1
|
Kim MC, De U, Borcherding N, Wang L, Paek J, Bhattacharyya I, Yu Q, Kolb R, Drashansky T, Thatayatikom A, Zhang W, Cha S. Single-cell transcriptomics unveil profiles and interplay of immune subsets in rare autoimmune childhood Sjögren's disease. Commun Biol 2024; 7:481. [PMID: 38641668 PMCID: PMC11031574 DOI: 10.1038/s42003-024-06124-6] [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: 06/07/2023] [Accepted: 03/29/2024] [Indexed: 04/21/2024] Open
Abstract
Childhood Sjögren's disease represents critically unmet medical needs due to a complete lack of immunological and molecular characterizations. This study presents key immune cell subsets and their interactions in the periphery in childhood Sjögren's disease. Here we show that single-cell RNA sequencing identifies the subsets of IFN gene-enriched monocytes, CD4+ T effector memory, and XCL1+ NK cells as potential key players in childhood Sjögren's disease, and especially in those with recurrent parotitis, which is the chief symptom prompting clinical visits from young children. A unique cluster of monocytes with type I and II IFN-related genes is identified in childhood Sjögren's disease, compared to the age-matched control. In vitro regulatory T cell functional assay demonstrates intact functionality in childhood Sjögren's disease in contrast to reduced suppression in adult Sjögren's disease. Mapping this transcriptomic landscape and interplay of immune cell subsets will expedite the understanding of childhood Sjögren's disease pathogenesis and set the foundation for precision medicine.
Collapse
Affiliation(s)
- Myung-Chul Kim
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
- Diagnostic Laboratory Medicine, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea
- Research Institute of Veterinary Medicine, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea
- Center for Orphaned Autoimmune Disorders, University of Florida College of Dentistry, Gainesville, FL, 32610, USA
| | - Umasankar De
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Nicholas Borcherding
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St Louis, MO, 63110, USA
| | - Lei Wang
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Joon Paek
- Center for Orphaned Autoimmune Disorders, University of Florida College of Dentistry, Gainesville, FL, 32610, USA
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St Louis, MO, 63110, USA
| | - Indraneel Bhattacharyya
- Center for Orphaned Autoimmune Disorders, University of Florida College of Dentistry, Gainesville, FL, 32610, USA
- Department of Oral & Maxillofacial Diagnostic Sciences, University of Florida College of Dentistry, Gainesville, FL, 32610, USA
| | - Qing Yu
- The Forsyth Institute, Cambridge, MA, 02142, USA
| | - Ryan Kolb
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | | | | | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, 32610, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA.
| | - Seunghee Cha
- Center for Orphaned Autoimmune Disorders, University of Florida College of Dentistry, Gainesville, FL, 32610, USA.
- Department of Oral & Maxillofacial Diagnostic Sciences, University of Florida College of Dentistry, Gainesville, FL, 32610, USA.
| |
Collapse
|
2
|
Zhang Z, Chen X, Li B, Xia T, Wu X, Wu C. Helicobacter pylori induces urease subunit B-specific CD8 + T cell responses in infected individuals via cytosolic pathway of cross-presentation. Helicobacter 2023; 28:e13005. [PMID: 37382428 DOI: 10.1111/hel.13005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/04/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND Urease subunit B (UreB), a conserved and key virulence factor of Helicobacter pylori (H. pylori), can induce the host CD4+ T cell immune responses to provide protection, but less is known regarding CD8+ T cell responses. The characteristics of H. pylori-specific CD8+ T cell responses and the mechanism underlying antigen processing and presentation pathways remain unclear. This study was focus on protective antigen recombinant UreB (rUreb) to detect specific CD8+ T cell responses in vitro and elucidate the mechanism of UreB antigen processing and presentation. METHODS The peripheral blood mononuclear cells (PBMCs) collected from H. pylori-infected individuals were stimulated with rUreB in vitro to detect specific CD8+ T cell responses after co-culture with rUreB-pulsed autologous hMDCs. Through blocking assay, we investigated the potential pathway of UreB antigen processing and presentation via the cytosolic pathway or vacuolar pathway. The cytokines production of UreB specific CD8+ T cell were evaluated as well. RESULTS We demonstrated UreB can induce specific CD8+ T cell immune responses in H. pylori infected individuals. Importantly, we characterized that UreB were mainly processed by proteasome instead of lysosomal proteases and presented through cytosolic pathway of cross-presentation, which requires endoplasmic reticulum-Golgi transport and newly synthesized MHC-I molecules, to induce functional-specific CD8+ T cell (IFN-γ + TNF-α + Grz A+ Grz B+) responses. CONCLUSIONS These results suggest that H. pylori UreB induces specific CD8+ T cell responses through cytosolic pathway of cross-presentation in infected individuals.
Collapse
Affiliation(s)
- Zelin Zhang
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xingchi Chen
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Bin Li
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Tingting Xia
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiaobin Wu
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chao Wu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
3
|
Affandi AJ, Olesek K, Grabowska J, Nijen Twilhaar MK, Rodríguez E, Saris A, Zwart ES, Nossent EJ, Kalay H, de Kok M, Kazemier G, Stöckl J, van den Eertwegh AJM, de Gruijl TD, Garcia-Vallejo JJ, Storm G, van Kooyk Y, den Haan JMM. CD169 Defines Activated CD14 + Monocytes With Enhanced CD8 + T Cell Activation Capacity. Front Immunol 2021; 12:697840. [PMID: 34394090 PMCID: PMC8356644 DOI: 10.3389/fimmu.2021.697840] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/13/2021] [Indexed: 12/20/2022] Open
Abstract
Monocytes are antigen-presenting cells (APCs) that play diverse roles in promoting or regulating inflammatory responses, but their role in T cell stimulation is not well defined. In inflammatory conditions, monocytes frequently show increased expression of CD169/Siglec-1, a type-I interferon (IFN-I)-regulated protein. However, little is known about the phenotype and function of these CD169+ monocytes. Here, we have investigated the phenotype of human CD169+ monocytes in different diseases, their capacity to activate CD8+ T cells, and the potential for a targeted-vaccination approach. Using spectral flow cytometry, we detected CD169 expression by CD14+ CD16- classical and CD14+ CD16+ intermediate monocytes and unbiased analysis showed that they were distinct from dendritic cells, including the recently described CD14-expressing DC3. CD169+ monocytes expressed higher levels of co-stimulatory and HLA molecules, suggesting an increased activation state. IFNα treatment highly upregulated CD169 expression on CD14+ monocytes and boosted their capacity to cross-present antigen to CD8+ T cells. Furthermore, we observed CD169+ monocytes in virally-infected patients, including in the blood and bronchoalveolar lavage fluid of COVID-19 patients, as well as in the blood of patients with different types of cancers. Finally, we evaluated two CD169-targeting nanovaccine platforms, antibody-based and liposome-based, and we showed that CD169+ monocytes efficiently presented tumor-associated peptides gp100 and WT1 to antigen-specific CD8+ T cells. In conclusion, our data indicate that CD169+ monocytes are activated monocytes with enhanced CD8+ T cell stimulatory capacity and that they emerge as an interesting target in nanovaccine strategies, because of their presence in health and different diseases.
