1
|
Gehlhausen JR, Kong Y, Baker E, Ramachandran S, Koumpouras F, Ko CJ, Vesely M, Little AJ, Damsky W, King B, Iwasaki A. Cutaneous lupus features specialized stromal niches and altered retroelement expression. J Invest Dermatol 2025:S0022-202X(25)00488-9. [PMID: 40409678 DOI: 10.1016/j.jid.2025.04.033] [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: 09/29/2024] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 05/25/2025]
Abstract
Cutaneous Lupus is an inflammatory skin disease causing highly morbid inflamed skin and hair loss. In order to investigate the pathophysiology of cutaneous lupus, we performed single-cell RNA and spatial sequencing of lesional and non-lesional cutaneous lupus skin compared to healthy controls. Pathway enrichment analyses of lesional keratinocytes revealed elevated responses to type I interferon, type II interferon, tumor necrosis factor, and apoptotic signaling. Detailed clustering demonstrated unique fibroblasts specific to lupus skin with likely roles in inflammatory cell recruitment and fibrosis. We also evaluated the association of retroelement expression with type I interferons in the skin. We observed increased retroelement expression which correlated with interferon-stimulated genes across multiple cell types. Moreover, we saw elevated expression of genes involved in RIG-I and cGAS-STING pathways, which transduce elevated nucleic acid signals. Treatment of active cutaneous lupus with Anifrolumab reduced RIG-I and cGAS-STING pathways in addition to the most abundant retroelement family, L2b. Our studies better define type I interferon-mediated immunopathology in cutaneous lupus and identify an association between retroelement expression and interferon signatures in cutaneous lupus.
Collapse
Affiliation(s)
| | - Yong Kong
- Yale School of Public Health; New Haven, CT, USA
| | - Emily Baker
- Yale Department of Dermatology; New Haven, CT, USA
| | | | | | - Christine J Ko
- Yale Department of Dermatology; New Haven, CT, USA; Yale Department of Pathology; New Haven, CT, USA
| | | | | | - William Damsky
- Yale Department of Dermatology; New Haven, CT, USA; Yale Department of Pathology; New Haven, CT, USA
| | - Brett King
- Yale Department of Dermatology; New Haven, CT, USA
| | - Akiko Iwasaki
- Yale Department of Dermatology; New Haven, CT, USA; Yale Department of Immunobiology; New Haven, CT, USA; Department of Molecular Cellular and Developmental Biology, Yale University; New Haven, CT, USA; Howard Hughes Medical Institute; Chevy Chase, MD, USA.
| |
Collapse
|
2
|
Alvarez K, Anacona CA, Agudelo ER, Losada P, Orozco VH, Giraldo LF, Vásquez G, Rodriguez D, Díaz JC, Pineda R, Rojas M. Wheat germ agglutinin-nanoparticles encapsulating itacitinib target and suppress pro-inflammatory slan+ monocytes. Nanomedicine (Lond) 2025; 20:1113-1126. [PMID: 40289864 PMCID: PMC12068337 DOI: 10.1080/17435889.2025.2494500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 04/14/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND 6-sulfoLacNAc (slan)+ monocytes, a non-classical monocyte subset, play a pro-inflammatory role in autoimmune diseases like systemic lupus erythematosus (SLE). This study evaluates the therapeutic potential of itacitinib (ITA) encapsulated in wheat germ agglutinin-functionalized nanoparticles (WGA/F127/PNPs) to target and inhibit the JAK-STAT pathway in slan+ monocytes. METHODS We prepared ITA-loaded WGA/F127/PNPs and analyzed their binding and internalization in various leukocyte subsets using flow cytometry, focusing on slan+ and slan- monocytes. Further, peripheral blood samples from healthy controls (n = 37) and SLE patients (n = 50) were used to assess slan+ monocyte phenotypes. Co-cultures of slan+ and slan- monocytes stimulated with LPS revealed that slan+ monocytes significantly increased HLA-DR expression. RESULTS Results showed that slan+ monocytes from SLE patients were reduced compared to healthy controls (p < 0.001) and that slan+ monocytes effectively internalized WGA/F127/PNPs, unlike slan- cells. ITA-loaded nanoparticles decreased HLA-DR, CD69, and CD86 expression, STAT1 phosphorylation, and cytokine production in IFN-γ-stimulated slan+ monocytes. Findings support WGA/F127/PNPs as a promising drug delivery system for targeting slan+ monocytes, providing new therapeutic potential for SLE. CONCLUSION ITA-loaded WGA/F127/PNPs effectively target and suppress pro-inflammatory slan+ monocytes, presenting a promising, cell-specific therapeutic approach for managing systemic lupus erythematosus and related autoimmune disorders.
Collapse
Affiliation(s)
- Karen Alvarez
- Cellular Immunology and Immunogenetics Group (GICIG), Faculty of Medicine, University of Antioquia, Medellin, Colombia
| | - Cristian A. Anacona
- Cellular Immunology and Immunogenetics Group (GICIG), Faculty of Medicine, University of Antioquia, Medellin, Colombia
| | | | - Paula Losada
- Cellular Immunology and Immunogenetics Group (GICIG), Faculty of Medicine, University of Antioquia, Medellin, Colombia
| | - Víctor H. Orozco
- Polymer Research Laboratory, University of Antioquia, Medellin, Colombia
| | | | - Gloria Vásquez
- Cellular Immunology and Immunogenetics Group (GICIG), Faculty of Medicine, University of Antioquia, Medellin, Colombia
- Rheumatology Service, Hospital Universitario San Vicente Fundación, Medellin, Colombia
| | - Daniel Rodriguez
- ARTMEDICA Health Service Provider, Medellin, Antioquia, Colombia
| | - Juan Camilo Díaz
- ARTMEDICA Health Service Provider, Medellin, Antioquia, Colombia
| | - Ricardo Pineda
- ARTMEDICA Health Service Provider, Medellin, Antioquia, Colombia
| | - Mauricio Rojas
- Cellular Immunology and Immunogenetics Group (GICIG), Faculty of Medicine, University of Antioquia, Medellin, Colombia
- Flow Cytometry Core, University Research Headquarters (SIU), University of Antioquia, Medellin, Colombia
| |
Collapse
|
3
|
Dyba M, Berezenko V, Zabara D, Bezpala A, Donskoi B. Monocyte subpopulations in children with autoimmune liver disease. Pathol Res Pract 2024; 263:155622. [PMID: 39357182 DOI: 10.1016/j.prp.2024.155622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Patients with autoimmune liver diseases require individualized long-term immunosuppressive therapy, whose discontinuation is possible after complete histological remission and that requires repeated liver biopsy. In view of this, the search for non-invasive markers is essential for patients with autoimmune liver disease. PURPOSE The purpose of this research is to assess the possibility of predicting the recurrence of autoimmune liver disease in children. METHOD The biological material used in the study was blood serum from 80 children diagnosed with autoimmune hepatitis and autoimmune sclerosing cholangitis. Patients were divided into four groups according to disease activity and therapeutic approach. RESULTS The percentage of monocyte subpopulations was determined by flow cytometry, and disease activity, inflammation, and fibrosis markers were analyzed to study the relationship and diagnostic value of the parameters studied in detail. The results of the study indicate a significant relationship between disease activity and changes in the distribution of the percentage of monocyte subpopulations in the blood. The percentage of intermediate CD14++/CD16+ monocytes was found to correlate with disease activity, and non-classical CD14lowCD16+ monocytes were found to be of high diagnostic value in the diagnosis of disease relapse. CONCLUSIONS These findings not only expand the understanding of the pathogenesis of autoimmune liver disease but also point to the prospects of using monocyte subpopulations as potential biomarkers for predicting relapse, contributing to the development of more effective clinical management strategies.
Collapse
Affiliation(s)
- Maryna Dyba
- Department of Hepatology and Comorbidities in Children, Institute of Pediatrics, Obstetrics and Gynecology of the National Academy of Medical Sciences of Ukraine, Kyiv 04050, Ukraine.
| | - Valentyna Berezenko
- Department of Hepatology and Comorbidities in Children, Institute of Pediatrics, Obstetrics and Gynecology of the National Academy of Medical Sciences of Ukraine, Kyiv 04050, Ukraine
| | - Dariia Zabara
- Laboratory of Immunology, Institute of Pediatrics, Obstetrics and Gynecology of the National Academy of Medical Sciences of Ukraine, Kyiv 04050, Ukraine
| | - Anna Bezpala
- Department of Hepatology and Comorbidities in Children, Institute of Pediatrics, Obstetrics and Gynecology of the National Academy of Medical Sciences of Ukraine, Kyiv 04050, Ukraine
| | - Boris Donskoi
- Laboratory of Immunology, Institute of Pediatrics, Obstetrics and Gynecology of the National Academy of Medical Sciences of Ukraine, Kyiv 04050, Ukraine
| |
Collapse
|
4
|
Preuß SL, Oehrl S, Zhang H, Döbel T, Engel U, Young JL, Spatz JP, Schäkel K. Immune complex-induced haptokinesis in human non-classical monocytes. Front Immunol 2023; 14:1078241. [PMID: 36936904 PMCID: PMC10014541 DOI: 10.3389/fimmu.2023.1078241] [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: 10/24/2022] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Formation and deposition of immune complexes (ICs) are hallmarks of various autoimmune diseases. Detection of ICs by IC receptors on leukocytes induces downstream signaling and shapes the local immune response. In many cases the pathological relevance of ICs is not well understood. We here show that ICs induce a distinct migratory response, i.e. haptokinesis in 6-sulfo LacNAc+ monocytes (slanMo) and in non-classical monocytes (ncMo) but not in intermediate (imMo) and classical monocytes (cMo). Using live imaging combined with automated cell tracking, we show that the main features of IC-dependent haptokinesis are elongation of the cell body, actin polarization at the leading edge, and highly directional migration. We find that CD16-dependent signaling mediates haptokinesis as blocking of CD16 or blocking SYK-signaling inhibited the migratory response. The activity of the metalloproteinase ADAM17 also modifies IC-dependent haptokinesis, likely at least partially via cleavage of CD16. Furthermore, using matrices with defined ligand spacing, we show that ligand density impacts the magnitude of the migratory response. Taken together, we have demonstrated that ICs induce a specific migratory response in ncMo but not in other monocyte subsets. Therefore, our work lays the groundwork for the investigation of IC-dependent haptokinesis in ncMo as a potential pathomechanism in IC-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Sophie L. Preuß
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stephanie Oehrl
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Hao Zhang
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Döbel
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Ulrike Engel
- Nikon Imaging Center, Heidelberg University, Heidelberg, Germany
| | - Jennifer L. Young
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Heidelberg, Germany
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Biomedical Engineering Department, National University of Singapore, Singapore, Singapore
| | - Joachim P. Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Heidelberg, Germany
- Department of Biophysical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Knut Schäkel
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
- *Correspondence: Knut Schäkel,
| |
Collapse
|
5
|
Mahroum N, Elsalti A, Alwani A, Seida I, Alrais M, Seida R, Esirgun SN, Abali T, Kiyak Z, Zoubi M, Shoenfeld Y. The mosaic of autoimmunity - Finally discussing in person. The 13 th international congress on autoimmunity 2022 (AUTO13) Athens. Autoimmun Rev 2022; 21:103166. [PMID: 35932955 PMCID: PMC9349027 DOI: 10.1016/j.autrev.2022.103166] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 07/31/2022] [Indexed: 11/29/2022]
Abstract
While autoimmunity is a branch of medicine linked to every single organ system via direct and indirect pathways, meeting in person to discuss autoimmunity during the 13th international congress on autoimmunity (AUTO13) with participants from all over the world had a very good reason. The mechanisms involved in autoimmune diseases are of extreme importance and in fact critical in understanding the course of diseases as well as selecting proper therapies. COVID-19 has served as a great example of how autoimmunity is deeply involved in the disease and directly correlated to severity, morbidity, and mortality. For instance, initially the term cytokine storm dominated, then COVID-19 was addressed as the new member of the hyperferritinemic syndrome, and also the use of immunosuppressants in patients with COVID-19 throughout the pandemic, all shed light on the fundamental role of autoimmunity. Unsurprisingly, SARS-CoV-2 was called the “autoimmune virus” during AUTO13. Subsequently, the correlation between autoimmunity and COVID-19 vaccines and post-COVID, all were discussed from different autoimmune aspects during the congress. In addition, updates on the mechanisms of diseases, autoantibodies, novel diagnostics and therapies in regard to autoimmune diseases such as antiphospholipid syndrome, systemic lupus erythematosus, systemic sclerosis and others, were discussed in dedicated sessions. Due to the magnificence of the topics discussed, we aimed to bring in our article hereby, the pearls of AUTO13 in terms of updates, new aspects of autoimmunity, and interesting findings. While more than 500 abstract were presented, concluding all the topics was not in reach, hence major findings were summarized.
