associated Fibroblasts in 3D Collagen Matrices. Gels 2020; 6:E33. [PMID: 33008082 PMCID: PMC7709683 DOI: 10.3390/gels6040033] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/14/2020] [Accepted: 09/27/2020] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment is a key modulator in cancer progression and has become a novel target in cancer therapy. An increase in hyaluronan (HA) accumulation and metabolism can be found in advancing tumor progression and are often associated with aggressive malignancy, drug resistance and poor prognosis. Wound-healing related myofibroblasts or activated cancer-associated fibroblasts (CAF) are assumed to be the major sources of HA. Both cell types are capable to synthesize new matrix components as well as reorganize the extracellular matrix. However, to which extent myofibroblasts and CAF perform these actions are still unclear. In this work, we investigated the matrix remodeling and HA production potential in normal human dermal fibroblasts (NHFB) and CAF in the absence and presence of transforming growth factor beta -1 (TGF-β1), with TGF-β1 being a major factor of regulating fibroblast differentiation. Three-dimensional (3D) collagen matrix was utilized to mimic the extracellular matrix of the tumor microenvironment. We found that CAF appeared to response insensitively towards TGF-β1 in terms of cell proliferation and matrix remodeling when compared to NHFB. In regards of HA production, we found that both cell types were capable to produce matrix bound HA, rather than a soluble counterpart, in response to TGF-β1. However, activated CAF demonstrated higher HA production when compared to myofibroblasts. The average molecular weight of produced HA was found in the range of 480 kDa for both cells. By analyzing gene expression of HA metabolizing enzymes, namely hyaluronan synthase (HAS1-3) and hyaluronidase (HYAL1-3) isoforms, we found expression of specific isoforms in dependence of TGF-β1 present in both cells. In addition, HAS2 and HYAL1 are highly expressed in CAF, which might contribute to a higher production and degradation of HA in CAF matrix. Overall, our results suggested a distinct behavior of NHFB and CAF in 3D collagen matrices in the presence of TGF-β1 in terms of matrix remodeling and HA production pointing to a specific impact on tumor modulation.
Collapse
Affiliation(s)
- Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, UAE
- Institute of Biochemistry, Faculty of Life Sciences, Universität Leipzig, 04103 Leipzig, Germany; (C.D.M.); (S.M.); (T.P.)
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Universität Leipzig, 04103 Leipzig, Germany; (K.-T.N.); (U.A.)
| | - Claudia Damaris Müller
- Institute of Biochemistry, Faculty of Life Sciences, Universität Leipzig, 04103 Leipzig, Germany; (C.D.M.); (S.M.); (T.P.)
| | - Khiet-Tam Nguyen
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Universität Leipzig, 04103 Leipzig, Germany; (K.-T.N.); (U.A.)
| | - Steve Martin
- Institute of Biochemistry, Faculty of Life Sciences, Universität Leipzig, 04103 Leipzig, Germany; (C.D.M.); (S.M.); (T.P.)
| | - Ulf Anderegg
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Universität Leipzig, 04103 Leipzig, Germany; (K.-T.N.); (U.A.)
| | - Tilo Pompe
- Institute of Biochemistry, Faculty of Life Sciences, Universität Leipzig, 04103 Leipzig, Germany; (C.D.M.); (S.M.); (T.P.)
| |
Collapse
|
12
|
Hesse-Macabata J, Morgner B, Elsner P, Hipler UC, Wiegand C. Tryptanthrin promotes keratinocyte and fibroblast responses in vitro after infection with Trichophyton benhamiae DSM6916. Sci Rep 2020; 10:1863. [PMID: 32024909 PMCID: PMC7002663 DOI: 10.1038/s41598-020-58773-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 12/31/2019] [Indexed: 12/31/2022] Open
Abstract
Exceedingly virulent pathogens and growing antimicrobial resistances require new therapeutic approaches. The zoophilic dermatophyte Trichophyton benhamiae causes highly inflammatory, cutaneous fungal infections. Recently, it could be shown that the plant-derived alkaloid tryptanthrin (TRP) exhibits strong anti-microbial activities against yeasts and dermatophytes. The aim of this study was to analyse the bioactivity of TRP under infectious conditions using an in-vitro dermatophytosis model employing fibroblasts and keratinocytes infected with T. benhamiae DSM6916. Analyses comprised determination of cell viability, effects on the innate immune response including expression and secretion of pro-inflammatory cytokines/chemokines as well as expression of various antimicrobial peptides (AMP), toll-like receptor (TLR) 2 and proliferation marker MKI67. T. benhamiae caused severe inflammation in the cutaneous cell models. TRP almost fully prevented T. benhamiae-derived damage of dermal fibroblasts and substantially reduced it in epidermal keratinocytes. A distinct down-regulation of the expression and secretion of pro-inflammatory cytokines was observed. Further, TRP promoted AMP expression, especially of HBD2 and HBD3, in keratinocytes even without fungal presence. This study provides crucial evidence that TRP is not only a strong antifungal agent but also potentially modulates the innate immune response. This makes it interesting as a natural antimycotic drug for adjuvant treatment and prevention of fungal re-infection.
Collapse
Affiliation(s)
| | - Bianka Morgner
- Department of Dermatology, Jena University Hospital, Jena, Germany
| | - Peter Elsner
- Department of Dermatology, Jena University Hospital, Jena, Germany
| | | | - Cornelia Wiegand
- Department of Dermatology, Jena University Hospital, Jena, Germany.
| |
Collapse
|
13
|
Thélu A, Catoire S, Kerdine-Römer S. Immune-competent in vitro co-culture models as an approach for skin sensitisation assessment. Toxicol In Vitro 2020; 62:104691. [DOI: 10.1016/j.tiv.2019.104691] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/05/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022]
|
14
|
Ullm F, Riedl P, Machado de Amorim A, Patzschke A, Weiß R, Hauschildt S, Franke K, Anderegg U, Pompe T. 3D Scaffold-Based Macrophage Fibroblast Coculture Model Reveals IL-10 Dependence of Wound Resolution Phase. ACTA ACUST UNITED AC 2019; 4:e1900220. [PMID: 32293120 DOI: 10.1002/adbi.201900220] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/08/2019] [Indexed: 12/18/2022]
Abstract
Persistent inflammation and impaired repair in dermal wound healing are frequently associated with cell-cell and cell-matrix miscommunication. A direct coculture model of primary human myofibroblasts (MyoFB) and M-CSF-differentiated macrophages (M-Mɸ) in fibrillar three-dimensional Collagen I (Coll I) matrices is developed to study intercellular interactions. The coculture experiments reveal the number of M-Mɸ regulated MyoFB dedifferentiation in a dose-dependent manner. The amount of MyoFB decreases in dependence of the number of cocultured M-Mɸ, even in the presence of MyoFB-inducing transforming growth factor β1 (TGF-β1 ). Gene expression analysis of matrix proteins (collagen I, collagen III, ED-A-fibronectin) confirms the results of an altered MyoFB phenotype. Additionally, M-Mɸ is shown to be the main source of secreted cytokine interleukin-10 (IL-10), which is suggested to affect MyoFB dedifferentiation. These findings indicate a paracrine impact of IL-10 secretion by M-Mɸ on the MyoFB differentiation status counteracting the TGF-β1 -driven MyoFB activation. Hence, the in vitro coculture model simulates physiological situations during wound resolution and underlines the importance of paracrine IL-10 signals by M-Mɸ. In sum, the 3D Coll I-based matrices with a MyoFB-M-Mɸ coculture form a highly relevant biomimetic model of late stages of wound healing.
Collapse
Affiliation(s)
- Franziska Ullm
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, 04103, Leipzig, Germany
| | - Philipp Riedl
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, 04103, Leipzig, Germany
| | | | - Aline Patzschke
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, 04103, Leipzig, Germany
| | - Ronald Weiß
- Institute of Clinical Immunology, Medical Faculty, Leipzig University, 04103, Leipzig, Germany
| | - Sunna Hauschildt
- Institute of Biology, Faculty of Life Sciences, Leipzig University, 04103, Leipzig, Germany
| | - Katja Franke
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, 04103, Leipzig, Germany
| | - Ulf Anderegg
- Department of Dermatology, Venerology and Allergology, Medical Faculty, Leipzig University Leipzig, 04103, Leipzig, Germany
| | - Tilo Pompe
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, 04103, Leipzig, Germany
| |
Collapse
|
15
|
Thönes S, Rother S, Wippold T, Blaszkiewicz J, Balamurugan K, Moeller S, Ruiz-Gómez G, Schnabelrauch M, Scharnweber D, Saalbach A, Rademann J, Pisabarro MT, Hintze V, Anderegg U. Hyaluronan/collagen hydrogels containing sulfated hyaluronan improve wound healing by sustained release of heparin-binding EGF-like growth factor. Acta Biomater 2019; 86:135-147. [PMID: 30660005 DOI: 10.1016/j.actbio.2019.01.029] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/08/2019] [Accepted: 01/14/2019] [Indexed: 12/21/2022]
Abstract
Functional biomaterials that are able to bind, stabilize and release bioactive proteins in a defined manner are required for the controlled delivery of such to the desired place of action, stimulating wound healing in health-compromised patients. Glycosaminoglycans (GAG) represent a very promising group of components since they may be functionally engineered and are well tolerated by the recipient tissues due to their relative immunological inertness. Ligands of the Epidermal Growth Factor (EGF) receptor (EGFR) activate keratinocytes and dermal fibroblasts and, thus, contribute to skin wound healing. Heparin-binding EGF-like growth factor (HB-EGF) bound to GAG in biomaterials (e.g. hydrogels) might serve as a reservoir that induces prolonged activation of the EGF receptor and to recover disturbed wound healing. Based on previous findings, the capacity of hyaluronan (HA) and its sulfated derivatives (sHA) to bind and release HB-EGF from HA/collagen-based hydrogels was investigated. Docking and molecular dynamics analysis of a molecular model of HB-EGF led to the identification of residues in the heparin-binding domain of the protein being essential for the recognition of GAG derivatives. Furthermore, molecular modeling and surface plasmon resonance (SPR) analyses demonstrated that sulfation of HA increases binding strength to HB-EGF thus providing a rationale for the development of sHA-containing hydrogels. In line with computational observations and in agreement with SPR results, gels containing sHA displayed a retarded HB-EGF release in vitro compared to pure HA/collagen gels. Hydrogels containing HA and collagen or a mixture with sHA were shown to bind and release bioactive HB-EGF over at least 72 h, which induced keratinocyte migration, EGFR-signaling and HGF expression in dermal fibroblasts. Importantly, hydrogels containing sHA strongly increased the effectivity of HB-EGF in inducing epithelial tip growth in epithelial wounds shown in a porcine skin organ culture model. These findings suggest that hydrogels containing HA and sHA can be engineered for smart and effective wound dressings. STATEMENT OF SIGNIFICANCE: Immobilization and sustained release of recombinant proteins from functional biomaterials might overcome the limited success of direct application of non-protected solute growth factors during the treatment of impaired wound healing. We developed HA/collagen-based hydrogels supplemented with acrylated sulfated HA for binding and release of HB-EGF. We analyzed the molecular basis of HB-EGF interaction with HA and its chemical derivatives by in silico modeling and surface plasmon resonance. These hydrogels bind HB-EGF reversibly. Using different in vitro assays and organ culture we demonstrate that the introduction of sulfated HA into the hydrogels significantly increases the effectivity of HB-EGF action on target cells. Therefore, sulfated HA-containing hydrogels are promising functional biomaterials for the development of mediator releasing wound dressings.