Collapse
Affiliation(s)
- Alsya J Affandi
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Katarzyna Olesek
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Joanna Grabowska
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Maarten K Nijen Twilhaar
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ernesto Rodríguez
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Anno Saris
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Eline S Zwart
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Esther J Nossent
- Department of Pulmonary Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Amsterdam Cardiovascular Sciences Research Institute, Amsterdam UMC, Amsterdam, Netherlands
| | - Hakan Kalay
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Michael de Kok
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Geert Kazemier
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Johannes Stöckl
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Alfons J M van den Eertwegh
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Juan J Garcia-Vallejo
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Department of Biomaterials, Science and Technology, Faculty of Science and Technology, University of Twente, Enschede, Netherlands.,Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Joke M M den Haan
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
4
|
Soday L, Potts M, Hunter LM, Ravenhill BJ, Houghton JW, Williamson JC, Antrobus R, Wills MR, Matheson NJ, Weekes MP. Comparative Cell Surface Proteomic Analysis of the Primary Human T Cell and Monocyte Responses to Type I Interferon. Front Immunol 2021; 12:600056. [PMID: 33628210 PMCID: PMC7897682 DOI: 10.3389/fimmu.2021.600056] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/13/2021] [Indexed: 12/28/2022] Open
Abstract
The cellular response to interferon (IFN) is essential for antiviral immunity, IFN-based therapy and IFN-related disease. The plasma membrane (PM) provides a critical interface between the cell and its environment, and is the initial portal of entry for viruses. Nonetheless, the effect of IFN on PM proteins is surprisingly poorly understood, and has not been systematically investigated in primary immune cells. Here, we use multiplexed proteomics to quantify IFNα2a-stimulated PM protein changes in primary human CD14+ monocytes and CD4+ T cells from five donors, quantifying 606 and 482 PM proteins respectively. Comparison of cell surface proteomes revealed a remarkable invariance between donors in the overall composition of the cell surface from each cell type, but a marked donor-to-donor variability in the effects of IFNα2a. Furthermore, whereas only 2.7% of quantified proteins were consistently upregulated by IFNα2a at the surface of CD4+ T cells, 6.8% of proteins were consistently upregulated in primary monocytes, suggesting that the magnitude of the IFNα2a response varies according to cell type. Among these differentially regulated proteins, we found the viral target Endothelin-converting enzyme 1 (ECE1) to be an IFNα2a-stimulated protein exclusively upregulated at the surface of CD4+ T cells. We therefore provide a comprehensive map of the cell surface of IFNα2a-stimulated primary human immune cells, including previously uncharacterized interferon stimulated genes (ISGs) and candidate antiviral factors.
Collapse
Affiliation(s)
- Lior Soday
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Martin Potts
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Leah M. Hunter
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Benjamin J. Ravenhill
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Jack W. Houghton
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - James C. Williamson
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Mark R. Wills
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Nicholas J. Matheson
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), University of Cambridge, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge, United Kingdom
| | - Michael P. Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
5
|
Rubino E, Cruciani M, Tchitchek N, Le Tortorec A, Rolland AD, Veli Ö, Vallet L, Gaggi G, Michel F, Dejucq-Rainsford N, Pellegrini S. Human Ubiquitin-Specific Peptidase 18 Is Regulated by microRNAs via the 3'Untranslated Region, A Sequence Duplicated in Long Intergenic Non-coding RNA Genes Residing in chr22q11.21. Front Genet 2021; 11:627007. [PMID: 33633774 PMCID: PMC7901961 DOI: 10.3389/fgene.2020.627007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 12/30/2020] [Indexed: 12/16/2022] Open
Abstract
Ubiquitin-specific peptidase 18 (USP18) acts as gatekeeper of type I interferon (IFN) responses by binding to the IFN receptor subunit IFNAR2 and preventing activation of the downstream JAK/STAT pathway. In any given cell type, the level of USP18 is a key determinant of the output of IFN-stimulated transcripts. How the baseline level of USP18 is finely tuned in different cell types remains ill defined. Here, we identified microRNAs (miRNAs) that efficiently target USP18 through binding to the 3’untranslated region (3’UTR). Among these, three miRNAs are particularly enriched in circulating monocytes which exhibit low baseline USP18. Intriguingly, the USP18 3’UTR sequence is duplicated in human and chimpanzee genomes. In humans, four USP18 3’UTR copies were previously found to be embedded in long intergenic non-coding (linc) RNA genes residing in chr22q11.21 and known as FAM247A-D. Here, we further characterized their sequence and measured their expression profile in human tissues. Importantly, we describe an additional lincRNA bearing USP18 3’UTR (here linc-UR-B1) that is expressed only in testis. RNA-seq data analyses from testicular cell subsets revealed a positive correlation between linc-UR-B1 and USP18 expression in spermatocytes and spermatids. Overall, our findings uncover a set of miRNAs and lincRNAs, which may be part of a network evolved to fine-tune baseline USP18, particularly in cell types where IFN responsiveness needs to be tightly controlled.
Collapse
Affiliation(s)
- Erminia Rubino
- Unit of Cytokine Signaling, Institut Pasteur, INSERM U1221, Paris, France.,École Doctorale Physiologie, Physiopathologie et Thérapeutique, ED394, Sorbonne Université, Paris, France
| | - Melania Cruciani
- Unit of Cytokine Signaling, Institut Pasteur, INSERM U1221, Paris, France
| | - Nicolas Tchitchek
- École Doctorale Physiologie, Physiopathologie et Thérapeutique, ED394, Sorbonne Université, Paris, France.,i3 research unit, Hôpital Pitié-Salpêtrière-Sorbonne Université, Paris, France
| | - Anna Le Tortorec
- UMR_S1085, Institut de recherche en santé, environnement et travail (Irset), EHESP, Inserm, Univ Rennes, Rennes, France
| | - Antoine D Rolland
- UMR_S1085, Institut de recherche en santé, environnement et travail (Irset), EHESP, Inserm, Univ Rennes, Rennes, France
| | - Önay Veli
- Unit of Cytokine Signaling, Institut Pasteur, INSERM U1221, Paris, France
| | - Leslie Vallet
- Unit of Cytokine Signaling, Institut Pasteur, INSERM U1221, Paris, France
| | - Giulia Gaggi
- Unit of Cytokine Signaling, Institut Pasteur, INSERM U1221, Paris, France
| | - Frédérique Michel
- Unit of Cytokine Signaling, Institut Pasteur, INSERM U1221, Paris, France
| | - Nathalie Dejucq-Rainsford
- UMR_S1085, Institut de recherche en santé, environnement et travail (Irset), EHESP, Inserm, Univ Rennes, Rennes, France
| | - Sandra Pellegrini
- Unit of Cytokine Signaling, Institut Pasteur, INSERM U1221, Paris, France
| |
Collapse
|
6
|
Zhou L, Zhang Y, Wang Y, Zhang M, Sun W, Dai T, Wang A, Wu X, Zhang S, Wang S, Zhou F. A Dual Role of Type I Interferons in Antitumor Immunity. ACTA ACUST UNITED AC 2020; 4:e1900237. [PMID: 33245214 DOI: 10.1002/adbi.201900237] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/17/2020] [Accepted: 02/20/2020] [Indexed: 12/12/2022]
Abstract
Type I interferons (IFN-Is) are a family of cytokines that exert direct antiviral effects and regulate innate and adaptive immune responses through direct and indirect mechanisms. It is generally believed that IFN-Is repress tumor development via restricting tumor proliferation and inducing antitumor immune responses. However, recent emerging evidence suggests that IFN-Is play a dual role in antitumor immunity. That is, in the early stage of tumorigenesis, IFN-Is promote the antitumor immune response by enhancing antigen presentation in antigen-presenting cells and activating CD8+ T cells. However, in the late stage of tumor progression, persistent expression of IFN-Is induces the expression of immunosuppressive factors (PD-L1, IDO, and IL-10) on the surface of dendritic cells and other bone marrow cells and inhibits their antitumor immunity. This review outlines these dual functions of IFN-Is in antitumor immunity and elucidates the involved mechanisms, as well as their applications in tumor therapy.