Collapse
Affiliation(s)
- Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey.
| | - Abdulrahman Elsalti
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Abdulkarim Alwani
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Isa Seida
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Mahmoud Alrais
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ravend Seida
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Sevval Nil Esirgun
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Tunahan Abali
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Zeynep Kiyak
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Majdi Zoubi
- Department of Internal Medicine B, HaEmek Medical Center, Afula, Israel, Affiliated to Technion, Faculty of Medicine, Haifa, Israel
| | | |
Collapse
|
6
|
Ricard L, Eshagh D, Siblany L, de Vassoigne F, Malard F, Laurent C, Beurier P, Jachiet V, Rivière S, Fain O, Mohty M, Gaugler B, Mekinian A. 6-sulfo LacNAc monocytes are quantitatively and functionally disturbed in systemic sclerosis patients. Clin Exp Immunol 2022; 209:175-181. [PMID: 35758259 DOI: 10.1093/cei/uxac059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/17/2022] [Accepted: 06/24/2022] [Indexed: 11/14/2022] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease characterized by fibrosis, microangiopathy and autoantibodies. We previously reported that circulating follicular helper T (cTfh) cells are increased in SSc and induce plasmablast differentiation. However, mechanisms leading to cTfh cell expansion and activation in SSc remain to be established. Tfh cells require IL-12 for their expansion and differentiation. 6-sulfo LacNAc monocytes (slanMo), a subset of monocytes, have a higher capacity to produce IL-12 and to induce CD4 + T cell proliferation in comparison with dendritic cells (DC) or classical monocytes. The aim of this study was to perform a quantitative and functional analysis of monocytes and DC and to correlate them with cTfh cell expansion and clinical manifestations in SSc. Using flow cytometry, we analyzed different monocyte subsets including slanMo and DC from 36 SSc patients and 26 healthy controls (HC). In vitro culture experiments of sorted slanMo were performed for functional analysis and cytokine production. We observed that slanMo, intermediate and non-classical monocytes were increased in SSc in comparison with HC. Furthermore, the increase in slanMo cells was more potent in patients with diffuse SSc. We observed a significant positive correlation between slanMo and cTfh cell levels in SSc patients but not in HC. Other monocyte subsets did not correlate with cTfh cell expansion. In addition, we observed that in vitro, slanMo cells from SSc patients produced less IL-12 than slanMo from HC. SlanMo are increased in SSc and may participate in the activation of cTfh cells in SSc.
Collapse
Affiliation(s)
- Laure Ricard
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie clinique, F-75012, Paris, France
| | - Déborah Eshagh
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France.,AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne et de l'Inflammation-(DHU i2B), F-75012, Paris, France
| | - Lama Siblany
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie clinique, F-75012, Paris, France
| | - Frédéric de Vassoigne
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie clinique, F-75012, Paris, France
| | - Florent Malard
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie clinique, F-75012, Paris, France
| | - Charlotte Laurent
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France.,AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne et de l'Inflammation-(DHU i2B), F-75012, Paris, France
| | - Pauline Beurier
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France
| | - Vincent Jachiet
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France.,AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne et de l'Inflammation-(DHU i2B), F-75012, Paris, France
| | - Sébastien Rivière
- AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne et de l'Inflammation-(DHU i2B), F-75012, Paris, France
| | - Olivier Fain
- AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne et de l'Inflammation-(DHU i2B), F-75012, Paris, France
| | - Mohamad Mohty
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie clinique, F-75012, Paris, France
| | - Béatrice Gaugler
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie clinique, F-75012, Paris, France
| | - Arsène Mekinian
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France.,AP-HP, Hôpital Saint-Antoine, Service de Médecine Interne et de l'Inflammation-(DHU i2B), F-75012, Paris, France
| |
Collapse
|
7
|
Billi AC, Ma F, Plazyo O, Gharaee-Kermani M, Wasikowski R, Hile GA, Xing X, Yee CM, Rizvi SM, Maz MP, Berthier CC, Wen F, Tsoi LC, Pellegrini M, Modlin RL, Gudjonsson JE, Kahlenberg JM. Nonlesional lupus skin contributes to inflammatory education of myeloid cells and primes for cutaneous inflammation. Sci Transl Med 2022; 14:eabn2263. [PMID: 35476593 PMCID: PMC9169615 DOI: 10.1126/scitranslmed.abn2263] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cutaneous lupus erythematosus (CLE) is a disfiguring and poorly understood condition frequently associated with systemic lupus. Previous studies suggest that nonlesional keratinocytes play a role in disease predisposition, but this has not been investigated in a comprehensive manner or in the context of other cell populations. To investigate CLE immunopathogenesis, normal-appearing skin, lesional skin, and circulating immune cells from lupus patients were analyzed via integrated single-cell RNA sequencing and spatial RNA sequencing. We demonstrate that normal-appearing skin of patients with lupus represents a type I interferon-rich, prelesional environment that skews gene transcription in all major skin cell types and markedly distorts predicted cell-cell communication networks. We also show that lupus-enriched CD16+ dendritic cells undergo robust interferon education in the skin, thereby gaining proinflammatory phenotypes. Together, our data provide a comprehensive characterization of lesional and nonlesional skin in lupus and suggest a role for skin education of CD16+ dendritic cells in CLE pathogenesis.
Collapse
Affiliation(s)
| | - Feiyang Ma
- Dept of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Olesya Plazyo
- Dept of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Mehrnaz Gharaee-Kermani
- Dept of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Grace A. Hile
- Dept of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Xianying Xing
- Dept of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Christine M. Yee
- Dept of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Syed M. Rizvi
- Dept of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mitra P. Maz
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Celine C. Berthier
- Division of Nephrology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Fei Wen
- Dept of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Lam C. Tsoi
- Dept of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Matteo Pellegrini
- Dept of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Robert L. Modlin
- Dept of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | | | - J. Michelle Kahlenberg
- Dept of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Ligotti ME, Pojero F, Accardi G, Aiello A, Caruso C, Duro G, Candore G. Immunopathology and Immunosenescence, the Immunological Key Words of Severe COVID-19. Is There a Role for Stem Cell Transplantation? Front Cell Dev Biol 2021; 9:725606. [PMID: 34595175 PMCID: PMC8477205 DOI: 10.3389/fcell.2021.725606] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/06/2021] [Indexed: 01/08/2023] Open
Abstract
The outcomes of Coronavirus disease-2019 (COVID-19) vary depending on the age, health status and sex of an individual, ranging from asymptomatic to lethal. From an immunologic viewpoint, the final severe lung damage observed in COVID-19 should be caused by cytokine storm, driven mainly by interleukin-6 and other pro-inflammatory cytokines. However, which immunopathogenic status precedes this "cytokine storm" and why the male older population is more severely affected, are currently unanswered questions. The aging of the immune system, i.e., immunosenescence, closely associated with a low-grade inflammatory status called "inflammageing," should play a key role. The remodeling of both innate and adaptive immune response observed with aging can partly explain the age gradient in severity and mortality of COVID-19. This review discusses how aging impacts the immune response to the virus, focusing on possible strategies to rejuvenate the immune system with stem cell-based therapies. Indeed, due to immunomodulatory and anti-inflammatory properties, multipotent mesenchymal stem cells (MSCs) are a worth-considering option against COVID-19 adverse outcomes.
Collapse
Affiliation(s)
- Mattia Emanuela Ligotti
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- Institute for Biomedical Research and Innovation, National Research Council of Italy, Palermo, Italy
| | - Fanny Pojero
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- International Society on Aging and Disease, Fort Worth, TX, United States
| | - Giovanni Duro
- Institute for Biomedical Research and Innovation, National Research Council of Italy, Palermo, Italy
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| |
Collapse
|
9
|
Berndt N, Bippes CC, Michalk I, Bartsch T, Arndt C, Puentes-Cala E, Soto JA, Loureiro LR, Kegler A, Bachmann D, Gross JK, Gross T, Kurien BT, Scofield RH, Farris AD, James JA, Bergmann R, Schmitz M, Feldmann A, Bachmann MP. And Yet It Moves: Oxidation of the Nuclear Autoantigen La/SS-B Is the Driving Force for Nucleo-Cytoplasmic Shuttling. Int J Mol Sci 2021; 22:9699. [PMID: 34575862 PMCID: PMC8470643 DOI: 10.3390/ijms22189699] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 01/10/2023] Open
Abstract
Decades ago, we and many other groups showed a nucleo-cytoplasmic translocation of La protein in cultured cells. This shuttling of La protein was seen after UV irradiation, virus infections, hydrogen peroxide exposure and the Fenton reaction based on iron or copper ions. All of these conditions are somehow related to oxidative stress. Unfortunately, these harsh conditions could also cause an artificial release of La protein. Even until today, the shuttling and the cytoplasmic function of La/SS-B is controversially discussed. Moreover, the driving mechanism for the shuttling of La protein remains unclear. Recently, we showed that La protein undergoes redox-dependent conformational changes. Moreover, we developed anti-La monoclonal antibodies (anti-La mAbs), which are specific for either the reduced form of La protein or the oxidized form. Using these tools, here we show that redox-dependent conformational changes are the driving force for the shuttling of La protein. Moreover, we show that translocation of La protein to the cytoplasm can be triggered in a ligand/receptor-dependent manner under physiological conditions. We show that ligands of toll-like receptors lead to a redox-dependent shuttling of La protein. The shuttling of La protein depends on the redox status of the respective cell type. Endothelial cells are usually resistant to the shuttling of La protein, while dendritic cells are highly sensitive. However, the deprivation of intracellular reducing agents in endothelial cells makes endothelial cells sensitive to a redox-dependent shuttling of La protein.
Collapse
Affiliation(s)
- Nicole Berndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
| | - Claudia C. Bippes
- Institute of Immunology, Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, 01307 Dresden, Germany; (C.C.B.); (I.M.); (M.S.)
| | - Irene Michalk
- Institute of Immunology, Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, 01307 Dresden, Germany; (C.C.B.); (I.M.); (M.S.)
| | - Tabea Bartsch
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
| | - Claudia Arndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
| | - Edinson Puentes-Cala
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
- Corporación para la Investigación de la Corrosión (CIC), Piedecuesta 681011, Colombia
| | - Javier Andrés Soto
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
- Instituto de Investigación Masira, Facultad de Ciencias Médicas y de la Salud, Universidad de Santander, Cúcuta 540001, Colombia
| | - Liliana R. Loureiro
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
| | - Alexandra Kegler
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
| | - Dominik Bachmann
- Tumor Immunology, University Cancer Center (UCC), University Hospital Carl Gustav Carus Technische Universität Dresden, 01307 Dresden, Germany;
| | - Joanne K. Gross
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - Tim Gross
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - Biji T. Kurien
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - R. Hal Scofield
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - A. Darise Farris
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - Judith A. James
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (J.K.G.); (T.G.); (B.T.K.); (R.H.S.); (A.D.F.); (J.A.J.)
| | - Ralf Bergmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
- Department of Biophysics and Radiobiology, Semmelweis University, 1094 Budapest, Hungary
| | - Marc Schmitz
- Institute of Immunology, Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, 01307 Dresden, Germany; (C.C.B.); (I.M.); (M.S.)
- National Center for Tumor Diseases (NCT), 03128 Dresden, Germany
| | - Anja Feldmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
| | - Michael P. Bachmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany; (N.B.); (T.B.); (C.A.); (E.P.-C.); (J.A.S.); (L.R.L.); (A.K.); (R.B.); (A.F.)
- Institute of Immunology, Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, 01307 Dresden, Germany; (C.C.B.); (I.M.); (M.S.)
- National Center for Tumor Diseases (NCT), 03128 Dresden, Germany
| |
Collapse
|
10
|
Van Leeuwen-Kerkhoff N, Westers TM, Poddighe PJ, Povoleri GAM, Timms JA, Kordasti S, De Gruijl TD, Van de Loosdrecht AA. Reduced frequencies and functional impairment of dendritic cell subsets and non-classical monocytes in myelodysplastic syndromes. Haematologica 2021; 107:655-667. [PMID: 33567812 PMCID: PMC8883570 DOI: 10.3324/haematol.2020.268136] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Indexed: 11/09/2022] Open
Abstract
In myelodysplastic syndromes (MDS) the immune system is involved in pathogenesis as well as in disease progression. Dendritic cells (DC) are key players of the immune system by serving as regulators of immune responses. Their function has been scarcely studied in MDS and most of the reported studies didn't investigate naturally occurring DC subsets. Therefore, we here examined the frequency and function of DC subsets and slan+ non-classical monocytes in various MDS risk groups. Frequencies of DC as well as of slan+ monocytes were decreased in MDS bone marrow (BM) compared to normal bone marrow (NBM) samples. Transcriptional profiling revealed down-regulation of transcripts related to pro-inflammatory pathways in MDS-derived cells as compared to NBM. Additionally, their capacity to induce T cell proliferation was impaired. Multidimensional mass cytometry showed that whereas healthy donor-derived slan+ monocytes supported Th1/Th17/Treg differentiation/expansion their MDS-derived counterparts also mediated substantial Th2 expansion. Our findings point to a role for an impaired ability of DC subsets to adequately respond to cellular stress and DNA damage in the immune escape and progression of MDS. As such, it paves the way toward potential novel immunotherapeutic interventions.