Collapse
|
16
|
Hesse-Macabata J, Morgner B, Morgenstern S, Grimm MO, Elsner P, Hipler UC, Wiegand C. Innate immune response of human epidermal keratinocytes and dermal fibroblasts to in vitro incubation of Trichophyton benhamiae DSM 6916. J Eur Acad Dermatol Venereol 2019; 33:1177-1188. [PMID: 30720896 DOI: 10.1111/jdv.15472] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/14/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Superficial cutaneous infection caused by the zoophilic dermatophyte Trichophyton benhamiae is often associated with a highly inflammatory immune response. As non-professional immune cells, epidermal keratinocytes and dermal fibroblasts contribute to the first line of defence by producing pro-inflammatory cytokines and antimicrobial peptides (AMP). OBJECTIVE Purpose of this study was to gain a deeper understanding of the pathogenesis and the fungal-host interaction as not much is known about the innate immune response of these cutaneous cells against T. benhamiae. METHODS Using a dermatophytosis model of fibroblasts and keratinocytes incubated with T. benhamiae DSM 6916, analyses included determination of cell viability and cytotoxicity, effects on the innate immune response including expression and secretion of pro-inflammatory cytokines/chemokines and expression of AMP, as well as alterations of genes involved in cell adhesion. RESULTS Trichophyton benhamiae DSM 6916 infection led to severe cell damage and direct induction of a broad spectrum of pro-inflammatory cytokines and chemokines in both cutaneous cells. Only keratinocytes differentially up-regulated AMP genes expression after T. benhamiae DSM 6916 infection. Expression of AMPs in fibroblasts was not inducible by fungal infection, whereas their absences potentially contributed to a continuous increase in the fungal biomass on fibroblasts, which in turn was reduced in keratinocytes possibly due to the antimicrobial actions of induced AMPs. On mRNA level, T. benhamiae DSM 6916 infection altered cell-cell contact proteins in keratinocytes, indicating that targeting specific cell-cell adhesion proteins might be part of dermatophytes' virulence strategy. CONCLUSION This study showed that in addition to immune cells, keratinocytes and fibroblasts could participate in antimicrobial defence against an exemplary infection with T. benhamiae DSM 6916.
Collapse
Affiliation(s)
- J Hesse-Macabata
- Department of Dermatology, Jena University Hospital, Jena, Germany
| | - B Morgner
- Department of Dermatology, Jena University Hospital, Jena, Germany
| | - S Morgenstern
- Department of Urology, Jena University Hospital, Jena, Germany
| | - M O Grimm
- Department of Urology, Jena University Hospital, Jena, Germany
| | - P Elsner
- Department of Dermatology, Jena University Hospital, Jena, Germany
| | - U C Hipler
- Department of Dermatology, Jena University Hospital, Jena, Germany
| | - C Wiegand
- Department of Dermatology, Jena University Hospital, Jena, Germany
| |
Collapse
|
17
|
Mainzer C, Packard T, Bordes S, Closs B, Greene WC, Elias PM, Uchida Y. Tissue microenvironment initiates an immune response to structural components of Staphylococcus aureus. Exp Dermatol 2019; 28:161-168. [PMID: 30566255 PMCID: PMC6706075 DOI: 10.1111/exd.13864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/13/2018] [Accepted: 12/13/2018] [Indexed: 01/10/2023]
Abstract
Cell-to-cell communication in skin participates to the maintenance of homeostatic responses to foreign substances. Certain strains of Staphylococcus (S) aureus are vicious pathogens that cause deleterious effects in host cells and tissues. Both secreted toxins and structural components of S. aureus trigger an immune response, though how S. aureus stimulates host immune responses is poorly understood. We explored here how keratinocytes and fibroblasts initiate the first steps of an immune response by activating dendritic cells (DCs) through recognition of structural components of S. aureus. We treated monocyte-derived Langerhans cells (moLCs) and monocyte-derived DCs (moDCs) with conditioned media from keratinocytes (K-CM) and fibroblasts (F-CM) treated with heat-killed S. aureus (HKSA) respectively, or directly with HKSA. Immune and inflammatory responses from keratinocytes, fibroblasts, moLCs and moDCs were assessed by analysis of cell surface markers and cytokine production using flow cytometry, real-time PCR and ELISA assays. K-CM and F-CM increased the expression of CD86 and HLA-DR on moLCs and moDCs, in association with a specific cytokine profile. K-CM upregulated TNFA, IL-1B and GM-CSF mRNA expression in moLCs, while F-CM upregulated IL-12 and downregulated TNFA and TGFB mRNA expression in moDCs. Additionally, F-CM attenuated the induction of an inflammatory profile in monocytes. The recognition of structural components from S. aureus by cutaneous microenvironment induces the activation and the expression of specific cytokines from LCs and DCs.
Collapse
Affiliation(s)
- Carine Mainzer
- Department of Dermatology, School of Medicine, University of California San Francisco, San Francisco, California
- SILAB Inc., Hazlet, New Jersey
| | - Thomas Packard
- Gladstone Institute of Virology and Immunology, University of California San Francisco, San Francisco, California
| | | | | | - Warner C. Greene
- Gladstone Institute of Virology and Immunology, University of California San Francisco, San Francisco, California
| | - Peter M. Elias
- Department of Dermatology, School of Medicine, University of California San Francisco, San Francisco, California
| | - Yoshikazu Uchida
- Department of Dermatology, School of Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
18
|
Walch-Rückheim B, Ströder R, Theobald L, Pahne-Zeppenfeld J, Hegde S, Kim YJ, Bohle RM, Juhasz-Böss I, Solomayer EF, Smola S. Cervical Cancer-Instructed Stromal Fibroblasts Enhance IL23 Expression in Dendritic Cells to Support Expansion of Th17 Cells. Cancer Res 2019; 79:1573-1586. [PMID: 30696656 DOI: 10.1158/0008-5472.can-18-1913] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 11/16/2018] [Accepted: 01/23/2019] [Indexed: 11/16/2022]
Abstract
Persistent infection with high-risk human papillomavirus (HPV) is a prerequisite for the development of cervical cancer. HPV-transformed cells actively instruct their microenvironment, promoting chronic inflammation and cancer progression. We previously demonstrated that cervical cancer cells contribute to Th17 cell recruitment, a cell type with protumorigenic properties. In this study, we analyzed the expression of the Th17-promoting cytokine IL23 in the cervical cancer micromilieu and found CD83+ mature dendritic cells (mDC) coexpressing IL23 in the stroma of cervical squamous cell carcinomas in situ. This expression of IL23 correlated with stromal Th17 cells, advanced tumor stage, lymph node metastasis, and cervical cancer recurrence. Cocultures of cervical cancer-instructed mDCs and cervical fibroblasts led to potent protumorigenic expansion of Th17 cells in vitro but failed to induce antitumor Th1 differentiation. Correspondingly, cervical cancer-instructed fibroblasts increased IL23 production in cocultured cervical cancer-instructed mDCs, which mediated subsequent Th17 cell expansion. In contrast, production of the Th1-polarizing cytokine IL12 in the cancer-instructed mDCs was strongly reduced. This differential IL23 and IL12 regulation was the consequence of an increased expression of the IL23 subunits IL23p19 and IL12p40 but decreased expression of the IL12 subunit IL12p35 in cervical cancer-instructed mDCs. Cervical cancer cell-derived IL6 directly suppressed IL12p35 in mDCs but indirectly induced IL23 expression in fibroblast-primed mDCs via CAAT/enhancer-binding protein β (C/EBPβ)-dependent induction of IL1β. In summary, our study defines a mechanism by which the cervical cancer micromilieu supports IL23-mediated Th17 expansion associated with cancer progression. SIGNIFICANCE: Cervical cancer cells differentially regulate IL23 and IL12 in DC fibroblast cocultures in an IL6/C/EBPβ/IL1β-dependent manner, thereby supporting the expansion of Th17 cells during cancer progression.