Collapse
Affiliation(s)
- Lili Zhou
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Yuqi Zhang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Yongqiang Wang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Meirong Zhang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Wenhuan Sun
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Tong Dai
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Aijun Wang
- Department of Surgery, School of Medicine, UC Davis, Davis, CA, 95817, USA
| | - Xiaojin Wu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Suping Zhang
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, Base for international Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518055, China
| | - Shuai Wang
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Fangfang Zhou
- Jiangsu Key Laboratory of Infection and Immunity, The Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
7
|
Romoli MR, Di Gennaro P, Gerlini G, Sestini S, Brandani P, Ferrone S, Borgognoni L. High Antigen Processing Machinery component expression in Langerhans cells from melanoma patients' sentinel lymph nodes. Cell Immunol 2017; 320:29-37. [PMID: 28870403 DOI: 10.1016/j.cellimm.2017.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 08/18/2017] [Accepted: 08/26/2017] [Indexed: 11/25/2022]
Abstract
Langerhans cells (LCs) from melanoma patients sentinel lymph nodes (SLN) are poor T cell activators mostly due to an immature immunophenotype. However Antigen Presenting Machinery (APM) role is unknown. We investigated HLA-class I APM components (Delta, LMP-7/10, TAP-1, Calnexin, Tapasin, β2-microglobulin and HLA-A,B,C) in LCs from healthy donors skin and melanoma patients SLN. APM component levels were low in immature epidermal LCs and significantly increased after maturation (p<0.05); their levels were significantly high in SLN LCs (p<0.01). APM component expression correlated with melanoma Breslow's thickness and SLN metastases: HLA-A,B,C level was significantly lower in SLN LCs from thick lesions patients compared with those from thin/intermediate lesions (p<0.05); β2-microglobulin level was significantly higher in positive SLN LCs compared to negative ones (p<0.05). Functionally, SLN LCs did not phagocytose exogenous antigens. These findings extend LCs knowledge indicating that they are not fully impaired by melanoma, contributing to design new LCs-based therapeutic approaches.
Collapse
Affiliation(s)
- Maria Raffaella Romoli
- Plastic and Reconstructive Surgery Unit, Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Tuscan Tumour Institute (ITT) - S.M. Annunziata Hospital, Florence, Italy
| | - Paola Di Gennaro
- Plastic and Reconstructive Surgery Unit, Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Tuscan Tumour Institute (ITT) - S.M. Annunziata Hospital, Florence, Italy; Dept. Surgery and Translational Medicine, Dermatology Section, University of Florence, Florence, Italy.
| | - Gianni Gerlini
- Plastic and Reconstructive Surgery Unit, Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Tuscan Tumour Institute (ITT) - S.M. Annunziata Hospital, Florence, Italy
| | - Serena Sestini
- Plastic and Reconstructive Surgery Unit, Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Tuscan Tumour Institute (ITT) - S.M. Annunziata Hospital, Florence, Italy
| | - Paola Brandani
- Plastic and Reconstructive Surgery Unit, Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Tuscan Tumour Institute (ITT) - S.M. Annunziata Hospital, Florence, Italy
| | - Soldano Ferrone
- Dept. Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lorenzo Borgognoni
- Plastic and Reconstructive Surgery Unit, Regional Melanoma Referral Center and Melanoma & Skin Cancer Unit, Tuscan Tumour Institute (ITT) - S.M. Annunziata Hospital, Florence, Italy
| |
Collapse
|
8
|
Ju X, Silveira PA, Hsu WH, Elgundi Z, Alingcastre R, Verma ND, Fromm PD, Hsu JL, Bryant C, Li Z, Kupresanin F, Lo TH, Clarke C, Lee K, McGuire H, Fazekas de St Groth B, Larsen SR, Gibson J, Bradstock KF, Clark GJ, Hart DNJ. The Analysis of CD83 Expression on Human Immune Cells Identifies a Unique CD83+-Activated T Cell Population. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:4613-4625. [PMID: 27837105 DOI: 10.4049/jimmunol.1600339] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 10/10/2016] [Indexed: 02/02/2023]
Abstract
CD83 is a member of the Ig gene superfamily, first identified in activated lymphocytes. Since then, CD83 has become an important marker for defining activated human dendritic cells (DC). Several potential CD83 mRNA isoforms have been described, including a soluble form detected in human serum, which may have an immunosuppressive function. To further understand the biology of CD83, we examined its expression in different human immune cell types before and after activation using a panel of mouse and human anti-human CD83 mAb. The mouse anti-human CD83 mAbs, HB15a and HB15e, and the human anti-human CD83 mAb, 3C12C, were selected to examine cytoplasmic and surface CD83 expression, based on their different binding characteristics. Glycosylation of CD83, the CD83 mRNA isoforms, and soluble CD83 released differed among blood DC, monocytes, and monocyte-derived DC, and other immune cell types. A small T cell population expressing surface CD83 was identified upon T cell stimulation and during allogeneic MLR. This subpopulation appeared specifically during viral Ag challenge. We did not observe human CD83 on unstimulated human natural regulatory T cells (Treg), in contrast to reports describing expression of CD83 on mouse Treg. CD83 expression was increased on CD4+, CD8+ T, and Treg cells in association with clinical acute graft-versus-host disease in allogeneic hematopoietic cell transplant recipients. The differential expression and function of CD83 on human immune cells reveal potential new roles for this molecule as a target of therapeutic manipulation in transplantation, inflammation, and autoimmune diseases.