Collapse
Affiliation(s)
- Nathalie Van Leeuwen-Kerkhoff
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam
| | - Theresia M Westers
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam
| | - Pino J Poddighe
- Department of Clinical Genetics, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam
| | - Giovanni A M Povoleri
- Department Inflammation Biology, King's College London, Centre for Inflammation Biology and Cancer Immunology, London
| | - Jessica A Timms
- Systems Cancer Immunology Lab, Comprehensive Cancer Center, King's College London, London
| | - Shahram Kordasti
- Systems Cancer Immunology Lab, Comprehensive Cancer Center, King's College London, London, United Kingdom; Dipartimento Scienze Cliniche e Molecolari, UNIVPM, Ancona
| | - Tanja D De Gruijl
- Department of Medical Oncology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam
| | - Arjan A Van de Loosdrecht
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam.
| |
Collapse
|
11
|
Bianchetto-Aguilera F, Tamassia N, Gasperini S, Calzetti F, Finotti G, Gardiman E, Montioli R, Bresciani D, Vermi W, Cassatella MA. Deciphering the fate of slan + -monocytes in human tonsils by gene expression profiling. FASEB J 2020; 34:9269-9284. [PMID: 32413173 DOI: 10.1096/fj.202000181r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/24/2020] [Accepted: 04/24/2020] [Indexed: 12/24/2022]
Abstract
Monocytic cells perform crucial homeostatic and defensive functions. However, their fate and characterization at the transcriptomic level in human tissues are partially understood, often as a consequence of the lack of specific markers allowing their unequivocal identification. The 6-sulfo LacNAc (slan) antigen identifies a subset of non-classical (NC) monocytes in the bloodstream, namely the slan+ -monocytes. In recent studies, we and other groups have reported that, in tonsils, slan marks dendritic cell (DC)-like cells, as defined by morphological, phenotypical, and functional criteria. However, subsequent investigations in lymphomas have uncovered a significant heterogeneity of tumor-infiltrating slan+ -cells, including a macrophage-like state. Based on their emerging role in tissue inflammation and cancer, herein we investigated slan+ -cell fate in tonsils by using a molecular-based approach. Hence, RNA from tonsil slan+ -cells, conventional CD1c+ DCs (cDC2) and CD11b+ CD14+ -macrophages was subjected to gene expression analysis. For comparison, transcriptomes were also obtained from blood cDC2, classical (CL), intermediate (INT), NC, and slan+ -monocytes. Data demonstrate that the main trajectory of human slan+ -monocytes infiltrating the tonsil tissue is toward a macrophage-like population, displaying molecular features distinct from those of tonsil CD11b+ CD14+ -macrophages and cDC2. These findings provide a novel view on the terminal differentiation path of slan+ -monocytes, which is relevant for inflammatory diseases and lymphomas.
Collapse
Affiliation(s)
| | - Nicola Tamassia
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Sara Gasperini
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Federica Calzetti
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Giulia Finotti
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Elisa Gardiman
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Riccardo Montioli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Debora Bresciani
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
12
|
Haynes WA, Haddon DJ, Diep VK, Khatri A, Bongen E, Yiu G, Balboni I, Bolen CR, Mao R, Utz PJ, Khatri P. Integrated, multicohort analysis reveals unified signature of systemic lupus erythematosus. JCI Insight 2020; 5:122312. [PMID: 31971918 DOI: 10.1172/jci.insight.122312] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 01/17/2020] [Indexed: 12/27/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease that follows an unpredictable disease course and affects multiple organs and tissues. We performed an integrated, multicohort analysis of 7,471 transcriptomic profiles from 40 independent studies to identify robust gene expression changes associated with SLE. We identified a 93-gene signature (SLE MetaSignature) that is differentially expressed in the blood of patients with SLE compared with healthy volunteers; distinguishes SLE from other autoimmune, inflammatory, and infectious diseases; and persists across diverse tissues and cell types. The SLE MetaSignature correlated significantly with disease activity and other clinical measures of inflammation. We prospectively validated the SLE MetaSignature in an independent cohort of pediatric patients with SLE using a microfluidic quantitative PCR (qPCR) array. We found that 14 of the 93 genes in the SLE MetaSignature were independent of IFN-induced and neutrophil-related transcriptional profiles that have previously been associated with SLE. Pathway analysis revealed dysregulation associated with nucleic acid biosynthesis and immunometabolism in SLE. We further refined a neutropoiesis signature and identified underappreciated transcripts related to immune cells and oxidative stress. In our multicohort, transcriptomic analysis has uncovered underappreciated genes and pathways associated with SLE pathogenesis, with the potential to advance clinical diagnosis, biomarker development, and targeted therapeutics for SLE.
Collapse
Affiliation(s)
- Winston A Haynes
- Institute for Immunity, Transplantation and Infection.,Division of Biomedical Informatics Research
| | - D James Haddon
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Vivian K Diep
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Avani Khatri
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Erika Bongen
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Gloria Yiu
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Imelda Balboni
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | | | - Rong Mao
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Paul J Utz
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection.,Division of Biomedical Informatics Research
| |
Collapse
|
13
|
Rammensee HG, Wiesmüller KH, Chandran PA, Zelba H, Rusch E, Gouttefangeas C, Kowalewski DJ, Di Marco M, Haen SP, Walz JS, Gloria YC, Bödder J, Schertel JM, Tunger A, Müller L, Kießler M, Wehner R, Schmitz M, Jakobi M, Schneiderhan-Marra N, Klein R, Laske K, Artzner K, Backert L, Schuster H, Schwenck J, Weber ANR, Pichler BJ, Kneilling M, la Fougère C, Forchhammer S, Metzler G, Bauer J, Weide B, Schippert W, Stevanović S, Löffler MW. A new synthetic toll-like receptor 1/2 ligand is an efficient adjuvant for peptide vaccination in a human volunteer. J Immunother Cancer 2019; 7:307. [PMID: 31730025 PMCID: PMC6858783 DOI: 10.1186/s40425-019-0796-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 10/30/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND We previously showed that the bacterial lipopeptide Pam3Cys-Ser-Ser, meanwhile established as a toll-like receptor (TLR) 1/2 ligand, acts as a strong adjuvant for the induction of virus specific CD8+ T cells in mice, when covalently coupled to a synthetic peptide. CASE PRESENTATION We now designed a new water-soluble synthetic Pam3Cys-derivative, named XS15 and characterized it in vitro by a TLR2 NF-κB luciferase reporter assay. Further, the capacity of XS15 to activate immune cells and stimulate peptide-specific CD8+ T and NK cells by 6-sulfo LacNAc+ monocytes was assessed by flow cytometry as well as cytokine induction using immunoassays. The induction of a functional immune response after vaccination of a volunteer with viral peptides was assessed by ELISpot assay and flow cytometry in peripheral blood cells and infiltrating cells at the vaccination site, as well as by immunohistochemistry and imaging. XS15 induced strong ex vivo CD8+ and TH1 CD4+ responses in a human volunteer upon a single injection of XS15 mixed to uncoupled peptides in a water-in-oil emulsion (Montanide™ ISA51 VG). A granuloma formed locally at the injection site containing highly activated functional CD4+ and CD8+ effector memory T cells. The total number of vaccine peptide-specific functional T cells was experimentally assessed and estimated to be 3.0 × 105 in the granuloma and 20.5 × 106 in peripheral blood. CONCLUSION Thus, in one volunteer we show a granuloma forming by peptides combined with an efficient adjuvant in a water-in-oil-emulsion, inducing antigen specific T cells detectable in circulation and at the vaccination site, after one single vaccination only. The ex vivo T cell responses in peripheral blood were detectable for more than one year and could be strongly boosted by a second vaccination. Hence, XS15 is a promising adjuvant candidate for peptide vaccination, in particular for tumor peptide vaccines in a personalized setting.
Collapse
Affiliation(s)
- Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany. .,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany. .,Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tubingen, Germany.
| | | | - P Anoop Chandran
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Henning Zelba
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Elisa Rusch
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tubingen, Germany
| | - Daniel J Kowalewski
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.,Present address: Immatics Biotechnologies GmbH, Tübingen, Germany
| | - Moreno Di Marco
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Sebastian P Haen
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany.,Department of Oncology, Hematology, Immunology, Rheumatology and Pulmonology, University Hospital of Tübingen, Tübingen, Germany
| | - Juliane S Walz
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tubingen, Germany.,Department of Oncology, Hematology, Immunology, Rheumatology and Pulmonology, University Hospital of Tübingen, Tübingen, Germany
| | - Yamel Cardona Gloria
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Johanna Bödder
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Jill-Marie Schertel
- Faculty of Medicine Carl Gustav Carus, Institute of Immunology, Technische Universität Dresden, Dresden, Germany
| | - Antje Tunger
- Faculty of Medicine Carl Gustav Carus, Institute of Immunology, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany and Helmholtz Association/ Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Luise Müller
- Faculty of Medicine Carl Gustav Carus, Institute of Immunology, Technische Universität Dresden, Dresden, Germany
| | - Maximilian Kießler
- Faculty of Medicine Carl Gustav Carus, Institute of Immunology, Technische Universität Dresden, Dresden, Germany
| | - Rebekka Wehner
- Faculty of Medicine Carl Gustav Carus, Institute of Immunology, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany and Helmholtz Association/ Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marc Schmitz
- Faculty of Medicine Carl Gustav Carus, Institute of Immunology, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany and Helmholtz Association/ Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Meike Jakobi
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | | | - Reinhild Klein
- Department of Oncology, Hematology, Immunology, Rheumatology and Pulmonology, University Hospital of Tübingen, Tübingen, Germany
| | - Karoline Laske
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Kerstin Artzner
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany
| | - Linus Backert
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.,Present address: Immatics Biotechnologies GmbH, Tübingen, Germany
| | - Heiko Schuster
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.,Present address: Immatics Biotechnologies GmbH, Tübingen, Germany
| | - Johannes Schwenck
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tubingen, Germany.,Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital of Tübingen, Tübingen, Germany.,Werner Siemens Imaging Center, Medical Faculty, University of Tübingen, Tübingen, Germany
| | - Alexander N R Weber
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.,Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tubingen, Germany
| | - Bernd J Pichler
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tubingen, Germany.,Werner Siemens Imaging Center, Medical Faculty, University of Tübingen, Tübingen, Germany
| | - Manfred Kneilling
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tubingen, Germany.,Werner Siemens Imaging Center, Medical Faculty, University of Tübingen, Tübingen, Germany.,Department of Dermatology, University Hospital of Tübingen, Tübingen, Germany
| | - Christian la Fougère
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tubingen, Germany.,Department of Nuclear Medicine and Clinical Molecular Imaging, University Hospital of Tübingen, Tübingen, Germany
| | - Stephan Forchhammer
- Department of Dermatology, University Hospital of Tübingen, Tübingen, Germany
| | - Gisela Metzler
- Department of Dermatology, University Hospital of Tübingen, Tübingen, Germany
| | - Jürgen Bauer
- Department of Dermatology, University Hospital of Tübingen, Tübingen, Germany
| | - Benjamin Weide
- Department of Dermatology, University Hospital of Tübingen, Tübingen, Germany
| | - Wilfried Schippert
- Department of Dermatology, University Hospital of Tübingen, Tübingen, Germany
| | - Stefan Stevanović
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tubingen, Germany
| | - Markus W Löffler
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076, Tübingen, Germany. .,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany. .,Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tubingen, Germany. .,Department of General, Visceral and Transplant Surgery, University Hospital of Tübingen, Tübingen, Germany. .,Department of Clinical Pharmacology, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
14
|
Günther C. Nucleic Acid Immunity in the Pathogenesis of Cutaneous Lupus Erythematosus. Front Immunol 2019; 10:1636. [PMID: 31379837 PMCID: PMC6646723 DOI: 10.3389/fimmu.2019.01636] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/01/2019] [Indexed: 12/12/2022] Open
Abstract
Cutaneous lupus erythematosus can be a devastating painful and mutilating disease that is associated with an inflammatory response in the skin driven by type I interferon activation. Clearance defects in the extra- and intracellular space lead to an enhanced prevalence of nucleic acids that represent danger signals for the innate immune system. Self nucleic acids can stimulate DNA and RNA sensors that have originally evolved to ensure viral defense. Their activation can induce a type I interferon dominated response in resident skin cells, macrophages and dendritic cells that subsequently progresses to adaptive immune stimulation. The genetic exploration of rare monogenic type I interferon driven diseases helped to identify these pathogenic concepts. Based on a genetic susceptibility lupus patients are more vulnerable to environmental trigger factors such as UV-irradiation that can provoke inflammation with local tissue destruction and eventually systemic disease. Understanding of these pathogenic concepts is a prerequisite for development of targeted therapies.
Collapse
Affiliation(s)
- Claudia Günther
- Department of Dermatology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| |
Collapse
|
15
|
Ahmad F, Döbel T, Schmitz M, Schäkel K. Current Concepts on 6-sulfo LacNAc Expressing Monocytes (slanMo). Front Immunol 2019; 10:948. [PMID: 31191513 PMCID: PMC6540605 DOI: 10.3389/fimmu.2019.00948] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 04/12/2019] [Indexed: 12/25/2022] Open
Abstract
The human mononuclear phagocytes system consists of dendritic cells (DCs), monocytes, and macrophages having different functions in bridging innate and adaptive immunity. Among the heterogeneous population of monocytes the cell surface marker slan (6-sulfo LacNAc) identifies a specific subset of human CD14- CD16+ non-classical monocytes, called slan+ monocytes (slanMo). In this review we discuss the identity and functions of slanMo, their contributions to immune surveillance by pro-inflammatory cytokine production, and cross talk with T cells and NK cells. We also consider the role of slanMo in the regulation of chronic inflammatory diseases and cancer. Finally, we highlight unresolved questions that should be the focus of future research.