Collapse
Affiliation(s)
- Barbara Walch-Rückheim
- Institute of Virology and Center of Human and Molecular Biology, Saarland University, Homburg/Saar, Germany.
| | - Russalina Ströder
- Department of Obstetrics and Gynecology, Saarland University, Homburg/Saar, Germany
| | - Laura Theobald
- Institute of Virology and Center of Human and Molecular Biology, Saarland University, Homburg/Saar, Germany
| | - Jennifer Pahne-Zeppenfeld
- Center for Molecular Medicine Cologne and Institute of Virology, University of Cologne, Cologne, Germany
| | - Subramanya Hegde
- Center for Molecular Medicine Cologne and Institute of Virology, University of Cologne, Cologne, Germany
| | - Yoo-Jin Kim
- Institute of Pathology, Saarland University, Homburg/Saar, Germany
| | | | - Ingolf Juhasz-Böss
- Department of Obstetrics and Gynecology, Saarland University, Homburg/Saar, Germany
| | | | - Sigrun Smola
- Institute of Virology and Center of Human and Molecular Biology, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
19
|
Sapudom J, Wu X, Chkolnikov M, Ansorge M, Anderegg U, Pompe T. Fibroblast fate regulation by time dependent TGF-β1 and IL-10 stimulation in biomimetic 3D matrices. Biomater Sci 2018; 5:1858-1867. [PMID: 28676875 DOI: 10.1039/c7bm00286f] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The presentation of TGF-β1 during the early stage of wound healing is a prerequisite for extracellular matrix (ECM) synthesis and remodeling by activated fibroblasts, called myofibroblasts. At later stages, clearance of myofibroblasts is needed to avoid overshooting ECM production. Apoptosis of myofibroblasts and the macrophage-released anti-inflammatory cytokine IL-10 are controversially discussed as regulating cues in this context. To reveal the regulating cues, defined biomaterial scaffolds are needed to conduct in-depth in vitro studies in a physiologically relevant context. In this work, we used an in vitro biomimetic wound healing model. It consists of a 3D fibrillar matrix from collagen I and fibronectin and different temporal stimuli by TGF-β1 and IL-10. Human dermal fibroblast behavior was investigated in terms of myofibroblast differentiation (αSMA expression), matrix remodeling, proliferation and migration in the permanent or sequential presence of TGF-β1 and IL-10 over 4 days. We could show that removal of TGF-β1 after initial stimulation resulted in an increase of apoptosis of myofibroblasts. In contrast, TGF-β1 stimulation followed by IL-10 treatment did not result in increased cell apoptosis but instead led to a significant increase of cell motility and reduction of myofibroblasts. The findings suggest that myofibroblasts are a transiently "activated" fibroblastic phenotype and can be de-differentiated to fibroblasts in the presence of IL-10. Overall, our 3D ECM model allows mimicking the early and late stages of wound healing and highlights the temporal sequence of TGF-β1 and IL-10 as an important cue for completion of tissue formation and maintenance of tissue homeostasis.
Collapse
Affiliation(s)
- Jiranuwat Sapudom
- Institute of Biochemistry, Faculty of Biosciences, Pharmacy and Psychology, Universität Leipzig, Leipzig 04103, Germany.
| | | | | | | | | | | |
Collapse
|
20
|
Sagi Z, Hieronymus T. The Impact of the Epithelial-Mesenchymal Transition Regulator Hepatocyte Growth Factor Receptor/Met on Skin Immunity by Modulating Langerhans Cell Migration. Front Immunol 2018; 9:517. [PMID: 29616031 PMCID: PMC5864859 DOI: 10.3389/fimmu.2018.00517] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/27/2018] [Indexed: 01/16/2023] Open
Abstract
Langerhans cells (LCs), the epidermal dendritic cell (DC) subset, express the transmembrane tyrosine kinase receptor Met also known as hepatocyte growth factor (HGF) receptor. HGF is the exclusive ligand of Met and upon binding executes mitogenic, morphogenic, and motogenic activities to various cells. HGF exerts anti-inflammatory activities via Met signaling and was found to regulate various functions of immune cells, including differentiation and maturation, cytokine production, cellular migration and adhesion, and T cell effector function. It has only recently become evident that a number of HGF-regulated functions in inflammatory processes and immune responses are imparted via DCs. However, the mechanisms by which Met signaling in DCs conveys its immunoregulatory effects have not yet been fully understood. In this review, we focus on the current knowledge of Met signaling in DCs with particular attention on the morphogenic and motogenic activities. Met signaling was shown to promote DC mobility by regulating matrix metalloproteinase activities and adhesion. This is a striking resemblance to the role of Met in regulating a cell fate program during embryonic development, wound healing, and in tumor invasion known as epithelial–mesenchymal transition (EMT). Hence, we propose the concept that an EMT program is executed by Met signaling in LCs.
Collapse
Affiliation(s)
- Zsofia Sagi
- Department of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany.,Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Thomas Hieronymus
- Department of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany.,Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
21
|
Buechler MB, Turley SJ. A short field guide to fibroblast function in immunity. Semin Immunol 2017; 35:48-58. [PMID: 29198601 DOI: 10.1016/j.smim.2017.11.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/06/2017] [Indexed: 12/27/2022]
Abstract
Fibroblasts in secondary lymphoid organs, or fibroblastic reticular cells (FRC), are gate-keepers of immune responses. Here, we frame how these cells regulate immune responses via a three-part scheme in which FRC can setup, support or suppress immune responses. We also review how fibroblasts from non-lymphoid tissues influence immunity and highlight how they resemble and differ from FRC. Overall, we aim to focus attention on the emerging roles of lymphoid tissue and non-lymphoid tissue fibroblasts in control of innate and adaptive immunity.
Collapse
Affiliation(s)
- Matthew B Buechler
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, United States
| | - Shannon J Turley
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, United States.
| |
Collapse
|
22
|
Ansorge M, Sapudom J, Chkolnikov M, Wilde M, Anderegg U, Möller S, Schnabelrauch M, Pompe T. Mimicking Paracrine TGFβ1 Signals during Myofibroblast Differentiation in 3D Collagen Networks. Sci Rep 2017; 7:5664. [PMID: 28720779 PMCID: PMC5515936 DOI: 10.1038/s41598-017-05912-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 06/05/2017] [Indexed: 01/06/2023] Open
Abstract
TGFβ1 is a key regulator for induction of tissue remodeling after dermal wounding. We present a model of paracrine delivery of TGFβ1 for differentiation of dermal fibroblasts based on a fibrillar 3D collagen matrix and embedded TGFβ1 releasing microparticles. We found differentiation into myofibroblasts was achieved in a TGFβ1 dependent manner at much lower doses than systemic delivery. This effect is accounted to the slow and sustained TGFβ1 release mimicking paracrine cell signals.
Collapse
Affiliation(s)
- Michael Ansorge
- Universität Leipzig, Institute of Biochemistry, Johannisallee 21/23, 04103, Leipzig, Germany
| | - Jiranuwat Sapudom
- Universität Leipzig, Institute of Biochemistry, Johannisallee 21/23, 04103, Leipzig, Germany
| | - Marina Chkolnikov
- Universität Leipzig, Institute of Biochemistry, Johannisallee 21/23, 04103, Leipzig, Germany
| | - Martin Wilde
- Universität Leipzig, Institute of Biochemistry, Johannisallee 21/23, 04103, Leipzig, Germany
| | - Ulf Anderegg
- Universitätsklinikum Leipzig, Department of Dermatology, Venereology and Allergology, 04103, Leipzig, Germany
| | - Stephanie Möller
- Biomaterials Department, INNOVENT e. V., Prüssingstr. 27B, 07745, Jena, Germany
| | | | - Tilo Pompe
- Universität Leipzig, Institute of Biochemistry, Johannisallee 21/23, 04103, Leipzig, Germany.
| |
Collapse
|
23
|
New E-beam-initiated hyaluronan acrylate cryogels support growth and matrix deposition by dermal fibroblasts. Int J Biol Macromol 2017; 94:611-620. [DOI: 10.1016/j.ijbiomac.2016.10.065] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/07/2016] [Accepted: 10/18/2016] [Indexed: 12/27/2022]
|
24
|
Bustos-Arriaga J, Mita-Mendoza NK, Lopez-Gonzalez M, García-Cordero J, Juárez-Delgado FJ, Gromowski GD, Méndez-Cruz RA, Fairhurst RM, Whitehead SS, Cedillo-Barrón L. Soluble mediators produced by the crosstalk between microvascular endothelial cells and dengue-infected primary dermal fibroblasts inhibit dengue virus replication and increase leukocyte transmigration. Immunol Res 2016; 64:392-403. [PMID: 26130295 DOI: 10.1007/s12026-015-8675-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
When dengue virus (DENV)-infected mosquitoes use their proboscis to probe into human skin during blood feeding, both saliva and virus are released. During this process, cells from the epidermis and dermis layers of the skin, along with small blood vessels, may get exposed to or infected with DENV. In these microenvironments of the skin, the presence of DENV initiates a complex interplay among the DENV-infected and non-infected neighboring cells at the initial bite site. Previous studies suggested that DENV-infected human dermal fibroblasts (HDFs) participate in the immune response against DENV by secreting soluble mediators of innate immunity. In the present study, we investigated whether DENV-infected HDFs activate human dermal microvascular endothelial cells (HDMECs) in co-cultures. Our results suggest that co-cultures of DENV-infected HDFs and HDMECs elicit soluble mediators that are sufficient to reduce viral replication, activate HDMECs, and induce leukocyte migration through HDMEC monolayers. These effects were partly dependent on HDF donor and DENV serotype, which may provide novel insights into the natural variation in host susceptibility to DENV disease.
Collapse
Affiliation(s)
- José Bustos-Arriaga
- Departmento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Mexico, Distrito Federal, Mexico
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Neida K Mita-Mendoza
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Moises Lopez-Gonzalez
- Departmento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Mexico, Distrito Federal, Mexico
| | - Julio García-Cordero
- Departmento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Mexico, Distrito Federal, Mexico
| | | | - Gregory D Gromowski
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - René A Méndez-Cruz
- Laboratorio de Inmunología UMF, FES Iztacala, Universidad Autónoma de México, Mexico, Distrito Federal, Mexico
| | - Rick M Fairhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Stephen S Whitehead
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Leticia Cedillo-Barrón
- Departmento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados, Instituto Politécnico Nacional, Mexico, Distrito Federal, Mexico.
| |
Collapse
|
25
|
Mason LMK, Wagemakers A, van ‘t Veer C, Oei A, van der Pot WJ, Ahmed K, van der Poll T, Geijtenbeek TBH, Hovius JWR. Borrelia burgdorferi Induces TLR2-Mediated Migration of Activated Dendritic Cells in an Ex Vivo Human Skin Model. PLoS One 2016; 11:e0164040. [PMID: 27695100 PMCID: PMC5047638 DOI: 10.1371/journal.pone.0164040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/19/2016] [Indexed: 12/13/2022] Open
Abstract
Borrelia burgdorferi is transmitted into the skin of the host where it encounters and interacts with two dendritic cell (DC) subsets; Langerhans cells (LCs) and dermal DCs (DDCs). These cells recognize pathogens via pattern recognition receptors, mature and migrate out of the skin into draining lymph nodes, where they orchestrate adaptive immune responses. In order to investigate the response of skin DCs during the early immunopathogenesis of Lyme borreliosis, we injected B. burgdorferi intradermally into full-thickness human skin and studied the migration of DCs out of the skin, the activation profile and phenotype of migrated cells. We found a significant increase in the migration of LCs and DDCs in response to B. burgdorferi. Notably, migration was prevented by blocking TLR2. DCs migrated from skin inoculated with higher numbers of spirochetes expressed significantly higher levels of CD83 and produced pro-inflammatory cytokines. No difference was observed in the expression of HLA-DR, CD86, CD38, or CCR7. To conclude, we have established an ex vivo human skin model to study DC-B. burgdorferi interactions. Using this model, we have demonstrated that B. burgdorferi-induced DC migration is mediated by TLR2. Our findings underscore the utility of this model as a valuable tool to study immunity to spirochetal infections.