Collapse
Affiliation(s)
- Xinsheng Ju
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
| | - Pablo A Silveira
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Wei-Hsun Hsu
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Zehra Elgundi
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
| | - Renz Alingcastre
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
| | - Nirupama D Verma
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
| | - Phillip D Fromm
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Jennifer L Hsu
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Institute of Haematology, Royal Prince Alfred Hospital, University of Sydney, Sydney, New South Wales 2050, Australia
| | - Christian Bryant
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Institute of Haematology, Royal Prince Alfred Hospital, University of Sydney, Sydney, New South Wales 2050, Australia
| | - Ziduo Li
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Fiona Kupresanin
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
| | - Tsun-Ho Lo
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Candice Clarke
- Anatomical Pathology Department, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia; and
| | - Kenneth Lee
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
- Anatomical Pathology Department, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia; and
| | - Helen McGuire
- Centenary Institute, Royal Prince Alfred Hospital, Sydney, New South Wales 2050, Australia
| | | | - Stephen R Larsen
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
- Institute of Haematology, Royal Prince Alfred Hospital, University of Sydney, Sydney, New South Wales 2050, Australia
| | - John Gibson
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
- Institute of Haematology, Royal Prince Alfred Hospital, University of Sydney, Sydney, New South Wales 2050, Australia
| | - Kenneth F Bradstock
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Georgina J Clark
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Derek N J Hart
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia;
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
9
|
Zhao S, Zhang L, Han J, Chu J, Wang H, Chen X, Wang Y, Tun N, Lu L, Bai XF, Yearsley M, Devine S, He X, Yu J. Conformal Nanoencapsulation of Allogeneic T Cells Mitigates Graft-versus-Host Disease and Retains Graft-versus-Leukemia Activity. ACS NANO 2016; 10:6189-200. [PMID: 27224853 PMCID: PMC5514314 DOI: 10.1021/acsnano.6b02206] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Allogeneic transplantation of hematopoietic stem cells (HSC) in combination with T cells has a curative potential for hematopoietic malignancies through graft-versus-leukemia (GVL) effects, but is often compromised by the notorious side effect of graft-versus-host disease (GVHD) resulting from alloreactivity of the donor T cells. Here, we tested if temporary immunoisolation achieved by conformally encapsulating the donor T cells within a biocompatible and biodegradable porous film (∼450 nm in thickness) of chitosan and alginate could attenuate GVHD without compromising GVL. The nanoencapsulation was found not to affect the phenotype of T cells in vitro in terms of size, viability, proliferation, cytokine secretion, and cytotoxicity against tumor cells. Moreover, the porous nature of the nanoscale film allowed the encapsulated T cells to communicate with their environment, as evidenced by their intact capability of binding to antibodies. Lethally irradiated mice transplanted with bone marrow cells (BMCs) and the conformally encapsulated allogeneic T cells exhibited significantly improved survival and reduced GVHD together with minimal liver damage and enhanced engraftment of donor BMCs, compared to the transplantation of BMCs and non-encapsulated allogeneic T cells. Moreover, the conformal nanoencapsulation did not compromise the GVL effect of the donor T cells. These data show that conformal nanoencapsulation of T cells within biocompatible and biodegradable nanoscale porous materials is a potentially safe and effective approach to improve allogeneic HSC transplantation for treating hematological malignancies and possibly other diseases.
Collapse
Affiliation(s)
- Shuting Zhao
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, United States
| | - Lingling Zhang
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Jianfeng Han
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jianhong Chu
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Suzhou Institute of Blood and Marrow Transplantation, Soochow University, Suzhou 215000, China
| | - Hai Wang
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, United States
| | - Xilin Chen
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - Youwei Wang
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - Norm Tun
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - Lanchun Lu
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Xue-Feng Bai
- Department of Pathology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Martha Yearsley
- Department of Pathology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Steven Devine
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
- The James Cancer Hospital, The Ohio State University, Columbus, Ohio 43210, United States
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, United States
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jianhua Yu
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
- The James Cancer Hospital, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
10
|
Jungebluth P, Holzgraefe B, Lim ML, Duru AD, Lundin V, Heldring N, Wiklander OPB, Nordin JZ, Chrobok M, Roderburg C, Sjöqvist S, Anderstam B, Beltrán Rodríguez A, Haag JC, Gustafsson Y, Roddewig KG, Jones P, Wood MJA, Luedde T, Teixeira AI, Hermanson O, Winqvist O, Kalzén H, El Andaloussi S, Alici E, Macchiarini P. Autologous Peripheral Blood Mononuclear Cells as Treatment in Refractory Acute Respiratory Distress Syndrome. Respiration 2015; 90:481-492. [PMID: 26613253 DOI: 10.1159/000441799] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 10/12/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a devastating disorder. Despite enormous efforts in clinical research, effective treatment options are lacking, and mortality rates remain unacceptably high. OBJECTIVES A male patient with severe ARDS showed no clinical improvement with conventional therapies. Hence, an emergent experimental intervention was performed. METHODS We performed intratracheal administration of autologous peripheral blood-derived mononuclear cells (PBMCs) and erythropoietin (EPO). RESULTS We found that after 2 days of initial PBMC/EPO application, lung function improved and extracorporeal membrane oxygenation (ECMO) support was reduced. Bronchoscopy and serum inflammatory markers revealed reduced inflammation. Additionally, serum concentration of miR-449a, b, c and miR-34a, a transient upregulation of E-cadherin and associated chromatin marks in PBMCs indicated airway epithelial differentiation. Extracellular vesicles from PBMCs demonstrated anti-inflammatory capacity in a TNF-α-mediated nuclear factor-x03BA;B in vitro assay. Despite improving respiratory function, the patient died of multisystem organ failure on day 38 of ECMO treatment. CONCLUSIONS This case report provides initial encouraging evidence to use locally instilled PBMC/EPO for treatment of severe refractory ARDS. The observed clinical improvement may partially be due to the anti-inflammatory effects of PBMC/EPO to promote tissue regeneration. Further studies are needed for more in-depth understanding of the underlying mechanisms of in vivo regeneration.
Collapse
Affiliation(s)
- Philipp Jungebluth
- Division of Ear, Nose and Throat, Advanced Center for Translational Regenerative Medicine, Department for Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Niessen A, Heyder P, Krienke S, Blank N, Tykocinski LO, Lorenz HM, Schiller M. Apoptotic-cell-derived membrane microparticles and IFN-α induce an inflammatory immune response. J Cell Sci 2015; 128:2443-53. [PMID: 26034070 DOI: 10.1242/jcs.162735] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 05/27/2015] [Indexed: 12/29/2022] Open
Abstract
A dysregulation in the clearance of apoptotic material is considered a major pathogenetic factor for the emergence of autoimmune diseases. Apoptotic-cell-derived membrane microparticles (AdMPs), which are released from the cell surface during apoptosis, have been implicated in the pathogenesis of autoimmunity. Also of importance are cytokines, such as interferon-α (IFN-α), which is known to be a major player in patients with systemic lupus erythematosus (SLE). This study investigates the combined effect of AdMPs and IFN-α on professional phagocytes. In the presence of IFN-α, phagocytosis of AdMPs by human monocytes was significantly increased in a dose-dependent manner. The combination of AdMPs and raised IFN-α concentrations resulted in an increase in the secretion of pro-inflammatory cytokines and an upregulation of surface molecule expression involved in antigen uptake. In addition, macrophage polarisation was shifted towards a more inflammatory type of cell. The synergism between IFN-α and AdMPs seemed to be mediated by an upregulation of phosphorylated STAT1. Our results indicate that IFN-α, together with AdMPs, amplify the initiation and maintenance of inflammation. This mechanism might especially play a crucial role in disorders with a defective clearance of apoptotic material.