Collapse
Affiliation(s)
- Fareed Ahmad
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Döbel
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany.,Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD, United States
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universtät Dresden, Dresden, Germany.,Partner Site Dresden, National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Knut Schäkel
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
16
|
Rhodes JW, Tong O, Harman AN, Turville SG. Human Dendritic Cell Subsets, Ontogeny, and Impact on HIV Infection. Front Immunol 2019; 10:1088. [PMID: 31156637 PMCID: PMC6532592 DOI: 10.3389/fimmu.2019.01088] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/29/2019] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) play important roles in orchestrating host immunity against invading pathogens, representing one of the first responders to infection by mucosal invaders. From their discovery by Ralph Steinman in the 1970s followed shortly after with descriptions of their in vivo diversity and distribution by Derek Hart, we are still continuing to progressively elucidate the spectrum of DCs present in various anatomical compartments. With the power of high-dimensional approaches such as single-cell sequencing and multiparameter cytometry, recent studies have shed new light on the identities and functions of DC subtypes. Notable examples include the reclassification of plasmacytoid DCs as purely interferon-producing cells and re-evaluation of intestinal conventional DCs and macrophages as derived from monocyte precursors. Collectively, these observations have changed how we view these cells not only in steady-state immunity but also during disease and infection. In this review, we will discuss the current landscape of DCs and their ontogeny, and how this influences our understanding of their roles during HIV infection.
Collapse
Affiliation(s)
- Jake William Rhodes
- Centre for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Orion Tong
- Centre for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Andrew Nicholas Harman
- Centre for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia.,Discipline of Applied Medical Sciences, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Stuart Grant Turville
- University of New South Wales, Sydney, NSW, Australia.,Kirby Institute, Kensington, NSW, Australia
| |
Collapse
|
17
|
Zaal A, van Ham SM, Ten Brinke A. Differential effects of anaphylatoxin C5a on antigen presenting cells, roles for C5aR1 and C5aR2. Immunol Lett 2019; 209:45-52. [PMID: 30959077 DOI: 10.1016/j.imlet.2019.03.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/21/2019] [Accepted: 03/22/2019] [Indexed: 12/24/2022]
Abstract
The anaphylatoxin C5a is well-known for its role as chemoattractant and contributes to immune cell recruitment into inflamed tissue and local inflammation. C5a has recently been implicated in modulation of antigen presenting cell function, such as macrophages and dendritic cells, which are pivotal for T cell activation and final T cell effector function. The published data on the effect of C5a on APC function and subsequent adaptive immune responses are in part conflicting, as both pro and anti-inflammatory effects have been described. In this review the opposing effects of C5a on APC function in mice and human are summarized and discussed in relation to origin of the involved APC subset, being either of the monocyte-derived lineage or dendritic cell lineage. In addition, the current knowledge on the expression of C5aR1 and C5aR2 on the different APC subsets is summarized. Based on the combined data, we propose that the differential effects of C5a on APC function may be attributed to absence or presence of co-expression of C5aR2 and C5aR1 on the specific APC.
Collapse
Affiliation(s)
- Anouk Zaal
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Swammerdam Institute for Life Sciences, University of Amsterdam, the Netherlands
| | - Anja Ten Brinke
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
18
|
Schierer S, Ostalecki C, Zinser E, Lamprecht R, Plosnita B, Stich L, Dörrie J, Lutz MB, Schuler G, Baur AS. Extracellular vesicles from mature dendritic cells (DC) differentiate monocytes into immature DC. Life Sci Alliance 2018; 1:e201800093. [PMID: 30519676 PMCID: PMC6277684 DOI: 10.26508/lsa.201800093] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 11/15/2018] [Accepted: 11/20/2018] [Indexed: 01/01/2023] Open
Abstract
Mature dendritic cells (DC) secrete substantial amounts of vesicles that are primarily ingested by monocytes, leading to differentiation processes in these target cells towards monocyte-derived DC. During inflammation, murine and human monocytes can develop into dendritic cells (DC), but this process is not entirely understood. Here, we demonstrate that extracellular vesicles (EV) secreted by mature human DC (maDC) differentiate peripheral monocytes into immature DC, expressing a unique marker pattern, including 6-sulfo LacNAc (slan), Zbtb46, CD64, and CD14. While EV from both maDC and immature DC differentiated monocytes similar to GM-CSF/IL-4 stimulation, only maDC-EV produced precursors, which upon maturation stimulus developed into T-cell–activating and IL-12p70–secreting maDC. Mechanistically, maDC-EV induced cell signaling through GM-CSF, which was abundant in EV as were IL-4 and other cytokines and chemokines. When injected into the mouse skin, murine maDC-EV attracted immune cells including monocytes that developed activation markers typical for inflammatory cells. Skin-injected EV also reached lymph nodes, causing a similar immune cell infiltration. We conclude that DC-derived EV likely serve to perpetuate an immune reaction and may contribute to chronic inflammation.
Collapse
Affiliation(s)
- Stefan Schierer
- Department of Dermatology, University Hospital Erlangen, Kussmaul Campus, Erlangen, Germany
| | - Christian Ostalecki
- Department of Dermatology, University Hospital Erlangen, Kussmaul Campus, Erlangen, Germany
| | - Elisabeth Zinser
- Department of Immune Modulation, University Hospital Erlangen, Kussmaul Campus, Erlangen, Germany
| | - Ricarda Lamprecht
- Department of Dermatology, University Hospital Erlangen, Kussmaul Campus, Erlangen, Germany
| | | | - Lena Stich
- Department of Immune Modulation, University Hospital Erlangen, Kussmaul Campus, Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, University Hospital Erlangen, Kussmaul Campus, Erlangen, Germany
| | - Manfred B Lutz
- Institute of Virology and Immunobiology, Würzburg, Germany
| | - Gerold Schuler
- Department of Dermatology, University Hospital Erlangen, Kussmaul Campus, Erlangen, Germany
| | - Andreas S Baur
- Department of Dermatology, University Hospital Erlangen, Kussmaul Campus, Erlangen, Germany
| |
Collapse
|
19
|
Iannetta M, Savinelli S, Rossi R, Mascia C, Marocco R, Vita S, Zuccalà P, Zingaropoli MA, Mengoni F, Massetti AP, Falciano M, d'Ettorre G, Ciardi MR, Mastroianni CM, Vullo V, Lichtner M. Myeloid and lymphoid activation markers in AIDS and non-AIDS presenters. Immunobiology 2018; 224:231-241. [PMID: 30522891 DOI: 10.1016/j.imbio.2018.11.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 11/27/2018] [Accepted: 11/27/2018] [Indexed: 01/21/2023]
Abstract
HIV infection is characterized by a state of chronic activation of the immune system, which is not completely reversed by antiretroviral treatment (ART). The aim of this study was to assess myeloid and lymphoid activation markers during HIV infection, before and one year after ART initiation, in AIDS and non-AIDS presenters. Treatment naïve HIV positive patients were enrolled in this study. Myeloid dendritic cell (mDC), plasmacytoid dendritic cell (pDC), slanDC, monocyte and T-lymphocyte cell counts and activation status, were assessed by flow cytometry in peripheral blood samples. Soluble (s)CD14 and sCD163 were assessed in plasma samples using ELISA assays. Statistical analyses were performed using GraphPad Prism and Minitab Express. Thirty-four ART naïve HIV-1 infected subjects were enrolled in this study (22 non-AIDS and 12 AIDS presenters). Seventeen healthy donors (HD) were included as control group. Although circulating mDC levels resulted unchanged, HLA-DR expression was decreased on mDCs of HIV positive subjects compared to HD (p < 0,0001). AIDS presenters showed the lowest level of expression of HLA-DR on mDCs. Circulating levels of pDCs were decreased in HIV patients compared to HD (p < 0,001), without any changes in HLA-DR expression. SlanDC cell counts were extremely reduced in AIDS presenters, compared to non-AIDS presenters and HD (p < 0,01 and p < 0,0001, respectively) and showed higher HLA-DR expression in HIV patients compared to HD (p < 0,01). Intermediate monocyte (IM) cell counts were increased in AIDS and non-AIDS presenters compared to HD (p < 0,001 and p < 0,001 respectively). Furthermore, IM expansion was directly correlated to HIV viral load (p = 0,036) and independent from CD4 cell counts and activation levels. Plasma concentrations of sCD14 and sCD163 resulted increased in HIV infected subjects compared to HD (p < 0,0001 and p < 0,001), with the highest levels observed in AIDS presenters. After 1 year, ART was able to increase pDC and decrease IM absolute cell counts and modify HLA-DR expression on mDCs and slanDCs, approaching the levels observed in HD. ART reduced also CD4 and CD8 activation levels. In conclusion, in untreated HIV infected subjects circulating dendritic cells resulted altered either in numbers or in HLA-DR expression, especially in AIDS presenters. IM absolute counts were equally increased in AIDS and non-AIDS presenters. ART was able to reduce myeloid and lymphoid inflammation in both advanced and non-advanced HIV patients, confirming the role of ART in hampering disease progression and immune activation associated non-AIDS events.
Collapse
Affiliation(s)
- Marco Iannetta
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Stefano Savinelli
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Raffaella Rossi
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Claudia Mascia
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Raffaella Marocco
- Infectious Diseases Unit, Sapienza University, Santa Maria Goretti Hospital, Via Canova, 04100, Latina, Italy
| | - Serena Vita
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy; Infectious Diseases Unit, Sapienza University, Santa Maria Goretti Hospital, Via Canova, 04100, Latina, Italy
| | - Paola Zuccalà
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Maria Antonella Zingaropoli
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Fabio Mengoni
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Anna Paola Massetti
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Mario Falciano
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Gabriella d'Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Maria Rosa Ciardi
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Vincenzo Vullo
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Miriam Lichtner
- Department of Public Health and Infectious Diseases, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy; Infectious Diseases Unit, Sapienza University, Santa Maria Goretti Hospital, Via Canova, 04100, Latina, Italy
| |
Collapse
|
20
|
Okumura M, Ozawa T, Hamana H, Norimatsu Y, Tsuda R, Kobayashi E, Shinoda K, Taki H, Tobe K, Imura J, Sugiyama E, Kishi H, Muraguchi A. Autoantibodies reactive to PEP08 are clinically related with morbidity and severity of interstitial lung disease in connective tissue diseases. Eur J Immunol 2018; 48:1717-1727. [DOI: 10.1002/eji.201747381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 05/15/2018] [Accepted: 07/05/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Maiko Okumura
- Department of Immunology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
- The First Department of Internal Medicine; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
| | - Tatsuhiko Ozawa
- Department of Immunology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
| | - Hiroshi Hamana
- Department of Immunology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
| | - Yu Norimatsu
- Department of Immunology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
- School of Medicine Program of Medicine; Hiroshima University; Minami Hiroshima Japan
| | - Reina Tsuda
- Department of Immunology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
- The First Department of Internal Medicine; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
| | - Eiji Kobayashi
- Department of Immunology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
| | - Koichiro Shinoda
- The First Department of Internal Medicine; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
| | - Hirofumi Taki
- The First Department of Internal Medicine; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
| | - Kazuyuki Tobe
- The First Department of Internal Medicine; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
| | - Johji Imura
- Department of Diagnostic Pathology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
| | - Eiji Sugiyama
- Department of Clinical Immunology and Rheumatology; Hiroshima University Hospital; Minami Hiroshima Japan
| | - Hiroyuki Kishi
- Department of Immunology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
| | - Atsushi Muraguchi
- Department of Immunology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama Japan
| |
Collapse
|
21
|
Baran W, Oehrl S, Ahmad F, Döbel T, Alt C, Buske-Kirschbaum A, Schmitz M, Schäkel K. Phenotype, Function, and Mobilization of 6-Sulfo LacNAc-Expressing Monocytes in Atopic Dermatitis. Front Immunol 2018; 9:1352. [PMID: 29977237 PMCID: PMC6021776 DOI: 10.3389/fimmu.2018.01352] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/31/2018] [Indexed: 01/31/2023] Open
Abstract
Mononuclear phagocytes (MPs) are important immune regulatory cells in atopic dermatitis (AD). We previously identified 6-sulfo LacNAc-expressing monocytes (slanMo) as TNF-α- and IL-23-producing cells in psoriatic skin lesions and as inducers of IFN-γ-, IL-17-, and IL-22-producing T cells. These cytokines are also upregulated in AD and normalize with treatment, as recently shown for dupilumab-treated patients. We here asked for the role of slanMo in AD. Increased numbers of slanMo were found in AD skin lesions. In difference to other MPs in AD, slanMo lacked expression of FcɛRI, CD1a, CD14, and CD163. slanMo from blood of patients with AD expressed increased levels of CD86 and produced IL-12 and TNF-α at higher amounts than CD14+ monocytes and myeloid dendritic cells. While CD14+ monocytes from patients with AD revealed a reduced IL-12 production, we observed no difference in the cytokine production comparing slanMo in AD and healthy controls. Interestingly, experimentally induced mental stress, a common trigger of flares in patients with AD, rapidly mobilized slanMo which retained their high TNF-α-producing capacity. This study identifies slanMo as a distinct population of inflammatory cells in skin lesions and as proinflammatory blood cells in patients with AD. slanMo may, therefore, represent a potent future target for treatment of AD.