Collapse
Affiliation(s)
- Lauren M. K. Mason
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
- * E-mail:
| | - Alex Wagemakers
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - Cornelis van ‘t Veer
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - Anneke Oei
- Department of Medical Microbiology, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Kalam Ahmed
- Department of Plastic Surgery, Kennemer Gasthuis, Haarlem, the Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
- Division of Infectious Diseases, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Joppe W. R. Hovius
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands
- Division of Infectious Diseases, Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Here, we explore an emerging theme in the literature, which is the role of dendritic cells in the causation of fibrosis. To fully appreciate this pathway to disease, we also review the most recent literature regarding dendritic cell biology as it pertains to ocular surface tissues. On the basis of this information, we propose a unifying hypothesis for how dendritic cells may cause conjunctival fibrosis in the allergy setting. RECENT FINDINGS Work in models of airway remodeling and liver fibrosis has pointed to a potentially central role for dendritic cells in the pathobiology of fibrosis. Indeed, these cells are recognized as the most potent antigen-presenting cells, and as such activate T lymphocytes that are profibrotic under certain conditions. However, recent findings suggest a more direct role for dendritic cells, which opens up the possibility that a similar pathway may be relevant in the causation of conjunctival fibrosis, particularly in allergic eye disease. SUMMARY Conjunctival fibrosis is a serious clinical concern and is associated with chronic inflammation of the ocular surface tissue, such as in allergic eye disease. Dendritic cells are required in mediating allergic disease by activating pathologic T lymphocytes. Recent findings pointing to a central role for dendritic cell in fibrosis may, however, mean that these cells could also be contributing directly to conjunctival fibrosis. If so, furthering our understanding of dendritic cells could lead to the identification of novel and more effective therapeutic strategies to treat this disease.
Collapse
|
27
|
Damiani E, Ullrich SE. Understanding the connection between platelet-activating factor, a UV-induced lipid mediator of inflammation, immune suppression and skin cancer. Prog Lipid Res 2016; 63:14-27. [PMID: 27073146 DOI: 10.1016/j.plipres.2016.03.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/16/2016] [Accepted: 03/31/2016] [Indexed: 01/08/2023]
Abstract
Lipid mediators of inflammation play important roles in several diseases including skin cancer, the most prevalent type of cancer found in the industrialized world. Ultraviolet (UV) radiation is a complete carcinogen and is the primary cause of skin cancer. UV radiation is also a potent immunosuppressive agent, and UV-induced immunosuppression is a well-known risk factor for skin cancer induction. An essential mediator in this process is the glyercophosphocholine 1-alkyl-2-acetyl-sn-glycero-3-phosphocholine commonly referred to as platelet-activating factor (PAF). PAF is produced by keratinocytes in response to diverse stimuli and exerts its biological effects by binding to a single specific G-protein-coupled receptor (PAF-R) expressed on a variety of cells. This review will attempt to describe how this lipid mediator is involved in transmitting the immunosuppressive signal from the skin to the immune system, starting from its production by keratinocytes, to its role in activating mast cell migration in vivo, and to the mechanisms involved that ultimately lead to immune suppression. Recent findings related to its role in regulating DNA repair and activating epigenetic mechanisms, further pinpoint the importance of this bioactive lipid, which may serve as a critical molecular mediator that links the environment (UVB radiation) to the immune system and the epigenome.
Collapse
Affiliation(s)
- Elisabetta Damiani
- Dipartimento di Scienze della Vita e dell'Ambiente, Universita' Politecnica delle Marche, Ancona, Italy
| | - Stephen E Ullrich
- Department of Immunology and The Center for Cancer Immunology Research, The University of Texas Graduate School for Biomedical Sciences at Houston, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
28
|
Malecka A, Wang Q, Shah S, Sutavani RV, Spendlove I, Ramage JM, Greensmith J, Franks HA, Gough MJ, Saalbach A, Patel PM, Jackson AM. Stromal fibroblasts support dendritic cells to maintain IL-23/Th17 responses after exposure to ionizing radiation. J Leukoc Biol 2016; 100:381-9. [PMID: 27049023 PMCID: PMC4945355 DOI: 10.1189/jlb.3a1015-474r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 03/18/2016] [Indexed: 12/30/2022] Open
Abstract
Cross talk between DCs and FBs in understanding the effects of IR in DC function. Dendritic cell function is modulated by stromal cells, including fibroblasts. Although poorly understood, the signals delivered through this crosstalk substantially alter dendritic cell biology. This is well illustrated with release of TNF-α/IL-1β from activated dendritic cells, promoting PGE2 secretion from stromal fibroblasts. This instructs dendritic cells to up-regulate IL-23, a key Th17-polarizing cytokine. We previously showed that ionizing radiation inhibited IL-23 production by human dendritic cells in vitro. In the present study, we investigated the hypothesis that dendritic cell-fibroblast crosstalk overcomes the suppressive effect of ionizing radiation to support appropriately polarized Th17 responses. Radiation (1–6 Gy) markedly suppressed IL-23 secretion by activated dendritic cells (P < 0.0001) without adversely impacting their viability and consequently, inhibited the generation of Th17 responses. Cytokine suppression by ionizing radiation was selective, as there was no effect on IL-1β, -6, -10, and -27 or TNF-α and only a modest (11%) decrease in IL-12p70 secretion. Coculture with fibroblasts augmented IL-23 secretion by irradiated dendritic cells and increased Th17 responses. Importantly, in contrast to dendritic cells, irradiated fibroblasts maintained their capacity to respond to TNF-α/IL-1β and produce PGE2, thus providing the key intermediary signals for successful dendritic cell-fibroblasts crosstalk. In summary, stromal fibroblasts support Th17-polarizing cytokine production by dendritic cells that would otherwise be suppressed in an irradiated microenvironment. This has potential ramifications for understanding the immune response to local radiotherapy. These findings underscore the need to account for the impact of microenvironmental factors, including stromal cells, in understanding the control of immunity.
Collapse
Affiliation(s)
- Anna Malecka
- Host-Tumour Interactions Group, University of Nottingham, Nottingham, United Kingdom
| | - Qunwei Wang
- Host-Tumour Interactions Group, University of Nottingham, Nottingham, United Kingdom
| | - Sabaria Shah
- Host-Tumour Interactions Group, University of Nottingham, Nottingham, United Kingdom
| | - Ruhcha V Sutavani
- Cancer Immunotherapy Group, Division of Cancer and Stem Cells, University of Nottingham, Nottingham, United Kingdom; Cell Signalling and Immunology, University of Dundee, Scotland, United Kingdom
| | - Ian Spendlove
- Cancer Immunotherapy Group, Division of Cancer and Stem Cells, University of Nottingham, Nottingham, United Kingdom
| | - Judith M Ramage
- Cancer Immunotherapy Group, Division of Cancer and Stem Cells, University of Nottingham, Nottingham, United Kingdom
| | - Julie Greensmith
- Host-Tumour Interactions Group, University of Nottingham, Nottingham, United Kingdom; Intelligent Modelling and Analysis Research Group, University of Nottingham, Nottingham, United Kingdom
| | - Hester A Franks
- Host-Tumour Interactions Group, University of Nottingham, Nottingham, United Kingdom
| | - Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, Oregon, USA
| | - Anja Saalbach
- Klinik fur Dermatologie, University of Leipzig, Germany; and
| | - Poulam M Patel
- Host-Tumour Interactions Group, University of Nottingham, Nottingham, United Kingdom
| | - Andrew M Jackson
- Host-Tumour Interactions Group, University of Nottingham, Nottingham, United Kingdom;
| |
Collapse
|
29
|
Freynet O, Marchal-Sommé J, Jean-Louis F, Mailleux A, Crestani B, Soler P, Michel L. Human lung fibroblasts may modulate dendritic cell phenotype and function: results from a pilot in vitro study. Respir Res 2016; 17:36. [PMID: 27044262 PMCID: PMC4820963 DOI: 10.1186/s12931-016-0345-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 03/09/2016] [Indexed: 01/14/2023] Open
Abstract
In human lung fibrotic lesions, fibroblasts were shown to be closely associated with immature dendritic cell (DC) accumulation. The aim of the present pilot study was to characterize the role of pulmonary fibroblasts on DC phenotype and function, using co-culture of lung fibroblasts from patients with idiopathic pulmonary fibrosis (IPF) and from control patients, with a DC cell line MUTZ-3. We observed that co-culture of lung control and IPF fibroblasts with DCs reduced the expression of specific DC markers and down-regulated their T-cell stimulatory activity. This suggests that pulmonary fibroblasts might sustain chronic inflammation in the fibrotic lung by maintaining in situ a pool of immature DCs.