Collapse
Affiliation(s)
- Anna Niessen
- Department of Internal Medicine V, Division of Rheumatology, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Petra Heyder
- Department of Internal Medicine V, Division of Rheumatology, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Stefan Krienke
- Department of Internal Medicine V, Division of Rheumatology, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Norbert Blank
- Department of Internal Medicine V, Division of Rheumatology, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Lars-Oliver Tykocinski
- Department of Internal Medicine V, Division of Rheumatology, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Hanns-Martin Lorenz
- Department of Internal Medicine V, Division of Rheumatology, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Martin Schiller
- Department of Internal Medicine V, Division of Rheumatology, University Hospital Heidelberg, Heidelberg 69120, Germany
| |
Collapse
|
12
|
Sluijter BJR, van den Hout MFCM, Koster BD, van Leeuwen PAM, Schneiders FL, van de Ven R, Molenkamp BG, Vosslamber S, Verweij CL, van den Tol MP, van den Eertwegh AJM, Scheper RJ, de Gruijl TD. Arming the Melanoma Sentinel Lymph Node through Local Administration of CpG-B and GM-CSF: Recruitment and Activation of BDCA3/CD141(+) Dendritic Cells and Enhanced Cross-Presentation. Cancer Immunol Res 2015; 3:495-505. [PMID: 25633713 DOI: 10.1158/2326-6066.cir-14-0165] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 01/22/2015] [Indexed: 11/16/2022]
Abstract
Melanoma-induced suppression of dendritic cells (DC) in the sentinel lymph node (SLN) interferes with the generation of protective antitumor immunity. In an effort to strengthen immune defense against metastatic spread, we performed a three-arm phase II study comprising 28 patients with stage I-II melanoma randomized to receive intradermal injections around the primary tumor excision site of saline or low-dose CpG-B, alone or combined with GM-CSF, before excision of the SLNs. After pathologic examination, 5 patients were diagnosed with stage III melanoma based on the presence of tumor cells in the SLNs. Combined CpG/GM-CSF administration resulted in enhanced maturation of all identifiable conventional (cDC) and plasmacytoid (pDC) DC subsets and selectively induced increased frequencies of SLN-resident BDCA3/CD141(+) cDC subsets that also expressed the C-type lectin receptor CLEC9A. Correlative in vivo analyses and in vitro studies provided evidence that these subsets were derived from BDCA3(+) cDC precursors in the blood that were recruited to the SLNs in a type I IFN-dependent manner and subsequently matured under the combined influence of CpG and GM-CSF. In line with their reported functional abilities, frequencies of in vivo CpG/GM-CSF-induced BDCA3/CD141(+) DCs correlated with increased ex vivo cross-presenting capacity of SLN suspensions. Combined local CpG/GM-CSF delivery thus supports protective antimelanoma immunity through concerted activation of pDC and cDC subsets and recruitment of BDCA3(+) cDC subsets with T cell-stimulatory and cross-priming abilities.
Collapse
Affiliation(s)
- Berbel J R Sluijter
- Department of Surgical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | | | - Bas D Koster
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Paul A M van Leeuwen
- Department of Surgical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Famke L Schneiders
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Rieneke van de Ven
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Barbara G Molenkamp
- Department of Surgical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Saskia Vosslamber
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Cornelis L Verweij
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | | | | | - Rik J Scheper
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
13
|
Paccosi S, Musilli C, Caporale R, Gelli AMG, Guasti D, Clemente AM, Torcia MG, Filippelli A, Romagnoli P, Parenti A. Stimulatory interactions between human coronary smooth muscle cells and dendritic cells. PLoS One 2014; 9:e99652. [PMID: 24932497 PMCID: PMC4059651 DOI: 10.1371/journal.pone.0099652] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 05/16/2014] [Indexed: 01/26/2023] Open
Abstract
Despite inflammatory and immune mechanisms participating to atherogenesis and dendritic cells (DCs) driving immune and non-immune tissue injury response, the interactions between DCs and vascular smooth muscle cells (VSMCs) possibly relevant to vascular pathology including atherogenesis are still unclear. To address this issue, immature DCs (iDCs) generated from CD14+ cells isolated from healthy donors were matured either with cytokines (mDCs), or co-cultured (ccDCs) with human coronary artery VSMCs (CASMCs) using transwell chambers. Co-culture induced DC immunophenotypical and functional maturation similar to cytokines, as demonstrated by flow cytometry and mixed lymphocyte reaction. In turn, factors from mDCs and ccDCs induced CASMC migration. MCP-1 and TNFα, secreted from DCs, and IL-6 and MCP-1, secreted from CASMCs, were primarily involved. mDCs adhesion to CASMCs was enhanced by CASMC pre-treatment with IFNγ and TNFα ICAM-1 and VCAM-1 were involved, since the expression of specific mRNAs for these molecules increased and adhesion was inhibited by neutralizing antibodies to the counter-receptors CD11c and CD18. Adhesion was also inhibited by CASMC pre-treatment with the HMG-CoA-reductase inhibitor atorvastatin and the PPARγ agonist rosiglitazone, which suggests a further mechanism for the anti-inflammatory action of these drugs. Adhesion of DCs to VSMCs was shown also in vivo in rat carotid 7 to 21 days after crush and incision injury. The findings indicate that DCs and VSMCs can interact with reciprocal stimulation, possibly leading to perpetuate inflammation and vascular wall remodelling, and that the interaction is enhanced by a cytokine-rich inflammatory environment and down-regulated by HMGCoA-reductase inhibitors and PPARγ agonists.
Collapse
Affiliation(s)
- Sara Paccosi
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence, Italy
| | - Claudia Musilli
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence, Italy
| | - Roberto Caporale
- Central Laboratory, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | | | - Daniele Guasti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Ann Maria Clemente
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Maria Gabriella Torcia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Amelia Filippelli
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| | - Paolo Romagnoli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Astrid Parenti
- Department of Health Sciences, Clinical Pharmacology and Oncology Unit, University of Florence, Florence, Italy
- * E-mail:
| |
Collapse
|
14
|
Monocyte-derived interferon-alpha primed dendritic cells in the pathogenesis of psoriasis: new pieces in the puzzle. Int Immunopharmacol 2012; 13:215-8. [PMID: 22522054 DOI: 10.1016/j.intimp.2012.04.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 03/21/2012] [Accepted: 04/03/2012] [Indexed: 12/14/2022]
Abstract
Psoriasis is a common chronic inflammatory skin disorder with serious clinical, psychosocial, and economic consequences. There is much evidence that different dendritic cell (DC) subsets, various proinflammatory cytokines and Toll-like receptors (TLRs) have a central role in the pathogenesis of the disease. One of the early events in psoriatic inflammation is the secretion of interferon (IFN)-α by activated plasmacytoid DCs, a special DC subset present in symptomless psoriatic skin. Secreted IFN-α along with other proinflammatory cytokines can lead to monocyte-derived DC (moDC) development, which might contribute to T-helper (Th)1 and Th17 lymphocyte differentiation/activation and to keratinocyte proliferation. Recently it was proven that interleukin (IL)-12 and IL-23 play a critical role in this process. Additionally in psoriatic lesions, Th1 and Th17 lympocytes can interact with monocytes and instruct these cells to differentiate into Th1- and Th17-promoting moDCs, further governing the formation and function of specialized moDC subsets. The concept we present here focuses on the initial and central role of IFN-α, on the importance of other proinflammatory cytokines, on TLR stimulation and on the effect of T lymphocytes in priming moDCs, which may play an important role in initiating and maintaining psoriasis.
Collapse
|
15
|
Abstract
Progress in vector design and an increased knowledge of mechanisms underlying tumor-induced immune suppression have led to a new and promising generation of Adenovirus (Ad)-based immunotherapies, which are discussed in this review. As vaccine vehicles Ad vectors (AdVs) have been clinically evaluated and proven safe, but a major limitation of the commonly used Ad5 serotype is neutralization by preexistent or rapidly induced immune responses. Genetic modifications in the Ad capsid can reduce intrinsic immunogenicity and facilitate escape from antibody-mediated neutralization. Further modification of the Ad hexon and fiber allows for liver and scavenger detargeting and selective targeting of, for example, dendritic cells. These next-generation Ad vaccines with enhanced efficacy are now becoming available for testing as tumor vaccines. In addition, AdVs encoding immune-modulating products may be used to convert the tumor microenvironment from immune-suppressive and proinvasive to proinflammatory, thus facilitating cell-mediated effector functions that can keep tumor growth and invasion in check. Oncolytic AdVs, that selectively replicate in tumor cells and induce an immunogenic form of cell death, can also be armed with immune-activating transgenes to amplify primed antitumor immune responses. These novel immunotherapy strategies, employing highly efficacious AdVs in optimized configurations, show great promise and warrant clinical exploration.