Collapse
Affiliation(s)
- Wojciech Baran
- Department of Dermatology, Venerology and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | - Stephanie Oehrl
- Department of Dermatology, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Fareed Ahmad
- Department of Dermatology, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Thomas Döbel
- Department of Dermatology, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Christina Alt
- Department of Dermatology, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | | | - Marc Schmitz
- Institute of Immunology, Medical Faculty, Technical University of Dresden, Dresden, Germany.,National Center for Tumor Diseases, University Hospital Carl Gustav Carus, Technical University of Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Regenerative Therapies Dresden (CRTD), Medical Faculty, Technical University of Dresden, Dresden, Germany
| | - Knut Schäkel
- Department of Dermatology, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
22
|
Olaru F, Döbel T, Lonsdorf AS, Oehrl S, Maas M, Enk AH, Schmitz M, Gröne EF, Gröne HJ, Schäkel K. Intracapillary immune complexes recruit and activate slan-expressing CD16+ monocytes in human lupus nephritis. JCI Insight 2018; 3:96492. [PMID: 29875315 DOI: 10.1172/jci.insight.96492] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 04/24/2018] [Indexed: 12/30/2022] Open
Abstract
Lupus nephritis is a major cause of morbidity in patients with systemic lupus erythematosus. Among the different types of lupus nephritis, intracapillary immune complex (IC) deposition and accumulation of monocytes are hallmarks of lupus nephritis class III and IV. The relevance of intracapillary ICs in terms of monocyte recruitment and activation, as well as the nature and function of these monocytes are not well understood. For the early focal form of lupus nephritis (class III) we demonstrate a selective accumulation of the proinflammatory population of 6-sulfo LacNAc+ (slan) monocytes (slanMo), which locally expressed TNF-α. Immobilized ICs induced a direct recruitment of slanMo from the microcirculation via interaction with Fc γ receptor IIIA (CD16). Interestingly, intravenous immunoglobulins blocked CD16 and prevented cell recruitment. Engagement of immobilized ICs by slanMo induced the production of neutrophil-attracting chemokine CXCL2 as well as TNF-α, which in a forward feedback loop stimulated endothelial cells to produce the slanMo-recruiting chemokine CX3CL1 (fractalkine). In conclusion, we observed that expression of CD16 equips slanMo with a unique capacity to orchestrate early IC-induced inflammatory responses in glomeruli and identified slanMo as a pathogenic proinflammatory cell type in lupus nephritis.
Collapse
Affiliation(s)
- Florina Olaru
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Döbel
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Anke S Lonsdorf
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stephanie Oehrl
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Maas
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander H Enk
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marc Schmitz
- Institute of Immunology, Medical Faculty, Technische Universität (TU) Dresden, Dresden, Germany.,National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Regenerative Therapies Dresden (CRTD), Medical Faculty, TU Dresden, Dresden, Germany
| | - Elisabeth F Gröne
- Department of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann-J Gröne
- Department of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Knut Schäkel
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
23
|
Thomas K, Metz I, Tumani H, Brück W, Ziemssen T. 6-sulpho LacNAc(+) dendritic cells accumulate in various inflammatory, but not ischaemic conditions of the central nervous system. Neuropathol Appl Neurobiol 2018; 42:394-8. [PMID: 26844946 DOI: 10.1111/nan.12313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 01/25/2016] [Accepted: 02/04/2016] [Indexed: 12/14/2022]
Affiliation(s)
- K Thomas
- Department of Neurology, University Hospital, Dresden, Germany
| | - I Metz
- Department of Neuropathology, University Medical Centre, Göttingen, Germany
| | - H Tumani
- Department of Neurology, University Hospital, Ulm, Germany
| | - W Brück
- Department of Neuropathology, University Medical Centre, Göttingen, Germany
| | - T Ziemssen
- Department of Neurology, University Hospital, Dresden, Germany
| |
Collapse
|
24
|
The Biology of Monocytes and Dendritic Cells: Contribution to HIV Pathogenesis. Viruses 2018; 10:v10020065. [PMID: 29415518 PMCID: PMC5850372 DOI: 10.3390/v10020065] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/01/2018] [Accepted: 02/03/2018] [Indexed: 02/07/2023] Open
Abstract
Myeloid cells such as monocytes, dendritic cells (DC) and macrophages (MΦ) are key components of the innate immune system contributing to the maintenance of tissue homeostasis and the development/resolution of immune responses to pathogens. Monocytes and DC, circulating in the blood or infiltrating various lymphoid and non-lymphoid tissues, are derived from distinct bone marrow precursors and are typically short lived. Conversely, recent studies revealed that subsets of tissue resident MΦ are long-lived as they originate from embryonic/fetal precursors that have the ability to self-renew during the life of an individual. Pathogens such as the human immunodeficiency virus type 1 (HIV-1) highjack the functions of myeloid cells for viral replication (e.g., MΦ) or distal dissemination and cell-to-cell transmission (e.g., DC). Although the long-term persistence of HIV reservoirs in CD4+ T-cells during viral suppressive antiretroviral therapy (ART) is well documented, the ability of myeloid cells to harbor replication competent viral reservoirs is still a matter of debate. This review summarizes the current knowledge on the biology of monocytes and DC during homeostasis and in the context of HIV-1 infection and highlights the importance of future studies on long-lived resident MΦ to HIV persistence in ART-treated patients.
Collapse
|
25
|
Steffen S, Abraham S, Herbig M, Schmidt F, Blau K, Meisterfeld S, Beissert S, Guck J, Günther C. Toll-Like Receptor-Mediated Upregulation of CXCL16 in Psoriasis Orchestrates Neutrophil Activation. J Invest Dermatol 2018; 138:344-354. [DOI: 10.1016/j.jid.2017.08.041] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/10/2017] [Accepted: 08/27/2017] [Indexed: 12/27/2022]
|
26
|
van Leeuwen-Kerkhoff N, Lundberg K, Westers TM, Kordasti S, Bontkes HJ, de Gruijl TD, Lindstedt M, van de Loosdrecht AA. Transcriptional profiling reveals functional dichotomy between human slan + non-classical monocytes and myeloid dendritic cells. J Leukoc Biol 2017; 102:1055-1068. [PMID: 28720687 DOI: 10.1189/jlb.3ma0117-037r] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/31/2017] [Accepted: 06/20/2017] [Indexed: 11/24/2022] Open
Abstract
Human 6-sulfo LacNac-positive (slan+) cells have been subject to a paradigm debate. They have previously been classified as a distinct dendritic cell (DC) subset. However, evidence has emerged that they may be more related to monocytes than to DCs. To gain deeper insight into the functional specialization of slan+ cells, we have compared them with both conventional myeloid DC subsets (CD1c+ and CD141+) in human peripheral blood (PB). With the use of genome-wide transcriptional profiling, as well as functional tests, we clearly show that slan+ cells form a distinct, non-DC-like population. They cluster away from both DC subsets, and their gene-expression profile evidently suggests involvement in distinct inflammatory processes. An extensive transcriptional meta-analysis confirmed the relationship of slan+ cells with the monocytic compartment rather than with DCs. From a functional perspective, their ability to prime CD4+ and CD8+ T cells is relatively low. Combined with the finding that "antigen presentation by MHC class II" is at the top of under-represented pathways in slan+ cells, this points to a minimal role in directing adaptive T cell immunity. Rather, the higher expression levels of complement receptors on their cell surface, together with their high secretion of IL-1β and IL-6, imply a specific role in innate inflammatory processes, which is consistent with their recent identification as non-classical monocytes. This study extends our knowledge on DC/monocyte subset biology under steady-state conditions and contributes to our understanding of their role in immune-mediated diseases and their potential use in immunotherapeutic strategies.
Collapse
Affiliation(s)
- Nathalie van Leeuwen-Kerkhoff
- Department of Hematology, Cancer Center Amsterdam, Vrije Universiteit Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Theresia M Westers
- Department of Hematology, Cancer Center Amsterdam, Vrije Universiteit Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Shahram Kordasti
- Department of Haematological Medicine, King's College London and King's College Hospital, London, United Kingdom
| | - Hetty J Bontkes
- Department of Oral Cell Biology, Academic Center for Dentistry, Amsterdam, The Netherlands; and
| | - Tanja D de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, Vrije Universiteit Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Malin Lindstedt
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Arjan A van de Loosdrecht
- Department of Hematology, Cancer Center Amsterdam, Vrije Universiteit Amsterdam University Medical Center, Amsterdam, The Netherlands;
| |
Collapse
|
27
|
Zaal A, Dieker M, Oudenampsen M, Turksma AW, Lissenberg-Thunnissen SN, Wouters D, van Ham SM, Ten Brinke A. Anaphylatoxin C5a Regulates 6-Sulfo-LacNAc Dendritic Cell Function in Human through Crosstalk with Toll-Like Receptor-Induced CREB Signaling. Front Immunol 2017; 8:818. [PMID: 28769928 PMCID: PMC5509794 DOI: 10.3389/fimmu.2017.00818] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 06/27/2017] [Indexed: 01/05/2023] Open
Abstract
Activation of antigen-presenting dendritic cells (DCs) and the complement system are essential early events in the immune defense against invading pathogens. Recently, we and others demonstrated immunological crosstalk between signaling from receptors recognizing complement activation products and PAMPs on DCs. This affects DC effector function, as demonstrated by the finding that C5a prevents induction of pro-inflammatory cytokines by toll-like receptor (TLR) ligands in human monocyte-derived DCs (moDCs). Here, we demonstrate that this regulatory crosstalk is specifically important in 6-sulfo LacNAc dendritic cells (slanDCs), the most pro-inflammatory DC subset found in human. C5aR and TLR signaling show profound interference in the ERK/p38/CREB1 signaling pathways. C5aR signaling accelerates TLR-induced CREB1 phosphorylation both in moDC and slanDC. This is key in the regulatory effect of C5a on pro-inflammatory DC maturation by mediating induction of IL-10, which subsequently inhibits pro-inflammatory cytokine production via negative feedback signaling. Importantly, the regulatory effect of C5a affects T-cell immunity by decreasing Th1 and cytotoxic CD8 T-cell responses. The finding that the pro-inflammatory effector function of slanDC can be down modulated by activation products of the complement system highlights the existence of intricate regulatory interactions between various arms of the immune system. Intensive immune monitoring of patients suffering from complement-mediated diseases or patients receiving complement modulating compounds can give more inside in the contribution of complement receptor and TLR crosstalk in APCs in disease.
Collapse
Affiliation(s)
- Anouk Zaal
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Miranda Dieker
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Manon Oudenampsen
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Annelies W Turksma
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Suzanne N Lissenberg-Thunnissen
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Diana Wouters
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Anja Ten Brinke
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
28
|
Controlling the pro-inflammatory function of 6-sulfo LacNAc (slan) dendritic cells with dimethylfumarate. J Dermatol Sci 2017; 87:278-284. [PMID: 28732748 DOI: 10.1016/j.jdermsci.2017.06.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 06/20/2017] [Indexed: 11/23/2022]
Abstract
BACKROUND The fumaric acid ester (FAE) dimethylfumarate (DMF) is a small molecule immunomodulator successfully used for the treatment of psoriasis and multiple sclerosis (MS). DMF is thought to inhibit pathogenic immune responses with Th17/Th1T cells, and IL-23/IL-12 producing dendritic cells (DCs). 6-sulfo LacNAc expressing dendritic cells (slanDCs) are a human pro-inflammatory cell type found frequently among the infiltrating leukocytes in skin lesions of psoriasis and brain lesions of MS. OBJECTIVE To explore the influence of DMF on functional properties and cell signaling pathways of slanDCs. METHODS In the context of slanDCs we studied the role of DMF in modulating cell migration, phenotypic maturation, cytokine production, cell signaling and T cell stimulation. RESULTS Initially, we observed the reduction of slanDCs numbers in psoriasis skin lesions of FAE treated patients. Studying whether DMF controls the migratory capacity of slanDCs to chemotactic factors expressed in psoriasis we observed an inhibition of the CX3CL1 and C5a depedent cell migration. DMF also attenuated the rapid spontaneous phenotypic maturation of slanDCs, as judged by a reduced CD80, CD86, CD83 and HLA-DR expression. In addition, we observed a DMF-dependent decrease of IL-23, IL-12, TNF-α and IL-10 secretion, and noticed a reduced capacity to stimulate Th17/Th1 responses. DMF targeted in slanDCs different intracellular cell signaling pathways including NFκB, STAT1 and HO-1. CONCLUSIONS With this study we identify a frequent pro-inflammatory cell type found in psoriasis and MS as a relevant target for the therapeutic immunomodulatory effects of DMF.
Collapse
|
29
|
Brief Report: HIV-1 Infection Results in Increased Frequency of Active and Inflammatory SlanDCs that Produce High Level of IL-1β. J Acquir Immune Defic Syndr 2017; 73:34-8. [PMID: 27243902 DOI: 10.1097/qai.0000000000001082] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
HIV infection is marked by phenotypic and functional alterations of immune cells. Different studies have shown both numerical and functional deterioration of dendritic cells in HIV-1-infected patients. In this study, we report an increase of inflammatory 6-sulfo LacNAc dendritic cells (slanDCs) that are more activated and produce higher amounts of interleukin (IL)-1β during HIV-1 infection as compared with healthy controls. IL-1β plays a regulatory role in chronic inflammatory disorders. Therefore, our findings might reveal a compensatory regulatory function of slanDCs during HIV-1 infection.