Collapse
Affiliation(s)
- Olivia Freynet
- Inserm U 1152, 46, rue Henri Huchard, Paris, 75018, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,DHU FIRE, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Service de Pneumologie A, Paris, France
| | - Joëlle Marchal-Sommé
- Inserm U 1152, 46, rue Henri Huchard, Paris, 75018, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,DHU FIRE, Paris, France
| | - Francette Jean-Louis
- Inserm UMR-S 976, Université Paris Diderot, Sorbonne Paris Cité, Hôpital Saint Louis, Paris, France
| | - Arnaud Mailleux
- Inserm U 1152, 46, rue Henri Huchard, Paris, 75018, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,DHU FIRE, Paris, France
| | - Bruno Crestani
- Inserm U 1152, 46, rue Henri Huchard, Paris, 75018, France. .,Université Paris Diderot, Sorbonne Paris Cité, Paris, France. .,DHU FIRE, Paris, France. .,Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Service de Pneumologie A, Paris, France. .,Service de Pneumologie, Hôpital Bichat, 46, rue Henri Huchard, Paris cedex 18, 75018, France.
| | - Paul Soler
- Inserm U 1152, 46, rue Henri Huchard, Paris, 75018, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,DHU FIRE, Paris, France
| | - Laurence Michel
- Inserm UMR-S 976, Université Paris Diderot, Sorbonne Paris Cité, Hôpital Saint Louis, Paris, France. .,Inserm UMR-S 976, Hôpital Saint-Louis, 1 avenue Claude Vellefaux, 75475, Paris, 75010, France.
| |
Collapse
|
30
|
Human Cytomegalovirus-Encoded Human Interleukin-10 (IL-10) Homolog Amplifies Its Immunomodulatory Potential by Upregulating Human IL-10 in Monocytes. J Virol 2016; 90:3819-3827. [PMID: 26792743 DOI: 10.1128/jvi.03066-15] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/14/2016] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED The human cytomegalovirus (HCMV) gene UL111A encodes cytomegalovirus-encoded human interleukin-10 (cmvIL-10), a homolog of the potent immunomodulatory cytokine human interleukin 10 (hIL-10). This viral homolog exhibits a range of immunomodulatory functions, including suppression of proinflammatory cytokine production and dendritic cell (DC) maturation, as well as inhibition of major histocompatibility complex (MHC) class I and class II. Here, we present data showing that cmvIL-10 upregulates hIL-10, and we identify CD14(+)monocytes and monocyte-derived macrophages and DCs as major sources of hIL-10 secretion in response to cmvIL-10. Monocyte activation was not a prerequisite for cmvIL-10-mediated upregulation of hIL-10, which was dose dependent and controlled at the transcriptional level. Furthermore, cmvIL-10 upregulated expression of tumor progression locus 2 (TPL2), which is a regulator of the positive hIL-10 feedback loop, whereas expression of a negative regulator of the hIL-10 feedback loop, dual-specificity phosphatase 1 (DUSP1), remained unchanged. Engagement of the hIL-10 receptor (hIL-10R) by cmvIL-10 led to upregulation of heme oxygenase 1 (HO-1), an enzyme linked with suppression of inflammatory responses, and this upregulation was required for cmvIL-10-mediated upregulation of hIL-10. We also demonstrate an important role for both phosphatidylinositol 3-kinase (PI3K) and STAT3 in the upregulation of HO-1 and hIL-10 by cmvIL-10. In addition to upregulating hIL-10, cmvIL-10 could exert a direct immunomodulatory function, as demonstrated by its capacity to upregulate expression of cell surface CD163 when hIL-10 was neutralized. This study identifies a mechanistic basis for cmvIL-10 function, including the capacity of this viral cytokine to potentially amplify its immunosuppressive impact by upregulating hIL-10 expression. IMPORTANCE Human cytomegalovirus (HCMV) is a large, double-stranded DNA virus that causes significant human disease, particularly in the congenital setting and in solid-organ and hematopoietic stem cell transplant patients. A prominent feature of HCMV is the wide range of viral gene products that it encodes which function to modulate host defenses. One of these is cmvIL-10, which is a homolog of the potent immunomodulatory cytokine human interleukin 10 (hIL-10). In this study, we report that, in addition to exerting a direct biological impact, cmvIL-10 upregulates the expression of hIL-10 by primary blood-derived monocytes and that it does so by modulating existing cellular pathways. This capacity of cmvIL-10 to upregulate hIL-10 represents a mechanism by which HCMV may amplify its immunomodulatory impact during infection.
Collapse
|
31
|
Khosravi-Maharlooei M, Pakyari M, Jalili RB, Salimi-Elizei S, Lai JCY, Poormasjedi-Meibod M, Kilani RT, Dutz J, Ghahary A. Tolerogenic effect of mouse fibroblasts on dendritic cells. Immunology 2016; 148:22-33. [PMID: 26789277 DOI: 10.1111/imm.12584] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 12/28/2015] [Accepted: 01/11/2016] [Indexed: 12/30/2022] Open
Abstract
There is controversy about the immunomodulatory effect of fibroblasts on dendritic cells (DCs). To clarify this issue, in this study, we have evaluated different features of fibroblast-primed DCs including their ability to express co-inhibitory and co-stimulatory molecules, pro-inflammatory and anti-inflammatory cytokines and their ability to induce T-cell proliferation. We also examined migratory capacity of DCs to lymphatic tissues and present fibroblast-derived antigens after encountering fibroblasts. The results of our in vitro study showed that both co-inhibitory (programmed death ligand 1 and ligand 2 and B7H4) and co-stimulatory (CD86) molecules were up-regulated when DCs were co-cultured with fibroblasts. In an animal model, we showed that intra- peritoneal injection (IP) of both syngeneic and allogeneic fibroblasts significantly increased both total DC count and expression level of co-inhibitory and co-stimulatory molecules on DCs. Priming of DCs with syngeneic and allogeneic fibroblasts reduced the proliferation of CD4(+) and CD8(+) T cells. Even activation of fibroblast- primed DCs failed to restore their ability to induce T-cell proliferation. Likewise, priming of DCs with fibroblasts blocked the ability of ovalbumin-pulsed DCs to induce proliferation of ovalbumin-specific CD4(+) T cells. Compared with non-activated DCs, fibroblast-primed DCs had significantly higher expression levels of interleukin-10 and indoleamine 2, 3 dioxygenase. Fibroblast-primed DCs had a significantly reduced interleukin-12 expression level compared with that of activated DCs. After priming with fibroblasts, DCs were able to migrate to lymphatic tissues and present fibroblast-derived antigens (ovalbumin). In conclusion, after priming with fibroblasts, DCs gain tolerogenic features. This finding suggests the potential role of fibroblasts in the maintenance of immune tolerance.
Collapse
Affiliation(s)
| | - MohammadReza Pakyari
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| | - Reza B Jalili
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| | - Sanam Salimi-Elizei
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| | - Jacqueline C Y Lai
- Department of Dermatology and Skin Science, Faculty of Medicine, Child and Family Research Institute, The University of British Columbia, Vancouver, BC, Canada
| | | | - Ruhangiz T Kilani
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| | - Jan Dutz
- Department of Dermatology and Skin Science, Faculty of Medicine, Child and Family Research Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Aziz Ghahary
- Department of Surgery, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
32
|
Shankar SP, Griffith M, Forrester JV, Kuffová L. Dendritic cells and the extracellular matrix: A challenge for maintaining tolerance/homeostasis. World J Immunol 2015; 5:113-130. [DOI: 10.5411/wji.v5.i3.113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/18/2015] [Accepted: 11/11/2015] [Indexed: 02/05/2023] Open
Abstract
The importance of the extracellular matrix (ECM) in contributing to structural, mechanical, functional and tissue-specific features in the body is well appreciated. While the ECM was previously considered to be a passive bystander, it is now evident that it plays active, dynamic and flexible roles in shaping cell survival, differentiation, migration and death to varying extents depending on the specific site in the body. Dendritic cells (DCs) are recognized as potent antigen presenting cells present in many tissues and in blood, continuously scrutinizing the microenvironment for antigens and mounting local and systemic host responses against harmful agents. DCs also play pivotal roles in maintaining homeostasis to harmless self-antigens, critical for preventing autoimmunity. What is less understood are the complex interactions between DCs and the ECM in maintaining this balance between steady-state tissue residence and DC activation during inflammation. DCs are finely tuned to inflammation-induced variations in fragment length, accessible epitopes and post-translational modifications of individual ECM components and correspondingly interpret these changes appropriately by adjusting their profiles of cognate binding receptors and downstream immune activation. The successful design and composition of novel ECM-based mimetics in regenerative medicine and other applications rely on our improved understanding of DC-ECM interplay in homeostasis and the challenges involved in maintaining it.
Collapse
|
33
|
Schmidt-Lauber C, Bossaller L, Abujudeh HH, Vladimer GI, Christ A, Fitzgerald KA, Latz E, Gravallese EM, Marshak-Rothstein A, Kay J. Gadolinium-based compounds induce NLRP3-dependent IL-1β production and peritoneal inflammation. Ann Rheum Dis 2015; 74:2062-9. [PMID: 24914072 PMCID: PMC4428954 DOI: 10.1136/annrheumdis-2013-204900] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 05/22/2014] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Nephrogenic systemic fibrosis (NSF) is a progressive fibrosing disorder that may develop in patients with chronic kidney disease after administration of gadolinium (Gd)-based contrast agents (GBCAs). In the setting of impaired renal clearance of GBCAs, Gd deposits in various tissues and fibrosis subsequently develops. However, the precise mechanism by which fibrosis occurs in NSF is incompletely understood. Because other profibrotic agents, such as silica or asbestos, activate the nucleotide-binding oligomerisation domain (NOD)-like receptor protein 3 (NLRP3) inflammasome and initiate interleukin (IL)-1β release with the subsequent development of fibrosis, we evaluated the effects of GBCAs on inflammasome activation. METHODS Bone marrow derived macrophages from C57BL/6, Nlrp3(-/-) and Asc(-/-) mice were incubated with three Gd-containing compounds and IL-1β activation and secretion was detected by ELISA and western blot analysis. Inflammasome activation and regulation was investigated in IL-4- and interferon (IFN)γ-polarised macrophages by ELISA, quantitative real time (qRT)-PCR and NanoString nCounter analysis. Furthermore, C57BL/6 and Nlrp3(-/-)mice were intraperitoneally injected with GBCA and recruitment of inflammatory cells to the peritoneum was analysed by fluorescence-activated cell sorting (FACS). RESULTS Free Gd and GBCAs activate the NLRP3 inflammasome and induce IL-1β secretion in vitro. Gd-diethylenetriaminepentaacetic acid also induces the recruitment of neutrophils and inflammatory monocytes to the peritoneum in vivo. Gd activated IL-4-polarised macrophages more effectively than IFNγ-polarised macrophages, which preferentially expressed genes known to downregulate inflammasome activity. CONCLUSIONS These data suggest that Gd released from GBCAs triggers a NLRP3 inflammasome-dependent inflammatory response that leads to fibrosis in an appropriate clinical setting. The preferential activation of IL-4-differentiated macrophages is consistent with the predominantly fibrotic presentation of NSF.