Collapse
|
16
|
López CB, Hermesh T. Systemic responses during local viral infections: type I IFNs sound the alarm. Curr Opin Immunol 2011; 23:495-9. [PMID: 21752617 DOI: 10.1016/j.coi.2011.06.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 06/13/2011] [Accepted: 06/14/2011] [Indexed: 12/24/2022]
Abstract
Type I IFNs are well known for their role in controlling virus replication and spread. Type I IFNs produced by the infected tissue also signal beyond the boundaries of the infection to regulate different elements of the anti-viral immune response. Recent reports show that type I IFNs directly condition naive monocytes residing in the distal bone marrow (BM) and induce the expression of effector molecules in memory T cells, before their recruitment to the infected site. In addition, hematopoietic stem cells (HSCs) were shown to enter the cell cycle in response to systemically distributed type I IFNs. These discoveries expand our understanding of the pleiotropic effects of type I IFNs during infection and highlight the critical role of systemic signals in the development of an effective response to a localized viral infection.
Collapse
Affiliation(s)
- Carolina B López
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
17
|
Regulation of LRRK2 expression points to a functional role in human monocyte maturation. PLoS One 2011; 6:e21519. [PMID: 21738687 PMCID: PMC3124520 DOI: 10.1371/journal.pone.0021519] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 06/02/2011] [Indexed: 01/05/2023] Open
Abstract
Genetic variants of Leucine-Rich Repeat Kinase 2 (LRRK2) are associated with a significantly enhanced risk for Parkinson disease, the second most common human neurodegenerative disorder. Despite major efforts, our understanding of LRRK2 biological function and regulation remains rudimentary. In the present study we analyze LRRK2 mRNA and protein expression in sub-populations of human peripheral blood mononuclear cells (PBMCs). LRRK2 mRNA and protein was found in circulating CD19+ B cells and in CD14+ monocytes, whereas CD4+ and CD8+ T cells were devoid of LRRK2 mRNA. Within CD14+ cells the CD14+CD16+ sub-population of monocytes exhibited high levels of LRRK2 protein, in contrast to CD14+CD16- cells. However both populations expressed LRRK2 mRNA. As CD14+CD16+ cells represent a more mature subset of monocytes, we monitored LRRK2 expression after in vitro treatment with various stress factors known to induce monocyte activation. We found that IFN-γ in particular robustly increased LRRK2 mRNA and protein levels in monocytes concomitant with a shift of CD14+CD16− cells towards CD14+CD16+cells. Interestingly, the recently described LRRK2 inhibitor IN-1 attenuated this shift towards CD14+CD16+ after IFN-γ stimulation. Based on these findings we speculate that LRRK2 might have a role in monocyte maturation. Our results provide further evidence for the emerging role of LRRK2 in immune cells and regulation at the transcriptional and translational level. Our data might also reflect an involvement of peripheral and brain immune cells in the disease course of PD, in line with increasing awareness of the role of the immune system in PD.
Collapse
|
18
|
Farkas A, Kemény L. Interferon-α in the generation of monocyte-derived dendritic cells: recent advances and implications for dermatology. Br J Dermatol 2011; 165:247-54. [PMID: 21410666 DOI: 10.1111/j.1365-2133.2011.10301.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dendritic cells (DCs) have a critical role in antiviral responses, in autoimmune disease pathogenesis and in initiating and maintaining inflammatory skin disorders, and are candidates for cell-based immunotherapeutic approaches for tumours. Recent studies have shown the important role of type I interferons (IFNs) in DC differentiation and activation. In the presence of IFN-α and granulocyte/macrophage colony-stimulating factor monocytes differentiate into DCs referred to as IFN-DCs. In vitro generated IFN-DCs show a partially mature phenotype, are effective in taking up antigens, share features of myeloid DCs, plasmacytoid DCs and natural killer cells, exhibit an enhanced chemotactic response and are capable of migrating to the lymph nodes. IFN-DCs produce several chemokines and cytokines, including T-helper 1 (Th1) mediators belonging to the interleukin-12 family. IFN-DCs stimulate T- and B-cell responses and the production of IFN-γ in mixed lymphocyte reactions and have a capacity to produce IFN-γ themselves. IFN-DCs express several toll-like receptor (TLR) subtypes and TLR ligand stimulation improves their costimulatory molecule expression, increases their Th1 cytokine production and enhances their capacity to stimulate naive T-cell proliferation. Here we review the interaction of IFN-α and monocytes and the role of IFN-DCs in infections, in autoimmunity, in inflammation and in cancer immunotherapy focusing on dermatological conditions.
Collapse
Affiliation(s)
- A Farkas
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary.
| | | |
Collapse
|
19
|
Neagu M, Constantin C, Tanase C. Immune-related biomarkers for diagnosis/prognosis and therapy monitoring of cutaneous melanoma. Expert Rev Mol Diagn 2011; 10:897-919. [PMID: 20964610 DOI: 10.1586/erm.10.81] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Skin melanoma, a life-threatening disease, has a recently reported worldwide increase in incidence, despite primary prevention. Skin melanoma statistics emphasize the need for finding markers related to the immune response of the host. The mechanisms that are able to over-power the local immune surveillance comprise molecules that can be valuable markers for diagnosis and prognosis. This article summarizes the immune markers that can monitor the disease stage and evaluate the efficacy of therapeutic interventions. Recent data regarding immunotherapy are presented in the context of tumor escape from immune surveillance and the immune molecules that are both targets and a means of monitoring. Perspectives for developing immune interventions for skin melanoma management and the position of tissue or soluble immune markers as a diagnostic/prognostic panel are evaluated. State-of-the-art technology is emphasized for developing immune molecular signatures for a complex characterization of the patient's immunological status.
Collapse
Affiliation(s)
- Monica Neagu
- Victor Babes' National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania.
| | | | | |
Collapse
|
20
|
Oosterhoff D, Sluijter BJR, Hangalapura BN, de Gruijl TD. The dermis as a portal for dendritic cell-targeted immunotherapy of cutaneous melanoma. Curr Top Microbiol Immunol 2011; 351:181-220. [PMID: 21681685 DOI: 10.1007/82_2011_136] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Complete surgical excision at an early stage remains the only curative treatment for cutaneous melanoma with few available adjuvant therapy options. Nevertheless, melanoma is a relatively immunogenic tumor type and particularly amenable to immunotherapeutic approaches. A dense network of cutaneous dendritic cells (DC) may account for the reported efficacy of vaccination through the skin and provide an attractive target for the immunotherapy of melanoma. Several phenotypically distinct DC subsets are discernable in the skin, among others, epidermal Langerhans cells and dermal DC. Upon appropriate activation both subsets can efficiently migrate to melanoma-draining lymph nodes (LN) to prime T cell-mediated responses. Unfortunately, from an early stage, melanoma development is characterized by strong immune suppression, facilitating unchecked tumor growth and spread. Particularly the primary tumor site and the first-line tumor-draining LN, the so-called sentinel LN, bear the brunt of this melanoma-induced immune suppression-and these are exactly the sites where anti-melanoma effector T cell responses should be primed by DC in order to prevent early metastasis. Through local immunopotentiation or through DC-targeted vaccination, the dermis may be utilized as a portal to activate DC and kick-start or boost effective T cell-mediated anti-melanoma immunity, even in the face of this immune suppression.