Collapse
|
30
|
Qiu H, Wu H, Chan V, Lau CS, Lu Q. Transcriptional and epigenetic regulation of follicular T-helper cells and their role in autoimmunity. Autoimmunity 2017; 50:71-81. [PMID: 28263097 DOI: 10.1080/08916934.2017.1284821] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Hong Qiu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China and
| | - Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China and
| | - Vera Chan
- Division of Rheumatology & Clinical Immunology, Department of Medicine, University of Hong Kong, Hong Kong
| | - Chak-Sing Lau
- Division of Rheumatology & Clinical Immunology, Department of Medicine, University of Hong Kong, Hong Kong
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China and
| |
Collapse
|
31
|
Worbs T, Hammerschmidt SI, Förster R. Dendritic cell migration in health and disease. Nat Rev Immunol 2016; 17:30-48. [PMID: 27890914 DOI: 10.1038/nri.2016.116] [Citation(s) in RCA: 583] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are potent and versatile antigen-presenting cells, and their ability to migrate is key for the initiation of protective pro-inflammatory as well as tolerogenic immune responses. Recent comprehensive studies have highlighted the importance of DC migration in the maintenance of immune surveillance and tissue homeostasis, and also in the pathogenesis of a range of diseases. In this Review, we summarize the anatomical, cellular and molecular factors that regulate the migration of different DC subsets in health and disease. In particular, we focus on new insights concerning the role of migratory DCs in the pathogenesis of diseases of the skin, intestine, lung, and brain, as well as in autoimmunity and atherosclerosis.
Collapse
Affiliation(s)
- Tim Worbs
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Swantje I Hammerschmidt
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| |
Collapse
|
32
|
Micheletti A, Finotti G, Calzetti F, Lonardi S, Zoratti E, Bugatti M, Stefini S, Vermi W, Cassatella MA. slanDCs/M-DC8+ cells constitute a distinct subset of dendritic cells in human tonsils [corrected]. Oncotarget 2016; 7:161-75. [PMID: 26695549 PMCID: PMC4807990 DOI: 10.18632/oncotarget.6660] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/22/2015] [Indexed: 12/17/2022] Open
Abstract
Human blood dendritic cells (DCs) include three main distinct subsets, namely the CD1c+ and CD141+ myeloid DCs (mDCs) and the CD303+ plasmacytoid DCs (pDCs). More recently, a population of slan/M-DC8+ cells, also known as “slanDCs”, has been described in blood and detected even in inflamed secondary lymphoid organs and non-lymphoid tissues. Nevertheless, hallmarks of slan/M-DC8+ cells in tissues are poorly defined. Herein, we report a detailed characterization of the phenotype and function of slan/M-DC8+ cells present in human tonsils. We found that tonsil slan/M-DC8+ cells represent a unique DC cell population, distinct from their circulating counterpart and also from all other tonsil DC and monocyte/macrophage subsets. Phenotypically, slan/M-DC8+ cells in tonsils display a CD11c+HLA-DR+CD14+CD11bdim/negCD16dim/negCX3CR1dim/neg marker repertoire, while functionally they exhibit an efficient antigen presentation capacity and a constitutive secretion of TNFα. Notably, such DC phenotype and functions are substantially reproduced by culturing blood slan/M-DC8+ cells in tonsil-derived conditioned medium (TDCM), further supporting the hypothesis of a full DC-like differentiation program occurring within the tonsil microenvironment. Taken together, our data suggest that blood slan/M-DC8+ cells are immediate precursors of a previously unrecognizedcompetent DC subset in tonsils, and pave the way for further characterization of slan/M-DC8+ cells in other tissues.
Collapse
Affiliation(s)
- Alessandra Micheletti
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Giulia Finotti
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Federica Calzetti
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy
| | - Elisa Zoratti
- Applied Research on Cancer-Network (ARC-NET), University of Verona, Verona, Italy
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy
| | - Stefania Stefini
- Unit of Pediatric Otorhinolaryngology, Spedali Civili di Brescia, Brescia, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy.,Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Marco A Cassatella
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| |
Collapse
|
33
|
Dendritic Cells in Systemic Lupus Erythematosus: From Pathogenic Players to Therapeutic Tools. Mediators Inflamm 2016; 2016:5045248. [PMID: 27122656 PMCID: PMC4829720 DOI: 10.1155/2016/5045248] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 03/13/2016] [Indexed: 12/20/2022] Open
Abstract
System lupus erythematosus (SLE) is a multifactorial systemic autoimmune disease with a wide variety of presenting features. SLE is believed to result from dysregulated immune responses, loss of tolerance of CD4 T cells and B cells to ubiquitous self-antigens, and the subsequent production of anti-nuclear and other autoreactive antibodies. Recent research has associated lupus development with changes in the dendritic cell (DC) compartment, including altered DC subset frequency and localization, overactivation of mDCs and pDCs, and functional defects in DCs. Here we discuss the current knowledge on the role of DC dysfunction in SLE pathogenesis, with the focus on DCs as targets for interventional therapies.
Collapse
|
34
|
Blanco P, Ueno H, Schmitt N. T follicular helper (Tfh) cells in lupus: Activation and involvement in SLE pathogenesis. Eur J Immunol 2016; 46:281-90. [DOI: 10.1002/eji.201545760] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 11/17/2015] [Accepted: 11/23/2015] [Indexed: 12/28/2022]
Affiliation(s)
- Patrick Blanco
- Univ. Bordeaux; CIRID, UMR/CNRS; 5164 Bordeaux France
- CNRS; CIRID, UMR; 5164 Bordeaux France
- CHU de Bordeaux; Bordeaux France
| | - Hideki Ueno
- Baylor Institute for Immunology Research; Dallas USA
| | | |
Collapse
|
35
|
Langosch S, Wehner R, Malecka A, Franks HA, Schäkel K, Bachmann M, Jackson AM, Schmitz M. Impact of p38 mitogen-activated protein kinase inhibition on immunostimulatory properties of human 6-sulfo LacNAc dendritic cells. Immunobiology 2016; 221:166-74. [DOI: 10.1016/j.imbio.2015.09.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 08/13/2015] [Accepted: 09/07/2015] [Indexed: 02/07/2023]
|
36
|
Vermi W, Micheletti A, Lonardi S, Costantini C, Calzetti F, Nascimbeni R, Bugatti M, Codazzi M, Pinter PC, Schäkel K, Tamassia N, Cassatella MA. slanDCs selectively accumulate in carcinoma-draining lymph nodes and marginate metastatic cells. Nat Commun 2015; 5:3029. [PMID: 24398631 DOI: 10.1038/ncomms4029] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 11/27/2013] [Indexed: 01/01/2023] Open
Abstract
Dendritic cells (DCs) initiate adaptive immune responses to cancer cells by activating naive T lymphocytes. 6-sulfo LacNAc(+) DCs (slanDCs) represent a distinct population of circulating and tissue proinflammatory DCs, whose role in cancer immune surveillance is unknown. Herein, by screening a large set of clinical samples, we demonstrate accumulation of slanDCs in metastatic tumour-draining lymph nodes (M-TDLN) from carcinoma patients. Remarkably, slanDCs are absent at the primary carcinoma site, while their selective nodal recruitment follows the arrival of cancer cells to M-TDLN. slanDCs surround metastatic carcinoma deposits in close proximity to dead cells and efficiently phagocytose tumour cells. In colon carcinoma patients, the contingent of circulating slanDCs remains intact and competent in terms of IL-12p70 and tumour necrosis factor alpha production, induction of T-cell proliferation and migratory capacity to a set of chemokines produced in M-TDLN. We conclude that activated slanDCs represent previously unrecognized players of nodal immune responses to cancer cells.
Collapse
Affiliation(s)
- William Vermi
- 1] Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy [2] Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Alessandra Micheletti
- 1] Section of General Pathology, Department of Pathology and Diagnostics, University of Verona, Verona 37134, Italy [2]
| | - Silvia Lonardi
- 1] Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy [2]
| | - Claudio Costantini
- Section of General Pathology, Department of Pathology and Diagnostics, University of Verona, Verona 37134, Italy
| | - Federica Calzetti
- Section of General Pathology, Department of Pathology and Diagnostics, University of Verona, Verona 37134, Italy
| | - Riccardo Nascimbeni
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Mattia Bugatti
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Manuela Codazzi
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Patrick C Pinter
- Section of Otorhinolaryngology, Department of Surgery, University of Verona, Verona 37134, Italy
| | - Knut Schäkel
- Department of Dermatology, University Hospital Heidelberg, 69115 Heidelberg, Germany
| | - Nicola Tamassia
- Section of General Pathology, Department of Pathology and Diagnostics, University of Verona, Verona 37134, Italy
| | - Marco A Cassatella
- Section of General Pathology, Department of Pathology and Diagnostics, University of Verona, Verona 37134, Italy
| |
Collapse
|
37
|
slan-defined subsets of CD16-positive monocytes: impact of granulomatous inflammation and M-CSF receptor mutation. Blood 2015; 126:2601-10. [PMID: 26443621 DOI: 10.1182/blood-2015-06-651331] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 09/23/2015] [Indexed: 12/21/2022] Open
Abstract
Human monocytes are subdivided into classical, intermediate, and nonclassical subsets, but there is no unequivocal strategy to dissect the latter 2 cell types. We show herein that the cell surface marker 6-sulfo LacNAc (slan) can define slan-positive CD14(+)CD16(++) nonclassical monocytes and slan-negative CD14(++)CD16(+) intermediate monocytes. Gene expression profiling confirms that slan-negative intermediate monocytes show highest expression levels of major histocompatibility complex class II genes, whereas a differential ubiquitin signature is a novel feature of the slan approach. In unsupervised hierarchical clustering, the slan-positive nonclassical monocytes cluster with monocytes and are clearly distinct from CD1c(+) dendritic cells. In clinical studies, we show a selective increase of the slan-negative intermediate monocytes to >100 cells per microliter in patients with sarcoidosis and a fivefold depletion of the slan-positive monocytes in patients with hereditary diffuse leukoencephalopathy with axonal spheroids (HDLS), which is caused by macrophage colony-stimulating factor (M-CSF) receptor mutations. These data demonstrate that the slan-based definition of CD16-positive monocyte subsets is informative in molecular studies and in clinical settings.
Collapse
|
38
|
Kumar NA, Cheong K, Powell DR, da Fonseca Pereira C, Anderson J, Evans VA, Lewin SR, Cameron PU. The role of antigen presenting cells in the induction of HIV-1 latency in resting CD4(+) T-cells. Retrovirology 2015; 12:76. [PMID: 26362311 PMCID: PMC4567795 DOI: 10.1186/s12977-015-0204-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 09/01/2015] [Indexed: 12/24/2022] Open
Abstract
Background Combination antiretroviral therapy (cART) is able to control HIV-1 viral replication, however long-lived latent infection in resting memory CD4+ T-cells persist. The mechanisms for establishment and maintenance of latent infection in resting memory CD4+ T-cells remain unclear. Previously we have shown that HIV-1 infection of resting CD4+ T-cells co-cultured with CD11c+ myeloid dendritic cells (mDC) produced a population of non-proliferating T-cells with latent infection. Here we asked whether different antigen presenting cells (APC), including subpopulations of DC and monocytes, were able to induce post-integration latent infection in resting CD4+ T-cells, and examined potential cell interactions that may be involved using RNA-seq. Results mDC (CD1c+), SLAN+ DC and CD14+ monocytes were most efficient in stimulating proliferation of CD4+ T-cells during syngeneic culture and in generating post-integration latent infection in non-proliferating CD4+ T-cells following HIV-1 infection of APC-T cell co-cultures. In comparison, plasmacytoid DC (pDC) and B-cells did not induce latent infection in APC-T-cell co-cultures. We compared the RNA expression profiles of APC subpopulations that could and could not induce latency in non-proliferating CD4+ T-cells. Gene expression analysis, comparing the CD1c+ mDC, SLAN+ DC and CD14+ monocyte subpopulations to pDC identified 53 upregulated genes that encode proteins expressed on the plasma membrane that could signal to CD4+ T-cells via cell–cell interactions (32 genes), immune checkpoints (IC) (5 genes), T-cell activation (9 genes), regulation of apoptosis (5 genes), antigen presentation (1 gene) and through unknown ligands (1 gene). Conclusions APC subpopulations from the myeloid lineage, specifically mDC subpopulations and CD14+ monocytes, were able to efficiently induce post-integration HIV-1 latency in non-proliferating CD4+ T-cells in vitro. Inhibition of key pathways involved in mDC-T-cell interactions and HIV-1 latency may provide novel targets to eliminate HIV-1 latency. Electronic supplementary material The online version of this article (doi:10.1186/s12977-015-0204-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nitasha A Kumar
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC, 3004, Australia. .,Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, 3004, Australia. .,Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, 3010, Australia.
| | - Karey Cheong
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC, 3004, Australia. .,Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, 3004, Australia. .,Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, 3010, Australia.
| | - David R Powell
- Victorian Life Science Computational Initiative, Parkville, 3010, Australia. .,Monash Bioinformatics Platform, Monash University, Clayton, 3800, Australia.
| | | | - Jenny Anderson
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC, 3004, Australia. .,Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, 3004, Australia. .,Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, 3010, Australia.
| | - Vanessa A Evans
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC, 3004, Australia. .,Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, 3004, Australia. .,Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, 3010, Australia.
| | - Sharon R Lewin
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC, 3004, Australia. .,Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, 3004, Australia. .,Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, 3010, Australia.
| | - Paul U Cameron
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC, 3004, Australia. .,Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, 3004, Australia. .,Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, 3010, Australia.
| |
Collapse
|
39
|
Abstract
Hypersensitivity reactions to corticosteroids (CS) are rare in the general population, but they are not uncommon in high-risk groups such as patients who receive repeated doses of CS. Hypersensitivity reactions to steroids are broadly divided into two categories: immediate reactions, typically occurring within 1 h of drug administration, and non-immediate reactions, which manifest more than an hour after drug administration. The latter group is more common. We reviewed the literature using the search terms "hypersensitivity to steroids, adverse effects of steroids, steroid allergy, allergic contact dermatitis, corticosteroid side effects, and type I hypersensitivity" to identify studies or clinical reports of steroid hypersensitivity. We discuss the prevalence, mechanism, presentation, evaluation, and therapeutic options in corticosteroid hypersensitivity reactions. There is a paucity of literature on corticosteroid allergy, with most reports being case reports. Most reports involve non-systemic application of corticosteroids. Steroid hypersensitivity has been associated with type I IgE-mediated allergy including anaphylaxis. The overall prevalence of type I steroid hypersensitivity is estimated to be 0.3-0.5%. Allergic contact dermatitis (ACD) is the most commonly reported non-immediate hypersensitivity reaction and usually follows topical CS application. Atopic dermatitis and stasis dermatitis of the lower extremities are risk factors for the development of ACD from topical CS. Patients can also develop hypersensitivity reactions to nasal, inhaled, oral, and parenteral CS. A close and detailed evaluation is required for the clinician to confirm the presence of a true hypersensitivity reaction to the suspected drug and choose the safest alternative. Choosing an alternative CS is not only paramount to the patient's safety but also ameliorates the worry of developing an allergic, and potentially fatal, steroid hypersensitivity reaction. This evaluation becomes especially important in high-risk groups where steroids are a life-saving treatment. The assessment should be done when the patient's underlying condition is in a quiescent state.