Collapse
Affiliation(s)
- Christian Schmidt-Lauber
- Division of Rheumatology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Lukas Bossaller
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Hani H. Abujudeh
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gregory I. Vladimer
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Anette Christ
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Katherine A. Fitzgerald
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Eicke Latz
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
- Institute of Innate Immunity, University of Bonn and German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ellen M. Gravallese
- Division of Rheumatology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Ann Marshak-Rothstein
- Division of Rheumatology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jonathan Kay
- Division of Rheumatology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
34
|
Amoah BP, Yang H, Zhang P, Su Z, Xu H. Immunopathogenesis of Myocarditis: The Interplay Between Cardiac Fibroblast Cells, Dendritic Cells, Macrophages and CD4+T Cells. Scand J Immunol 2015; 82:1-9. [DOI: 10.1111/sji.12298] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 02/21/2015] [Accepted: 03/14/2015] [Indexed: 02/06/2023]
Affiliation(s)
- B. Prince Amoah
- Department of Immunology; School of Medical Science and Laboratory Medicine; Jiangsu University; Zhenjiang China
- Department of Biomedical and Forensic Sciences; School of Biological Sciences; University of Cape Coast; Cape Coast Ghana
| | - H. Yang
- Department of Immunology; School of Medical Science and Laboratory Medicine; Jiangsu University; Zhenjiang China
| | - P. Zhang
- Department of Immunology; School of Medical Science and Laboratory Medicine; Jiangsu University; Zhenjiang China
| | - Z. Su
- Department of Immunology; School of Medical Science and Laboratory Medicine; Jiangsu University; Zhenjiang China
- The Central Laboratory; The Fourth Affiliated Hospital of Jiangsu University; Zhenjiang China
| | - H. Xu
- Department of Immunology; School of Medical Science and Laboratory Medicine; Jiangsu University; Zhenjiang China
| |
Collapse
|
35
|
van der Smissen A, Hoffmeister PG, Friedrich N, Watarai A, Hacker MC, Schulz-Siegmund M, Anderegg U. Artificial extracellular matrices support cell growth and matrix synthesis of human dermal fibroblasts in macroporous 3D scaffolds. J Tissue Eng Regen Med 2015; 11:1390-1402. [DOI: 10.1002/term.2037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 01/26/2015] [Accepted: 04/22/2015] [Indexed: 01/10/2023]
Affiliation(s)
- Anja van der Smissen
- Department of Dermatology, Venereology and Allergology; Leipzig University; Germany
- Collaborative Research Centre (SFB-TR67); Matrix Engineering Leipzig and Dresden; Germany
| | - Peter-Georg Hoffmeister
- Pharmaceutical Technology, Institute of Pharmacy; Leipzig University; Germany
- Collaborative Research Centre (SFB-TR67); Matrix Engineering Leipzig and Dresden; Germany
| | - Nadja Friedrich
- Department of Dermatology, Venereology and Allergology; Leipzig University; Germany
- Collaborative Research Centre (SFB-TR67); Matrix Engineering Leipzig and Dresden; Germany
| | - Akira Watarai
- Department of Dermatology, Venereology and Allergology; Leipzig University; Germany
- Collaborative Research Centre (SFB-TR67); Matrix Engineering Leipzig and Dresden; Germany
- School of Medicine; Kitasato University; Japan
| | - Michael C. Hacker
- Pharmaceutical Technology, Institute of Pharmacy; Leipzig University; Germany
- Collaborative Research Centre (SFB-TR67); Matrix Engineering Leipzig and Dresden; Germany
| | - Michaela Schulz-Siegmund
- Pharmaceutical Technology, Institute of Pharmacy; Leipzig University; Germany
- Collaborative Research Centre (SFB-TR67); Matrix Engineering Leipzig and Dresden; Germany
| | - Ulf Anderegg
- Department of Dermatology, Venereology and Allergology; Leipzig University; Germany
- Collaborative Research Centre (SFB-TR67); Matrix Engineering Leipzig and Dresden; Germany
| |
Collapse
|
36
|
Saalbach A, Janik T, Busch M, Herbert D, Anderegg U, Simon JC. Fibroblasts support migration of monocyte-derived dendritic cells by secretion of PGE2 and MMP-1. Exp Dermatol 2015; 24:598-604. [PMID: 25865800 DOI: 10.1111/exd.12722] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2015] [Indexed: 12/14/2022]
Abstract
The outcome of a cutaneous immune response is critically dependent upon the ability of dendritic cells (DC) to migrate from skin to the draining lymph nodes - a process that is influenced by the cutaneous tissue microenvironment. Here, the role of fibroblasts - a major component of the dermal microenvironment - on the migratory capacity of monocyte-derived DC (MoDC) was investigated in a 3D collagen I matrix. Indeed, dermal fibroblasts supported the migration of pre-activated MoDC through a 3D collagen I matrix. Activation of human MoDC resulted in the release of TNFα and IL-1β that in turn stimulated MMP-1 (human collagenase) and PGE2 secretion by human dermal fibroblasts. Transmigration assays confirmed the importance of fibroblast-derived MMP-1 and PGE2 for the migration of MoDC through a 3D collagen I matrix. Finally, in mice initiation of inflammation by induction of an irritant contact dermatitis or a psoriasis-like skin inflammation, the expression of the PGE2 generating cox-2 and the mouse collagen I degrading enzyme matrix metalloproteinases (MMP)-13 was strongly up-regulated. Our study indicates that MoDC are able to instruct dermal fibroblasts resulting in enhanced migratory capability of MoDC, thus highlighting the role of a crosstalk of DC with their stromal microenvironment for the control of cutaneous immune responses.
Collapse
Affiliation(s)
- Anja Saalbach
- Department of Dermatology, Venerology and Allergology, Medical Faculty of Leipzig University, Leipzig, Germany
| | - Tobias Janik
- Department of Dermatology, Venerology and Allergology, Medical Faculty of Leipzig University, Leipzig, Germany
| | - Matthias Busch
- Department of Dermatology, Venerology and Allergology, Medical Faculty of Leipzig University, Leipzig, Germany
| | - Diana Herbert
- Department of Dermatology, Venerology and Allergology, Medical Faculty of Leipzig University, Leipzig, Germany
| | - Ulf Anderegg
- Department of Dermatology, Venerology and Allergology, Medical Faculty of Leipzig University, Leipzig, Germany
| | - Jan C Simon
- Department of Dermatology, Venerology and Allergology, Medical Faculty of Leipzig University, Leipzig, Germany
| |
Collapse
|
37
|
Mancini RJ, Stutts L, Moore T, Esser-Kahn AP. Controlling the origins of inflammation with a photoactive lipopeptide immunopotentiator. Angew Chem Int Ed Engl 2015; 54:5962-5. [PMID: 25800006 DOI: 10.1002/anie.201500416] [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] [Received: 01/15/2015] [Indexed: 11/06/2022]
Abstract
Inflammatory immune responses are mediated by signaling molecules that are both produced by and recognized across highly heterogeneous cell populations. As such, the study of inflammation using traditional immunostimulants is complicated by paracrine and autocrine signaling, which obscures the origin of a propagating response. To address this challenge, we developed a small-molecule probe that can photosensitize immune cells, thus allowing light-mediated inflammation. This probe was used to control the origin of inflammation using light. Following this motif, inflammation was initiated from fibroblasts or dendritic cells. The contributions of fibroblasts and dendritic cells in initiating inflammation in heterogeneous co-culture are reported, thus providing insights into the future development of vaccines and treatment of inflammation.
Collapse
Affiliation(s)
- Rock J Mancini
- Department of Chemistry, University of California, Irvine, 1102 Natural Sciences II, Irvine, CA 92697 (USA)
| | | | | | | |
Collapse
|
38
|
Mancini RJ, Stutts L, Moore T, Esser-Kahn AP. Controlling the Origins of Inflammation with a Photoactive Lipopeptide Immunopotentiator. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201500416] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
Hübel J, Hieronymus T. HGF/Met-Signaling Contributes to Immune Regulation by Modulating Tolerogenic and Motogenic Properties of Dendritic Cells. Biomedicines 2015; 3:138-148. [PMID: 28536404 PMCID: PMC5344228 DOI: 10.3390/biomedicines3010138] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/06/2015] [Accepted: 02/13/2015] [Indexed: 12/13/2022] Open
Abstract
Hepatocyte growth factor (HGF)-signaling via Met can induce mitogenic, morphogenic, and motogenic activity in various cell types. Met expression in the immune system is limited to cells with antigen-presenting capacities, including dendritic cells (DCs). Thus, it appears highly conceivable that Met-signaling impacts on adaptive immune responses. However, the mechanisms by which HGF imparts its effects on immunological responses are not yet fully understood. DCs possess unique functionalities that are critically involved in controlling both tolerance and immunity. HGF conveys immunoregulatory functions, which strongly correlate with that of DCs orchestrating the apt immune response in inflammation. Therefore, this review focuses on the current knowledge of Met-signaling in DCs with specific emphasis on the morphogenic and motogenic activities. HGF has been identified to play a role in peripheral immune tolerance by directing DC differentiation towards a tolerogenic phenotype. In skin immunity, Met-signaling was shown to drive mobilization of DCs by regulating matrix metalloproteinase activities. This is strikingly reminiscent of the role of Met for regulating a cell fate program during embryonic development, wound healing, and in tumor invasion known as epithelial-mesenchymal transition (EMT). Thus, the concept emerges that an EMT program is executed by Met-signaling in DCs, which will be also discussed.