Collapse
Affiliation(s)
- D Oosterhoff
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
21
|
Cunningham AL, Abendroth A, Jones C, Nasr N, Turville S. Viruses and Langerhans cells. Immunol Cell Biol 2010; 88:416-23. [PMID: 20445632 DOI: 10.1038/icb.2010.42] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Langerhans cells (LCs) are the resident dendritic cells (DCs) of epidermis in human mucosal stratified squamous epithelium and the skin. A phenotypically similar DC has recently been discovered as a minor population in the murine dermis. In epidermis, LCs function as sentinel antigen-presenting cells that can capture invading viruses such as herpes simplex virus (HSV), varicella-zoster virus (VZV) and human immunodeficiency virus (HIV). This interaction between LCs and viruses results in highly variable responses, depending on the virus as discussed in this review. For example, HSV induces apoptosis in LCs but HIV does not. LCs seem to be the first in a complex chain of antigen presentation to T cells in lymph nodes for HSV and possibly VZV, or they transport virus to T cells, as described for HIV and maybe VZV. Together with epidermal keratinocytes they may also have a role in the initial innate immune response at the site of infection in the epidermis, although this is not fully known. The full spectrum of biological responses of LCs even to these viruses has yet to be understood and will require complementary studies in human LCs in vitro and in murine models in vivo.
Collapse
Affiliation(s)
- Anthony L Cunningham
- Centre for Virus Research, Westmead Millennium Institute, New South Wales, Australia.
| | | | | | | | | |
Collapse
|
22
|
Abstract
Melanoma, one of the most aggressive forms of human cancer, has undergone an alarming increase in incidence in recent years. Early detection is a prerequisite for proper diagnosis and therapy orientation. Soluble biomarkers are an important tool for early diagnosis. Markers that are associated with melanocyte functions imply the enzymes involved in melanin synthesis and the melanin-related metabolites. Proteins such as autocrine melanocyte cell growth factor and melanoma metastasis suppressor have gained attention in the biomarkers domain. The antimelanoma immune response elicited in patients can not only provide new biomarkers but important therapeutic approaches in specific treatments. All the molecules generated during the metastasis process, invasion of neighboring tissue, angiogenesis, invading lymphatic/blood vessels and establishing new tumors at a distant site, are targets for biomarker discovery.
Collapse
Affiliation(s)
- Monica Neagu
- 'Victor Babes' National Institute of Pathology, Immunology Department, 99-101 Splaiul Independentei, 050096 Bucharest, Romania.
| | | | | | | |
Collapse
|
23
|
Hermesh T, Moltedo B, Moran TM, López CB. Antiviral instruction of bone marrow leukocytes during respiratory viral infections. Cell Host Microbe 2010; 7:343-53. [PMID: 20478536 PMCID: PMC2874206 DOI: 10.1016/j.chom.2010.04.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 02/04/2010] [Accepted: 03/26/2010] [Indexed: 12/19/2022]
Abstract
Respiratory viral infections trigger a robust inflammatory response in the lung, producing cytokines, chemokines, and growth factors that promote infiltration of effector leukocytes. Whereas the role of chemokines and infiltrating leukocytes in antiviral immunity is well studied, the effect that lung cytokines have on leukocytes in distal hematopoietic and lymphoid tissues and their role in antiviral immunity is unknown. We show that, during infection with influenza or Sendai virus, the lung communicates with the sterile bone marrow, the primary site of hematopoiesis, through type I interferons. While in the bone marrow, leukocytes exposed to type I interferons activate an antiviral transcriptional program and become resistant to infection with different viruses. The protected bone marrow leukocytes are capable of migrating to the infected lung and contribute to virus clearance. These findings show that appropriate instruction of cells during their development in the bone marrow is needed for effective control of infection.
Collapse
Affiliation(s)
- Tamar Hermesh
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Bruno Moltedo
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Thomas M. Moran
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | - Carolina B. López
- Department of Microbiology and Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| |
Collapse
|
24
|
Goetzl EJ, Huang MC, Kon J, Patel K, Schwartz JB, Fast K, Ferrucci L, Madara K, Taub DD, Longo DL. Gender specificity of altered human immune cytokine profiles in aging. FASEB J 2010; 24:3580-9. [PMID: 20453111 DOI: 10.1096/fj.10-160911] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cytokine generation by T cells and monocytes was determined for 50 subjects aged 65 yr or older and concurrently studied young subjects individually matched to each old subject for sex, race, and national origin. Highly significant differences between cytokine levels of old and young subjects all were gender specific. For T cells stimulated with anti-CD3 plus anti-CD28 antibodies, mean ratios of IFN-gamma generation for healthy old to young subjects were 0.22 for men (P<0.001; n=15) and 3.35 for women (P<0.001; n=13), and those of IL-17 were 0.30 for men (P<0.001) and no difference for women. CD8 T cells were the source of high IFN-gamma in healthy old women. For old men with an inflammatory or immune disease (n=10), mean old to young ratios of T-cell-generated IFN-gamma and IL-17 increased with disease severity up to 5.78 and 2.97 (both P<0.01), respectively, without changes for old women with similar diseases (n=12). For differentiated LPS-stimulated monocytes, old to young ratios of TNF-alpha and IL-6 generation were high only in women with immune or inflammatory disease (2.38, P<0.05 and 1.62, P<0.01, respectively), whereas ratios of IFN-gamma-evoked IP-10 chemokine were low in all groups. Alterations in immune cytokine profiles with aging show significant gender specificity.
Collapse
Affiliation(s)
- Edward J Goetzl
- Department of Medicine, University of California, San Francisco, CA 94143-0711, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Huch JH, Cunningham AL, Arvin AM, Nasr N, Santegoets SJAM, Slobedman E, Slobedman B, Abendroth A. Impact of varicella-zoster virus on dendritic cell subsets in human skin during natural infection. J Virol 2010; 84:4060-72. [PMID: 20130046 PMCID: PMC2849518 DOI: 10.1128/jvi.01450-09] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Accepted: 01/08/2010] [Indexed: 01/07/2023] Open
Abstract
Varicella-zoster virus (VZV) causes varicella and herpes zoster, diseases characterized by distinct cutaneous rashes. Dendritic cells (DC) are essential for inducing antiviral immune responses; however, the contribution of DC subsets to immune control during natural cutaneous VZV infection has not been investigated. Immunostaining showed that compared to normal skin, the proportion of cells expressing DC-SIGN (a dermal DC marker) or DC-LAMP and CD83 (mature DC markers) were not significantly altered in infected skin. In contrast, the frequency of Langerhans cells was significantly decreased in VZV-infected skin, whereas there was an influx of plasmacytoid DC, a potent secretor of type I interferon (IFN). Langerhans cells and plasmacytoid DC in infected skin were closely associated with VZV antigen-positive cells, and some Langerhans cells and plasmacytoid DC were VZV antigen positive. To extend these in vivo observations, both plasmacytoid DC (PDC) isolated from human blood and Langerhans cells derived from MUTZ-3 cells were shown to be permissive to VZV infection. In VZV-infected PDC cultures, significant induction of alpha IFN (IFN-alpha) did not occur, indicating the VZV inhibits the capacity of PDC to induce expression of this host defense cytokine. This study defines changes in the response of DC which occur during cutaneous VZV infection and implicates infection of DC subtypes in VZV pathogenesis.