Collapse
|
40
|
Abstract
The peer-reviewed publications in the field of autoimmunity published in 2013 represented a significant proportion of immunology articles and grew since the previous year to indicate that more immune-mediated phenomena may recognize an autoimmune mechanism and illustrated by osteoarthritis and atherosclerosis. As a result, our understanding of the mechanisms of autoimmunity is becoming the paradigm for translational research in which the progress in disease pathogenesis for both tolerance breakdown and inflammation perpetuation is rapidly followed by new treatment approaches and clinical management changes. The similarities across the autoimmune disease spectrum outnumber differences, particularly when treatments are compared. Indeed, the therapeutics of autoimmune diseases are based on a growing armamentarium that currently includes monoclonal antibodies and small molecules which act by targeting molecular markers or intracellular mediators with high specificity. Among the over 100 conditions considered as autoimmune, the common grounds are well illustrated by the data reported for systemic lupus erythematosus and rheumatoid arthritis or by the plethora of studies on Th17 cells and biomarkers, particularly serum autoantibodies. Further, we are particularly intrigued by studies on the genomics, epigenetics, and microRNA at different stages of disease development or on the safe and effective use of abatacept acting on the costimulation of T and B cells in rheumatoid arthritis. We are convinced that the data published in 2013 represent a promising background for future developments that will exponentially impact the work of laboratory and clinical scientists over the next years.
Collapse
|
41
|
Döbel T, Schäkel K. [The role of human 6-sulfo LacNAc dendritic cells (slanDCs) in autoimmunity and tumor diseases]. J Dtsch Dermatol Ges 2015; 12:874-80. [PMID: 25262889 DOI: 10.1111/ddg.12439_suppl] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dendritic cells play a central role in the regulation of immunological reactivity. The existence of functionally specialized populations of skin dendritic cells is a consequence of qualitatively different attacks on our organism. slanDCs are human inflammatory dendritic cells that are characterized by the specific expression of the carbohydrate 6-sulfo LacNAc (slan). After phenotypic maturation slanDCs are capable of producing very high amounts of proinflammatory mediators like IL-12, TNF-α, IL-1β â and IL-23. Recent data describe a potential role of slanDCs in a number of different diseases like psoriasis, lupus erythematosus but also tumor diseases and therefore open up new areas of research on their respective pathogenesis. Furthermore, as a basis of a directed therapeutic manipulation,a slanDC-specific targeting system has been developed. Future challenges of slanDC research include the elaboration of a deeper understanding of the significance of slanDCs for the regulation of adaptive and innate immune responses as well as a translation of this knowledge into therapeutic options.
Collapse
Affiliation(s)
- Thomas Döbel
- Hautklinik, Universitätsklinikum Heidelberg, Heidelberg
| | | |
Collapse
|
42
|
Abstract
BACKGROUND Lupus erythematosus is an autoimmune disease with a broad spectrum of cutaneous manifestations. The pathogenesis of lupus is based on a loss of tolerance against self antigens and can be mediated by defects in apoptosis, defects in eliminating cellular remnants and increased activation of the innate as well as the adaptive immune system. The increased activation of the innate immune system can be mediated by sensing of endogenous or exogenous nucleic acids, genetic variants in the components of the receptor cascade or disturbances in restriction of self nucleic acids. The inflammatory milieu is characterized by type I interferon expression and autoantibody production. The main trigger factors of the disease are sun exposure and viral infections. TREATMENT Lupus erythematosus is effectively treated by glucocorticosteroids. Approved alternatives for long-term treatment are antimalarial agents and the B-cell inhibitor belimumab for patients with systemic lupus erythematosus. CONCLUSION Future studies should more intensely analyse the effect of novel therapies on cutaneous manifestations to allow early detection of cutaneous lupus. Furthermore novel therapeutic strategies which specifically target the responsible pathogenetic mechanisms of the individual subtypes of lupus erythematosus are needed to improve the therapeutic success for this heterogeneous patient population.
Collapse
|
43
|
Schlitzer A, McGovern N, Ginhoux F. Dendritic cells and monocyte-derived cells: Two complementary and integrated functional systems. Semin Cell Dev Biol 2015; 41:9-22. [DOI: 10.1016/j.semcdb.2015.03.011] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 03/27/2015] [Accepted: 03/31/2015] [Indexed: 12/23/2022]
|
44
|
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that is characterized by the development of autoantibodies and immunologic attack of different organ systems, including the skin. This review aims to provide an overview of some of the pathogenic processes that may be important in the development of SLE, specifically cutaneous lupus erythematosus, and then illustrates how therapies might be tailored to modify these processes and treat disease.
Collapse
Affiliation(s)
- Mark G Kirchhof
- Department of Dermatology and Skin Science, University of British Columbia, 835 West 10th Avenue, Vancouver, British Columbia V5Z 4E8, Canada
| | - Jan P Dutz
- Department of Dermatology and Skin Science, University of British Columbia, 835 West 10th Avenue, Vancouver, British Columbia V5Z 4E8, Canada; Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, British Columbia V5Z 4H4, Canada.
| |
Collapse
|
45
|
Toma M, Wehner R, Kloß A, Hübner L, Fodelianaki G, Erdmann K, Füssel S, Zastrow S, Meinhardt M, Seliger B, Brech D, Noessner E, Tonn T, Schäkel K, Bornhäuser M, Bachmann MP, Wirth MP, Baretton G, Schmitz M. Accumulation of tolerogenic human 6-sulfo LacNAc dendritic cells in renal cell carcinoma is associated with poor prognosis. Oncoimmunology 2015; 4:e1008342. [PMID: 26155414 DOI: 10.1080/2162402x.2015.1008342] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/07/2015] [Accepted: 01/08/2015] [Indexed: 12/24/2022] Open
Abstract
Dendritic cells (DCs) essentially contribute to the induction and regulation of innate and adaptive immunity. Based on these important properties, DCs may profoundly influence tumor progression in patients. However, little is known about the role of distinct human DC subsets in primary tumors and their impact on clinical outcome. In the present study, we investigated the characteristics of human 6-sulfo LacNAc (slan) DCs in clear cell renal cell carcinoma (ccRCC). slanDCs have been shown to display various tumor-directed properties and to accumulate in tumor-draining lymph nodes from patients. When evaluating 263 ccRCC and 227 tumor-free tissue samples, we found increased frequencies of slanDCs in ccRCC tissues compared to tumor-free tissues. slanDCs were also detectable in the majority of 24 metastatic lymph nodes and 67 distant metastases from ccRCC patients. Remarkably, a higher density of slanDCs was significantly associated with a reduced progression-free, tumor-specific or overall survival of ccRCC patients. Tumor-infiltrating slanDCs displayed an immature phenotype expressing interleukin-10. ccRCC cells efficiently impaired slanDC-induced T-cell proliferation and programming as well as natural killer (NK) cell activation. In conclusion, these findings indicate that higher slanDC numbers in ccRCC tissues are associated with poor prognosis. The induction of a tolerogenic phenotype in slanDCs leading to an insufficient activation of innate and adaptive antitumor immunity may represent a novel immune escape mechanism of ccRCC. These observations may have implications for the design of therapeutic strategies that harness tumor-directed functional properties of DCs against ccRCC.
Collapse
Key Words
- CTLs, cytotoxic T cells
- DCs, dendritic cells
- FCS, fetal calf serum
- HLA, human leukocyte antigen
- IFNγ, interferonγ
- IL, interleukin
- ILT, immunoglobulin-like transcript
- LPS, lipopolysaccharide
- NK cells, natural killer cells
- PBMCs, peripheral blood mononuclear cells
- PMA, phorbol myristate acetate
- T cells
- TMAs, tissue microarrays
- TNF-α, tumor necrosis factor-α
- Th1 cells, T helper type I cells
- ccRCC, clear cell renal cell carcinoma
- dendritic cells
- renal cell carcinoma
- slan, 6-sulfo LacNAc
- tumor immunology
- tumor microenvironment
Collapse
Affiliation(s)
- Marieta Toma
- Institute of Pathology; University Hospital of Dresden ; Dresden, Germany
| | - Rebekka Wehner
- Institute of Immunology; Medical Faculty; TU Dresden ; Dresden, Germany
| | - Anja Kloß
- Institute of Immunology; Medical Faculty; TU Dresden ; Dresden, Germany
| | - Linda Hübner
- Institute of Immunology; Medical Faculty; TU Dresden ; Dresden, Germany
| | - Georgia Fodelianaki
- Institute of Immunology; Medical Faculty; TU Dresden ; Dresden, Germany ; Center for Regenerative Therapies Dresden ; Dresden, Germany
| | - Kati Erdmann
- Department of Urology; University Hospital of Dresden ; Dresden, Germany
| | - Susanne Füssel
- Department of Urology; University Hospital of Dresden ; Dresden, Germany
| | - Stefan Zastrow
- Department of Urology; University Hospital of Dresden ; Dresden, Germany
| | - Matthias Meinhardt
- Institute of Pathology; University Hospital of Dresden ; Dresden, Germany
| | - Barbara Seliger
- Institute for Medical Immunology; Martin Luther University Halle-Wittenberg ; Halle (Saale), Germany
| | - Dorothee Brech
- Institute of Molecular Immunology; Helmholtz Center Munich; German Research Center for Environmental Health Munich ; Munich, Germany
| | - Elfriede Noessner
- Institute of Molecular Immunology; Helmholtz Center Munich; German Research Center for Environmental Health Munich ; Munich, Germany
| | - Torsten Tonn
- Center for Regenerative Therapies Dresden ; Dresden, Germany ; German Red Cross Blood Service ; Dresden, Germany ; German Cancer Consortium (DKTK) ; Dresden, Germany ; German Cancer Research Center (DKFZ) ; Heidelberg, Germany
| | - Knut Schäkel
- Department of Dermatology; University Hospital of Heidelberg ; Heidelberg, Germany
| | - Martin Bornhäuser
- Center for Regenerative Therapies Dresden ; Dresden, Germany ; German Cancer Consortium (DKTK) ; Dresden, Germany ; German Cancer Research Center (DKFZ) ; Heidelberg, Germany ; Department of Medicine I; University Hospital of Dresden ; Dresden, Germany
| | - Michael P Bachmann
- Center for Regenerative Therapies Dresden ; Dresden, Germany ; German Cancer Consortium (DKTK) ; Dresden, Germany ; German Cancer Research Center (DKFZ) ; Heidelberg, Germany ; Department of Radioimmunology; Institute of Radiopharmaceutical Cancer Research; Helmholtz Center Dresden-Rossendorf ; Dresden, Germany
| | - Manfred P Wirth
- Department of Urology; University Hospital of Dresden ; Dresden, Germany ; German Cancer Consortium (DKTK) ; Dresden, Germany ; German Cancer Research Center (DKFZ) ; Heidelberg, Germany
| | - Gustavo Baretton
- Institute of Pathology; University Hospital of Dresden ; Dresden, Germany ; German Cancer Consortium (DKTK) ; Dresden, Germany ; German Cancer Research Center (DKFZ) ; Heidelberg, Germany
| | - Marc Schmitz
- Institute of Immunology; Medical Faculty; TU Dresden ; Dresden, Germany ; Center for Regenerative Therapies Dresden ; Dresden, Germany ; German Cancer Consortium (DKTK) ; Dresden, Germany ; German Cancer Research Center (DKFZ) ; Heidelberg, Germany
| |
Collapse
|
46
|
Döbel T, Schäkel K. The role of human 6-sulfo LacNAc dendritic cells (slanDCs) in autoimmunity and tumor diseases. J Dtsch Dermatol Ges 2014. [DOI: 10.1111/ddg.12439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Thomas Döbel
- Department of Dermatology; Heidelberg University Hospital; Heidelberg Germany
| | - Knut Schäkel
- Department of Dermatology; Heidelberg University Hospital; Heidelberg Germany
| |
Collapse
|
47
|
Thomas K, Dietze K, Wehner R, Metz I, Tumani H, Schultheiß T, Günther C, Schäkel K, Reichmann H, Brück W, Schmitz M, Ziemssen T. Accumulation and therapeutic modulation of 6-sulfo LacNAc(+) dendritic cells in multiple sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2014; 1:e33. [PMID: 25340085 PMCID: PMC4204231 DOI: 10.1212/nxi.0000000000000033] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 08/18/2014] [Indexed: 12/25/2022]
Abstract
Objective: To examine the potential role of 6-sulfo LacNAc+ (slan) dendritic cells (DCs) displaying pronounced proinflammatory properties in the pathogenesis of multiple sclerosis (MS). Methods: We determined the presence of slanDCs in demyelinated brain lesions and CSF samples of patients with MS. In addition, we explored the impact of methylprednisolone, interferon-β, glatiramer acetate, or natalizumab on the frequency of blood-circulating slanDCs in patients with MS. We also evaluated whether interferon-β modulates important proinflammatory capabilities of slanDCs. Results: SlanDCs accumulate in highly inflammatory brain lesions and are present in the majority of CSF samples of patients with MS. Short-term methylprednisolone administration reduces the percentage of slanDCs in blood of patients with MS and the proportion of tumor necrosis factor-α– or CD150-expressing slanDCs. Long-term interferon-β treatment decreases the percentage of blood-circulating slanDCs in contrast to glatiramer acetate or natalizumab. Furthermore, interferon-β inhibits the secretion of proinflammatory cytokines by slanDCs and their capacity to promote proliferation and differentiation of T cells. Conclusion: Accumulation of slanDCs in highly inflammatory brain lesions and their presence in CSF indicate that slanDCs may play an important role in the immunopathogenesis of MS. The reduction of blood-circulating slanDCs and the inhibition of their proinflammatory properties by methylprednisolone and interferon-β may contribute to the therapeutic efficiency of these drugs in patients with MS.