Collapse
Affiliation(s)
- Jessica Hübel
- Department of Cell Biology, Institute for Biomedical Engineering, Medical Faculty, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany.
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 20, 52074 Aachen, Germany.
| | - Thomas Hieronymus
- Department of Cell Biology, Institute for Biomedical Engineering, Medical Faculty, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany.
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 20, 52074 Aachen, Germany.
| |
Collapse
|
40
|
Vera J, Raatz Y, Wolkenhauer O, Kottek T, Bhattacharya A, Simon JC, Kunz M. Chk1 and Wee1 control genotoxic-stress induced G2-M arrest in melanoma cells. Cell Signal 2015; 27:951-60. [PMID: 25683911 DOI: 10.1016/j.cellsig.2015.01.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 01/31/2015] [Indexed: 11/29/2022]
Abstract
In the present report, the role of ATR-Chk1-Wee1 and ATM-Chk2-p53-p21 pathways in stress-induced cell cycle control is analysed in melanoma cells. Treatment of p53 wild-type melanoma cells with the genotoxic agent doxorubicin induces G2-M arrest, inhibitory phosphorylation of cell cycle kinase Cdc2 (CDK1) and enhanced expression of p53/p21. Wee1 inhibition under doxorubicin pulse-treatment reduces G2-M arrest and induces apoptosis. Inhibition of upstream kinase Chk1 under doxorubicin treatment almost completely abolishes stress-induced G2-M arrest and induces enhanced apoptosis. Interestingly, Chk1 inhibition alone even further increases apoptosis. While Chk1 inhibition alone almost completely abolishes G0-G1 arrest, combined treatment with doxorubicin re-establishes G0-G1 arrest. Moreover, Chk1 inhibition alone induces only a slight p53/p21 induction, while a strong induction of both proteins is observed by the combination with doxorubicin. These findings are suggestive for a particular role of p53/p21 in G0-G1, and Chk1 in G0-G1 and G2-M arrest. In line with this, the p53-mutant SK-Mel-28 melanoma cells do not mount a significant G0-G1 arrest under combined doxorubicin and Chk1 inhibitor treatment but rather show extensive apoptosis. Moreover, knockdown of p21 dramatically reduces stress-induced G0-G1 arrest under doxorubicin and Chk1 inhibitor treatment accompanied by massive DNA damage and apoptosis induction. Treatment of melanoma cells with an inhibitor of Chk2 upstream kinase ATM and doxorubicin almost completely abolishes G0-G1 arrest. Taken together, both Chk1 and Wee1 are mediators of G2-M arrest, while p53, p21 and Chk1 are mediators of G0-G1 arrest in melanoma cells. Combined treatment with chemotherapeutic agents such as doxorubicin and Chk1 inhibitors may help to overcome apoptosis resistance of p53-proficient melanoma cells. But treatment with Chk1 inhibitor alone may even be more efficient.
Collapse
Affiliation(s)
- Julio Vera
- Laboratory of Systems Tumor Immunology, Department of Dermatology, University Hospital Erlangen and Friedrich-Alexander-University Erlangen-Nürnberg, Ulmenweg 18, 91054 Erlangen, Germany
| | - Yvonne Raatz
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Philipp-Rosenthal-Str. 23, 04103 Leipzig, Germany
| | - Olaf Wolkenhauer
- Department of Systems Biology & Bioinformatics, University of Rostock, Ulmenstrasse 69, 18057 Rostock, Germany
| | - Tina Kottek
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Philipp-Rosenthal-Str. 23, 04103 Leipzig, Germany
| | - Animesh Bhattacharya
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Philipp-Rosenthal-Str. 23, 04103 Leipzig, Germany
| | - Jan C Simon
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Philipp-Rosenthal-Str. 23, 04103 Leipzig, Germany
| | - Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Philipp-Rosenthal-Str. 23, 04103 Leipzig, Germany
| |
Collapse
|
41
|
Sapudom J, Rubner S, Martin S, Thoenes S, Anderegg U, Pompe T. The interplay of fibronectin functionalization and TGF-β1 presence on fibroblast proliferation, differentiation and migration in 3D matrices. Biomater Sci 2015; 3:1291-301. [DOI: 10.1039/c5bm00140d] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
TGF-β1 dependent fibroblast behaviour in a wound healing context is mimicked by topologically and mechanically defined collagen matrices with fibronectin functionalization.
Collapse
Affiliation(s)
- Jiranuwat Sapudom
- Institute of Biochemistry
- Faculty of Biosciences
- Pharmacy and Psychology
- Universität Leipzig
- Leipzig 04103
| | - Stefan Rubner
- Institute of Biochemistry
- Faculty of Biosciences
- Pharmacy and Psychology
- Universität Leipzig
- Leipzig 04103
| | - Steve Martin
- Institute of Biochemistry
- Faculty of Biosciences
- Pharmacy and Psychology
- Universität Leipzig
- Leipzig 04103
| | - Stephan Thoenes
- Department of Dermatology
- Venereology and Allergology
- Universitätsklinikum Leipzig
- Leipzig 04103
- Germany
| | - Ulf Anderegg
- Department of Dermatology
- Venereology and Allergology
- Universitätsklinikum Leipzig
- Leipzig 04103
- Germany
| | - Tilo Pompe
- Institute of Biochemistry
- Faculty of Biosciences
- Pharmacy and Psychology
- Universität Leipzig
- Leipzig 04103
| |
Collapse
|
42
|
Kato J, Kamiya H, Himeno T, Shibata T, Kondo M, Okawa T, Fujiya A, Fukami A, Uenishi E, Seino Y, Tsunekawa S, Hamada Y, Naruse K, Oiso Y, Nakamura J. Mesenchymal stem cells ameliorate impaired wound healing through enhancing keratinocyte functions in diabetic foot ulcerations on the plantar skin of rats. J Diabetes Complications 2014; 28:588-95. [PMID: 25027388 DOI: 10.1016/j.jdiacomp.2014.05.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/29/2014] [Accepted: 05/14/2014] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Although the initial healing stage involves a re-epithelialization in humans, diabetic foot ulceration (DFU) has been investigated using rodent models with wounds on the thigh skin, in which a wound contraction is initiated. In this study, we established a rodent model of DFU on the plantar skin and evaluated the therapeutic efficacy of bone-marrow-derived mesenchymal stem cells (BM-MSCs) in this model. METHODS The wounds made on the hind paws or thighs of streptozotocin induced diabetic or control rats were treated with BM-MSCs. Expression levels of phosphorylated focal adhesion kinase (pFAK), matrix metaroprotease (MMP)-2, EGF, and IGF-1, were evaluated in human keratinocytes, which were cultured in conditioned media of BM-MSCs (MSC-CM) with high glucose levels. RESULTS Re-epithelialization initiated the healing process on the plantar, but not on the thigh, skin. The therapy utilizing BM-MSCs ameliorated the delayed healing in diabetic rats. In the keratinocytes cultured with MSC-CM, the decreased pFAK levels in the high glucose condition were restored, and the MMP2, EGF, and IGF-1 levels increased. CONCLUSIONS/INTERPRETATION Our study established a novel rat DFU model. The impaired healing process in diabetic rats was ameliorated by transplantation of BM-MSCs. This amelioration might be accounted for by the modification of keratinocyte functions.
Collapse
Affiliation(s)
- Jiro Kato
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Hideki Kamiya
- Department of CKD Initiatives, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Tatsuhito Himeno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Taiga Shibata
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Masaki Kondo
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tetsuji Okawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Atsushi Fujiya
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Ayako Fukami
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Eita Uenishi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yusuke Seino
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan; Department of Metabolic Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Shin Tsunekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yoji Hamada
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan; Department of Metabolic Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Keiko Naruse
- Department of Internal Medicine, School of Dentistry, Aichi-Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan
| | - Yutaka Oiso
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Jiro Nakamura
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
43
|
Schönherr M, Bhattacharya A, Kottek T, Szymczak S, Köberle M, Wickenhauser C, Siebolts U, Saalbach A, Koczan D, Magin TM, Simon JC, Kunz M. Genomewide RNAi screen identifies protein kinase Cb and new members of mitogen-activated protein kinase pathway as regulators of melanoma cell growth and metastasis. Pigment Cell Melanoma Res 2014; 27:418-30. [PMID: 24406113 DOI: 10.1111/pcmr.12216] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 01/07/2014] [Indexed: 01/13/2023]
Abstract
A large-scale RNAi screen was performed for eight different melanoma cell lines using a pooled whole-genome lentiviral shRNA library. shRNAs affecting proliferation of transduced melanoma cells were negatively selected during 10 days of culture. Overall, 617 shRNAs were identified by microarray hybridization. Pathway analyses identified mitogen-activated protein kinase (MAPK) pathway members such as ERK1/2, JNK1/2 and MAP3K7 and protein kinase C β (PKCβ) as candidate genes. Knockdown of PKCβ most consistently reduced cellular proliferation, colony formation and migratory capacity of melanoma cells and was selected for further validation. PKCβ showed enhanced expression in human primary melanomas and distant metastases as compared with benign melanocytic nevi. Moreover, treatment of melanoma cells with PKCβ-specific inhibitor enzastaurin reduced melanoma cell growth but had only small effects on benign fibroblasts. Finally, PKCβ-shRNA significantly reduced lung colonization capacity of stably transduced melanoma cells in mice. Taken together, this study identified new candidate genes for melanoma cell growth and proliferation. PKCβ seems to play an important role in these processes and might serve as a new target for the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Madeleine Schönherr
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Leipzig, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
van der Smissen A, Samsonov S, Hintze V, Scharnweber D, Moeller S, Schnabelrauch M, Pisabarro MT, Anderegg U. Artificial extracellular matrix composed of collagen I and highly sulfated hyaluronan interferes with TGFβ(1) signaling and prevents TGFβ(1)-induced myofibroblast differentiation. Acta Biomater 2013; 9:7775-86. [PMID: 23602877 DOI: 10.1016/j.actbio.2013.04.023] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 03/27/2013] [Accepted: 04/09/2013] [Indexed: 01/24/2023]
Abstract
Sulfated glycosaminoglycans are promising components for functional biomaterials since sulfate groups modulate the binding of growth factors and thereby influence wound healing. Here, we have investigated the influence of an artificial extracellular matrix (aECM) consisting of collagen I (coll) and hyaluronan (HA) or highly sulfated HA (hsHA) on dermal fibroblasts (dFb) with respect to their differentiation into myofibroblasts (MFb). Fibroblasts were cultured on aECM in the presence of aECM-adsorbed or soluble transforming growth factor β1 (TGFβ1). The synthesis of α-smooth muscle actin (αSMA), collagen and the ED-A splice variant of fibronectin (ED-A FN) were analyzed at the mRNA and protein levels. Furthermore, we investigated the bioactivity and signal transduction of TGFβ1 in the presence of aECM and finally made interaction studies of soluble HA or hsHA with TGFβ1. Artificial ECM composed of coll and hsHA prevents TGFβ1-stimulated αSMA, collagen and ED-A FN expression. Our data suggest an impaired TGFβ1 bioactivity and downstream signaling in the presence of aECM containing hsHA, shown by massively reduced Smad2/3 translocation to the nucleus. These data are explained by in silico docking experiments demonstrating the occupation of the TGFβ-receptor I binding site by hsHA. Possibly, HA sulfation has a strong impact on TGFβ1-driven differentiation of dFb and thus could be used to modulate the properties of biomaterials.