Collapse
Affiliation(s)
- Jennifer H. Huch
- Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, New South Wales 2006, Australia, Centre For Virus Research, Westmead Millennium Institute and University of Sydney, Westmead, New South Wales 2145, Australia, Departments of Pediatrics and Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam 1081HV, Netherlands, Laverty Pathology, North Ryde, New South Wales, 2113, Australia
| | - Anthony L. Cunningham
- Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, New South Wales 2006, Australia, Centre For Virus Research, Westmead Millennium Institute and University of Sydney, Westmead, New South Wales 2145, Australia, Departments of Pediatrics and Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam 1081HV, Netherlands, Laverty Pathology, North Ryde, New South Wales, 2113, Australia
| | - Ann M. Arvin
- Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, New South Wales 2006, Australia, Centre For Virus Research, Westmead Millennium Institute and University of Sydney, Westmead, New South Wales 2145, Australia, Departments of Pediatrics and Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam 1081HV, Netherlands, Laverty Pathology, North Ryde, New South Wales, 2113, Australia
| | - Najla Nasr
- Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, New South Wales 2006, Australia, Centre For Virus Research, Westmead Millennium Institute and University of Sydney, Westmead, New South Wales 2145, Australia, Departments of Pediatrics and Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam 1081HV, Netherlands, Laverty Pathology, North Ryde, New South Wales, 2113, Australia
| | - Saskia J. A. M. Santegoets
- Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, New South Wales 2006, Australia, Centre For Virus Research, Westmead Millennium Institute and University of Sydney, Westmead, New South Wales 2145, Australia, Departments of Pediatrics and Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam 1081HV, Netherlands, Laverty Pathology, North Ryde, New South Wales, 2113, Australia
| | - Eric Slobedman
- Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, New South Wales 2006, Australia, Centre For Virus Research, Westmead Millennium Institute and University of Sydney, Westmead, New South Wales 2145, Australia, Departments of Pediatrics and Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam 1081HV, Netherlands, Laverty Pathology, North Ryde, New South Wales, 2113, Australia
| | - Barry Slobedman
- Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, New South Wales 2006, Australia, Centre For Virus Research, Westmead Millennium Institute and University of Sydney, Westmead, New South Wales 2145, Australia, Departments of Pediatrics and Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam 1081HV, Netherlands, Laverty Pathology, North Ryde, New South Wales, 2113, Australia
| | - Allison Abendroth
- Discipline of Infectious Diseases and Immunology, University of Sydney, Sydney, New South Wales 2006, Australia, Centre For Virus Research, Westmead Millennium Institute and University of Sydney, Westmead, New South Wales 2145, Australia, Departments of Pediatrics and Microbiology and Immunology, Stanford University School of Medicine, Stanford, California 94305, Department of Pathology, VU University Medical Center, De Boelelaan 1117, Amsterdam 1081HV, Netherlands, Laverty Pathology, North Ryde, New South Wales, 2113, Australia
| |
Collapse
|
26
|
SEIPEL DANIELE, RIBEIRO-Gomes FLAVIALIMA, BARCELOS MICHELLEWILLMEN, RAMALHO ANDRÉVILLAÇA, KANASHIRO MILTONM, KIPNIS THEREZALIBERMAN, ARNHOLDT ANDREACRISTINAVETO. Monocytes/macrophages infected withToxoplasma gondiido not increase co-stimulatory molecules while maintaining their migratory ability. APMIS 2009; 117:672-80. [DOI: 10.1111/j.1600-0463.2009.02519.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
27
|
Poole JA, Thiele GM, Alexis NE, Burrell AM, Parks C, Romberger DJ. Organic dust exposure alters monocyte-derived dendritic cell differentiation and maturation. Am J Physiol Lung Cell Mol Physiol 2009; 297:L767-76. [PMID: 19648285 DOI: 10.1152/ajplung.00107.2009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Organic dust exposure in agricultural animal environments results in airway diseases. Dendritic cells (DCs) orchestrate inflammatory immune response in the airways, but little is known about how organic dust affects differentiation and maturation of monocyte-derived immature and mature DCs (iDCs, mDCs). Peripheral blood monocytes were differentiated in vitro into iDCs with granulocyte-macrophage colony stimulating factor + IL-4 (6 days) with and without swine facility organic dust extract (ODE, 0.1%). Unlike control iDCs, ODE-conditioned iDCs maintained key monocyte properties (increased mCD14, increased phagocytic ability) while expressing DC features [increased mCD83, HLA-DR, CD80, CD86, diminished cytokine (TNF-alpha, IL-6) responsiveness]. At day 6, iDCs were cultured for an additional 48 h (days 7 and 8) with lipopolysaccharide (LPS) to induce mDCs. ODE-conditioned mDCs maintained high expression of mCD14(+) and elevated phagocytosis while their DC features weakened as evidenced by decreased CD11c, CD83, HLA-DR, CD86, and CCR7 expression and reduced lymphocyte-stimulating capacity. Similar results were observed when monocytes were exposed to ODE for only the first 48 h and with ODE depleted of endotoxin. Control iDCs exposed to ODE during the final 2 days of iDC maturation (days 7 and 8) did not differ from control (no ODE) iDCs in surface marker expression and phagocytic ability, but exhibited enhanced lymphocyte-stimulating capacity. Dust exposure alters monocyte differentiation to iDCs and prevents maturation of iDC to mDCs. The first 48 h of monocyte differentiation appears to be the susceptible period to exposure. Environmental exposures present during early monocyte differentiation may impact the critical balance of DCs in the lung.
Collapse
Affiliation(s)
- Jill A Poole
- Omaha Veterans Administration Medical Center, Omaha, Nebraska, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Li H, Ciric B, Yang J, Xu H, Fitzgerald DC, Elbehi M, Fonseca-Kelly Z, Yu S, Zhang GX, Rostami A. Intravenous tolerance modulates macrophage classical activation and antigen presentation in experimental autoimmune encephalomyelitis. J Neuroimmunol 2009; 208:54-60. [PMID: 19187972 DOI: 10.1016/j.jneuroim.2009.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 12/24/2008] [Accepted: 01/05/2009] [Indexed: 12/11/2022]
Abstract
Macrophages act as the first line of self defense by mounting an inflammatory response to antigen and as antigen presenting cells to initiate the adaptive immune response. Inhibition of macrophage activation is one of the possible approaches to modulate inflammation. Intravenous (i.v.) tolerance has proved to be an effective method for ameliorating experimental autoimmune diseases. Whether macrophages are involved in tolerance induction is still largely undefined. In the present study we found that i.v. tolerance induction resulted in lower B7.1, B7.2 and MHC class II molecules, and reduced phagocytosis by both peritoneal macrophages and adherent splenocytes. Macrophages from tolerized mice were associated with a significantly impaired response of MOG-sensitized T cells to MOG. Macrophages from tolerized mice produced low levels of pro-inflammatory molecules IL-12, TNF-alpha, IL-1beta, RANTES and MCP-1 and high levels of IL-10 and TGF-beta. Administration of anti-TGF-beta led to a reduction of IL-10 in tolerized mice. Thus, i.v. tolerance inhibits macrophage classical activation and APC function, increases macrophage alternative activation and IL-10 and TGF-beta production. These cytokines, in turn, induce enhanced production of IL-10 in macrophages in MOG i.v. mice.
Collapse
Affiliation(s)
- Hongmei Li
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|