Collapse
Affiliation(s)
- Katja Thomas
- Departments of Neurology (K.T., T.S., H.R., T.Z.) and Dermatology (C.G.), University Hospital, Dresden; Institute of Immunology (K.D., R.W., M.S.), Medical Faculty, TU Dresden; Department of Neuropathology (I.M., W.B.), University Medical Centre, Göttingen; Department of Neurology (H.T.), University Hospital, Ulm; Department of Dermatology (K.S.), University Hospital, Heidelberg; and Center for Regenerative Therapies Dresden (M.S.), Dresden, Germany
| | - Kristin Dietze
- Departments of Neurology (K.T., T.S., H.R., T.Z.) and Dermatology (C.G.), University Hospital, Dresden; Institute of Immunology (K.D., R.W., M.S.), Medical Faculty, TU Dresden; Department of Neuropathology (I.M., W.B.), University Medical Centre, Göttingen; Department of Neurology (H.T.), University Hospital, Ulm; Department of Dermatology (K.S.), University Hospital, Heidelberg; and Center for Regenerative Therapies Dresden (M.S.), Dresden, Germany
| | - Rebekka Wehner
- Departments of Neurology (K.T., T.S., H.R., T.Z.) and Dermatology (C.G.), University Hospital, Dresden; Institute of Immunology (K.D., R.W., M.S.), Medical Faculty, TU Dresden; Department of Neuropathology (I.M., W.B.), University Medical Centre, Göttingen; Department of Neurology (H.T.), University Hospital, Ulm; Department of Dermatology (K.S.), University Hospital, Heidelberg; and Center for Regenerative Therapies Dresden (M.S.), Dresden, Germany
| | - Imke Metz
- Departments of Neurology (K.T., T.S., H.R., T.Z.) and Dermatology (C.G.), University Hospital, Dresden; Institute of Immunology (K.D., R.W., M.S.), Medical Faculty, TU Dresden; Department of Neuropathology (I.M., W.B.), University Medical Centre, Göttingen; Department of Neurology (H.T.), University Hospital, Ulm; Department of Dermatology (K.S.), University Hospital, Heidelberg; and Center for Regenerative Therapies Dresden (M.S.), Dresden, Germany
| | - Hayrettin Tumani
- Departments of Neurology (K.T., T.S., H.R., T.Z.) and Dermatology (C.G.), University Hospital, Dresden; Institute of Immunology (K.D., R.W., M.S.), Medical Faculty, TU Dresden; Department of Neuropathology (I.M., W.B.), University Medical Centre, Göttingen; Department of Neurology (H.T.), University Hospital, Ulm; Department of Dermatology (K.S.), University Hospital, Heidelberg; and Center for Regenerative Therapies Dresden (M.S.), Dresden, Germany
| | - Thorsten Schultheiß
- Departments of Neurology (K.T., T.S., H.R., T.Z.) and Dermatology (C.G.), University Hospital, Dresden; Institute of Immunology (K.D., R.W., M.S.), Medical Faculty, TU Dresden; Department of Neuropathology (I.M., W.B.), University Medical Centre, Göttingen; Department of Neurology (H.T.), University Hospital, Ulm; Department of Dermatology (K.S.), University Hospital, Heidelberg; and Center for Regenerative Therapies Dresden (M.S.), Dresden, Germany
| | - Claudia Günther
- Departments of Neurology (K.T., T.S., H.R., T.Z.) and Dermatology (C.G.), University Hospital, Dresden; Institute of Immunology (K.D., R.W., M.S.), Medical Faculty, TU Dresden; Department of Neuropathology (I.M., W.B.), University Medical Centre, Göttingen; Department of Neurology (H.T.), University Hospital, Ulm; Department of Dermatology (K.S.), University Hospital, Heidelberg; and Center for Regenerative Therapies Dresden (M.S.), Dresden, Germany
| | - Knut Schäkel
- Departments of Neurology (K.T., T.S., H.R., T.Z.) and Dermatology (C.G.), University Hospital, Dresden; Institute of Immunology (K.D., R.W., M.S.), Medical Faculty, TU Dresden; Department of Neuropathology (I.M., W.B.), University Medical Centre, Göttingen; Department of Neurology (H.T.), University Hospital, Ulm; Department of Dermatology (K.S.), University Hospital, Heidelberg; and Center for Regenerative Therapies Dresden (M.S.), Dresden, Germany
| | - Heinz Reichmann
- Departments of Neurology (K.T., T.S., H.R., T.Z.) and Dermatology (C.G.), University Hospital, Dresden; Institute of Immunology (K.D., R.W., M.S.), Medical Faculty, TU Dresden; Department of Neuropathology (I.M., W.B.), University Medical Centre, Göttingen; Department of Neurology (H.T.), University Hospital, Ulm; Department of Dermatology (K.S.), University Hospital, Heidelberg; and Center for Regenerative Therapies Dresden (M.S.), Dresden, Germany
| | - Wolfgang Brück
- Departments of Neurology (K.T., T.S., H.R., T.Z.) and Dermatology (C.G.), University Hospital, Dresden; Institute of Immunology (K.D., R.W., M.S.), Medical Faculty, TU Dresden; Department of Neuropathology (I.M., W.B.), University Medical Centre, Göttingen; Department of Neurology (H.T.), University Hospital, Ulm; Department of Dermatology (K.S.), University Hospital, Heidelberg; and Center for Regenerative Therapies Dresden (M.S.), Dresden, Germany
| | - Marc Schmitz
- Departments of Neurology (K.T., T.S., H.R., T.Z.) and Dermatology (C.G.), University Hospital, Dresden; Institute of Immunology (K.D., R.W., M.S.), Medical Faculty, TU Dresden; Department of Neuropathology (I.M., W.B.), University Medical Centre, Göttingen; Department of Neurology (H.T.), University Hospital, Ulm; Department of Dermatology (K.S.), University Hospital, Heidelberg; and Center for Regenerative Therapies Dresden (M.S.), Dresden, Germany
| | - Tjalf Ziemssen
- Departments of Neurology (K.T., T.S., H.R., T.Z.) and Dermatology (C.G.), University Hospital, Dresden; Institute of Immunology (K.D., R.W., M.S.), Medical Faculty, TU Dresden; Department of Neuropathology (I.M., W.B.), University Medical Centre, Göttingen; Department of Neurology (H.T.), University Hospital, Ulm; Department of Dermatology (K.S.), University Hospital, Heidelberg; and Center for Regenerative Therapies Dresden (M.S.), Dresden, Germany
| |
Collapse
|
48
|
Martin JC, Baeten DL, Josien R. Emerging role of IL-17 and Th17 cells in systemic lupus erythematosus. Clin Immunol 2014; 154:1-12. [DOI: 10.1016/j.clim.2014.05.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 05/10/2014] [Accepted: 05/14/2014] [Indexed: 12/14/2022]
|
49
|
Sommer U, Larsson B, Tuve S, Wehner R, Zimmermann N, Kramer M, Kloβ A, Günther C, Babatz J, Schmelz R, Brückner S, Schetelig J, Bornhäuser M, Schäkel K, Bachmann MP, Aust D, Baretton G, Schmitz M. Proinflammatory human 6-sulfo LacNAc-positive dendritic cells accumulate in intestinal acute graft-versus-host disease. Haematologica 2014; 99:e86-9. [PMID: 24682513 DOI: 10.3324/haematol.2013.101071] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Ulrich Sommer
- Institute of Pathology, University Hospital of Dresden, Dresden, Germany
| | - Brit Larsson
- Department of Medicine I, University Hospital of Dresden, Dresden, Germany
| | - Sebastian Tuve
- Department of Medicine I, University Hospital of Dresden, Dresden, Germany
| | - Rebekka Wehner
- Institute of Immunology, Medical Faculty, Dresden University of Technology, Dresden, Germany
| | - Nick Zimmermann
- Department of Dermatology, University Hospital of Dresden, Dresden, Germany
| | - Michael Kramer
- Department of Medicine I, University Hospital of Dresden, Dresden, Germany
| | - Anja Kloβ
- Institute of Immunology, Medical Faculty, Dresden University of Technology, Dresden, Germany
| | - Claudia Günther
- Department of Dermatology, University Hospital of Dresden, Dresden, Germany
| | - Jana Babatz
- Institute of Immunology, Medical Faculty, Dresden University of Technology, Dresden, Germany
| | - Renate Schmelz
- Institute of Immunology, Medical Faculty, Dresden University of Technology, Dresden, Germany
| | - Stefan Brückner
- Institute of Immunology, Medical Faculty, Dresden University of Technology, Dresden, Germany
| | - Johannes Schetelig
- Department of Medicine I, University Hospital of Dresden, Dresden, Germany
| | - Martin Bornhäuser
- Department of Medicine I, University Hospital of Dresden, Dresden, Germany Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Knut Schäkel
- Department of Dermatology, University Hospital of Heidelberg, Dresden, Germany
| | - Michael Philipp Bachmann
- Institute of Immunology, Medical Faculty, Dresden University of Technology, Dresden, Germany Center for Regenerative Therapies Dresden, Dresden, Germany Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Deparment of Radioimmunology, Dresden, Germany
| | - Daniela Aust
- Institute of Pathology, University Hospital of Dresden, Dresden, Germany
| | - Gustavo Baretton
- Institute of Pathology, University Hospital of Dresden, Dresden, Germany
| | - Marc Schmitz
- Institute of Immunology, Medical Faculty, Dresden University of Technology, Dresden, Germany Center for Regenerative Therapies Dresden, Dresden, Germany
| |
Collapse
|
50
|
Deciphering the stromal and hematopoietic cell network of the adventitia from non-aneurysmal and aneurysmal human aorta. PLoS One 2014; 9:e89983. [PMID: 24587165 PMCID: PMC3937418 DOI: 10.1371/journal.pone.0089983] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 01/23/2014] [Indexed: 01/09/2023] Open
Abstract
Aneurysm is associated to a complex remodeling of arteries that affects all their layers. Although events taking place in the intima and the media have received a particular attention, molecular and cellular events taking place in the adventitia have started to be deciphered only recently. In this study, we have precisely described the composition and distribution of stromal and hematopoietic cells in human arterial adventitia, both at steady state and in the setting of aortic aneurysm. Using polychromatic immunofluorescent and flow cytometry analyses, we observed that unlike the medial layer (which comprises mostly macrophages and T cells among leukocytes), the adventitia comprises a much greater variety of leukocytes. We observed an altered balance in macrophages subsets in favor of M2-like macrophages, an increased proliferation of macrophages, a greater number of all stromal cells in aneurysmal aortas. We also confirmed that in this pathological setting, adventitia comprised blood vessels and arterial tertiary lymphoid organs (ATLOs), which contained also M-DC8+ dendritic cells (slanDCs) that could participate in the induction of T-cell responses. Finally, we showed that lymphatic vessels can be detected in aneurysmal adventitia, the functionality of which will have to be evaluated in future studies. All together, these observations provide an integrative outlook of the stromal and hematopoietic cell network of the human adventitia both at steady state and in the context of aneurysm.
Collapse
|