Collapse
Affiliation(s)
- Anja van der Smissen
- Department of Dermatology, Venereology and Allergology, Leipzig University, 04103 Leipzig, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Kuroishi T, Bando K, Endo Y, Sugawara S. Metal Allergens Induce Nitric Oxide Production by Mouse Dermal Fibroblasts via the Hypoxia-Inducible Factor-2α–Dependent Pathway. Toxicol Sci 2013; 135:119-28. [DOI: 10.1093/toxsci/kft142] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
46
|
Matsuo K, Hirobe S, Okada N, Nakagawa S. Frontiers of transcutaneous vaccination systems: novel technologies and devices for vaccine delivery. Vaccine 2013; 31:2403-15. [PMID: 23523401 PMCID: PMC7125630 DOI: 10.1016/j.vaccine.2013.03.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2013] [Indexed: 12/24/2022]
Abstract
Transcutaneous immunization (TCI) systems that use the skin's immune function are promising needle-free, easy-to-use, and low-invasive vaccination alternative to conventional, injectable vaccination methods. To develop effective TCI systems, it is essential to establish fundamental techniques and technologies that deliver antigenic proteins to antigen-presenting cells in the epidermis and dermis while overcoming the barrier function of the stratum corneum. In this review, we provide an outline of recent trends in the development of techniques for the delivery of antigenic proteins and of the technologies used to enhance TCI systems. We also introduce basic and clinical research involving our TCI systems that incorporate several original devices.
Collapse
Affiliation(s)
- Kazuhiko Matsuo
- Laboratory of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | |
Collapse
|
47
|
Artificial extracellular matrices composed of collagen I and high-sulfated hyaluronan promote phenotypic and functional modulation of human pro-inflammatory M1 macrophages. Acta Biomater 2013; 9:5621-9. [PMID: 23168224 DOI: 10.1016/j.actbio.2012.11.016] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 11/02/2012] [Accepted: 11/07/2012] [Indexed: 01/22/2023]
Abstract
The sequential phases of biomaterial integration and wound healing require different macrophage functions mediated by distinct macrophage subsets. During the initial phase of healing, pro-inflammatory M1 macrophages (MΦ1) are required to clear the wound from microbes and debris; however, their unopposed, persistent activation often leads to disturbed integration of biomaterials and perturbed wound healing. Here we investigated whether pro-inflammatory macrophage functions are affected by immunomodulatory biomaterials based on artificial extracellular matrices (aECM). To address this issue, we tested the capacity of two-dimensional aECM consisting of collagen I and hyaluronan or sulfated derivatives of hyaluronan to affect functions of in vitro polarized human pro-inflammatory MΦ1. The aECM containing high-sulfated hyaluronan substantially decreased inflammatory macrophage functions, including pathogen uptake and release of the pro-inflammatory cytokines tumor necrosis factor alpha and interleukin-12 due to impaired activation of nuclear factor "kappa-light-chain-enhancer" of activated B-cells. Moreover, these macrophages secreted immunregulatory IL-10 and showed reduced activity of the transcription factors signal transducer and activator of transcription 1 and interferon-regulating factor 5, both controlling macrophage polarization to MΦ1 subsets. Our data reveal that the collagen I matrix containing high-sulfated hyaluronan possesses immunomodulating properties and dampens inflammatory macrophage activities by impeding signaling pathways crucial for polarization of pro-inflammatory MΦ1. We therefore suggest this aECM as a promising coating for biomaterials to modulate inflammatory macrophage functions during the healing response and recommend its further testing as a three-dimensional construct and in in vivo models.
Collapse
|
48
|
Müller SA, van der Smissen A, von Feilitzsch M, Anderegg U, Kalkhof S, von Bergen M. Quantitative proteomics reveals altered expression of extracellular matrix related proteins of human primary dermal fibroblasts in response to sulfated hyaluronan and collagen applied as artificial extracellular matrix. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2012; 23:3053-3065. [PMID: 22990618 PMCID: PMC3506194 DOI: 10.1007/s10856-012-4760-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 08/27/2012] [Indexed: 06/01/2023]
Abstract
Fibroblasts are the main matrix producing cells of the dermis and are also strongly regulated by their matrix environment which can be used to improve and guide skin wound healing processes. Here, we systematically investigated the molecular effects on primary dermal fibroblasts in response to high-sulfated hyaluronan [HA] (hsHA) by quantitative proteomics. The comparison of non- and high-sulfated HA revealed regulation of 84 of more than 1,200 quantified proteins. Based on gene enrichment we found that sulfation of HA alters extracellular matrix remodeling. The collagen degrading enzymes cathepsin K, matrix metalloproteinases-2 and -14 were found to be down-regulated on hsHA. Additionally protein expression of thrombospondin-1, decorin, collagen types I and XII were reduced, whereas the expression of trophoblast glycoprotein and collagen type VI were slightly increased. This study demonstrates that global proteomics provides a valuable tool for revealing proteins involved in molecular effects of growth substrates for further material optimization.
Collapse
Affiliation(s)
- Stephan A. Müller
- Department of Proteomics, UFZ, Helmholtz-Centre for Environmental Research Leipzig, 04318 Leipzig, Germany
- Collaborative Research Center (SFB-TR67), Matrixengineering, Leipzig, Germany
| | - Anja van der Smissen
- Department of Dermatology Venerology and Allergology, Leipzig University, 04103 Leipzig, Germany
- Collaborative Research Center (SFB-TR67), Matrixengineering, Leipzig, Germany
| | - Margarete von Feilitzsch
- Department of Dermatology Venerology and Allergology, Leipzig University, 04103 Leipzig, Germany
- Collaborative Research Center (SFB-TR67), Matrixengineering, Leipzig, Germany
| | - Ulf Anderegg
- Department of Dermatology Venerology and Allergology, Leipzig University, 04103 Leipzig, Germany
- Collaborative Research Center (SFB-TR67), Matrixengineering, Leipzig, Germany
| | - Stefan Kalkhof
- Department of Proteomics, UFZ, Helmholtz-Centre for Environmental Research Leipzig, 04318 Leipzig, Germany
- Collaborative Research Center (SFB-TR67), Matrixengineering, Leipzig, Germany
| | - Martin von Bergen
- Department of Proteomics, UFZ, Helmholtz-Centre for Environmental Research Leipzig, 04318 Leipzig, Germany
- Department of Metabolomics, UFZ, Helmholtz-Centre for Environmental Research Leipzig, 04318 Leipzig, Germany
- Collaborative Research Center (SFB-TR67), Matrixengineering, Leipzig, Germany
| |
Collapse
|
49
|
Kendall AC, Nicolaou A. Bioactive lipid mediators in skin inflammation and immunity. Prog Lipid Res 2012; 52:141-64. [PMID: 23124022 DOI: 10.1016/j.plipres.2012.10.003] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 10/15/2012] [Accepted: 10/17/2012] [Indexed: 12/20/2022]
Abstract
The skin is the primary barrier from the outside environment, protecting the host from injury, infectious pathogens, water loss and solar ultraviolet radiation. In this role, it is supported by a highly organized system comprising elements of innate and adaptive immunity, responsive to inflammatory stimuli. The cutaneous immune system is regulated by mediators such as cytokines and bioactive lipids that can initiate rapid immune responses with controlled inflammation, followed by efficient resolution. However, when immune responses are inadequate or mounted against non-infectious agents, these mediators contribute to skin pathologies involving unresolved or chronic inflammation. Skin is characterized by active lipid metabolism and fatty acids play crucial roles both in terms of structural integrity and functionality, in particular when transformed to bioactive mediators. Eicosanoids, endocannabinoids and sphingolipids are such key bioactive lipids, intimately involved in skin biology, inflammation and immunity. We discuss their origins, role and influence over various cells of the epidermis, dermis and cutaneous immune system and examine their function in examples of inflammatory skin conditions. We focus on psoriasis, atopic and contact dermatitis, acne vulgaris, wound healing and photodermatology that demonstrate dysregulation of bioactive lipid metabolism and examine ways of using this insight to inform novel therapeutics.
Collapse
Affiliation(s)
- Alexandra C Kendall
- School of Pharmacy and Centre for Skin Sciences, School of Life Sciences, University of Bradford, Richmond Road, Bradford BD7 1DP, UK
| | | |
Collapse
|
50
|
Wu TH, Hsu SH, Chang MH, Huang YY. Reducing scar formation by regulation of IL-1 and MMP-9 expression by using sustained release of prednisolone-loaded PDLL microspheres in a murine wound model. J Biomed Mater Res A 2012; 101:1165-72. [DOI: 10.1002/jbm.a.34413] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 06/09/2012] [Accepted: 07/25/2012] [Indexed: 01/04/2023]
|