101
|
Schlegel M, Koelwyn GJ, Moore KJ. Connecting Transcriptional and Functional Macrophage Heterogeneity in Atherosclerosis. Circ Res 2019; 125:1052-1054. [PMID: 31804906 DOI: 10.1161/circresaha.119.316168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Martin Schlegel
- From the Department of Medicine, Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine
| | - Graeme J Koelwyn
- From the Department of Medicine, Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine
| | - Kathryn J Moore
- From the Department of Medicine, Marc and Ruti Bell Program for Vascular Biology and Disease, The Leon H. Charney Division of Cardiology, New York University School of Medicine
| |
Collapse
|
102
|
El Shikh MEM, El Sayed R, Nerviani A, Goldmann K, John CR, Hands R, Fossati-Jimack L, Lewis MJ, Pitzalis C. Extracellular traps and PAD4 released by macrophages induce citrullination and auto-antibody production in autoimmune arthritis. J Autoimmun 2019; 105:102297. [PMID: 31277965 DOI: 10.1016/j.jaut.2019.06.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/20/2019] [Accepted: 06/24/2019] [Indexed: 11/18/2022]
Abstract
The mechanisms underlying the transition of rheumatoid arthritis (RA) systemic autoimmunity to the joints remain largely unknown. Here, we demonstrate that macrophages in the secondary lymphoid organs (SLOs) and synovial ectopic lymphoid-like structures (ELSs) express peptidylarginine deiminase 4 (PAD4) in murine collagen induced arthritis (CIA) and synovial biopsies from RA patients. Moreover, peptidyl citrulline colocalized with macrophages in SLOs and ELSs, and depletion of macrophages in CIA decreased lymphoid tissue citrullination and serum anti-citrullinated protein/peptide antibody (ACPA) levels. Furthermore, PAD was released from activated murine and RA synovial tissue and fluid (SF) macrophages which functionally deiminated extracellular proteins/peptides in vitro. Additionally, activated murine and SF macrophages displayed macrophage extracellular trap formation (METosis) and release of intracellular citrullinated histones. Moreover, presentation of citrullinated proteins induced ACPA production in vitro. Thus, lymphoid tissue macrophages contribute to self-antigen citrullination and ACPA production, indicating that their selective targeting would potentially ameliorate citrullination-dependent autoimmune disorders.
Collapse
Affiliation(s)
- Mohey Eldin M El Shikh
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Riham El Sayed
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Alessandra Nerviani
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Katriona Goldmann
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Christopher Robert John
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Rebecca Hands
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Liliane Fossati-Jimack
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK.
| |
Collapse
|
103
|
Ramel D, Gayral S, Sarthou MK, Augé N, Nègre-Salvayre A, Laffargue M. Immune and Smooth Muscle Cells Interactions in Atherosclerosis: How to Target a Breaking Bad Dialogue? Front Pharmacol 2019; 10:1276. [PMID: 31824304 PMCID: PMC6882774 DOI: 10.3389/fphar.2019.01276] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/07/2019] [Indexed: 12/14/2022] Open
Abstract
Inflammation is a well-known pathophysiological factor of atherosclerosis but its therapeutic targeting has long been ignored. However, recent advances in the understanding of the immune mechanisms implicated in atherosclerosis have unveiled several therapeutic targets currently undergoing clinical trials. These studies have also shed light on a dialogue between the immune compartment and vascular smooth muscle cells (VSMCs) that plays a critical role in atherosclerotic disease initiation, progression, and stabilization. Our review focuses on the link between cellular and soluble immune effectors and VSMC behavior at different phases of the pathology. Furthermore, we discuss the potential targeting of these interactions to efficiently prevent cardiovascular diseases.
Collapse
Affiliation(s)
- Damien Ramel
- Department of Vascular Biology of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse 3, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse, France
| | - Stéphanie Gayral
- Department of Vascular Biology of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse 3, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse, France
| | - Marie-Kerguelen Sarthou
- Department of Vascular Biology of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse 3, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse, France
| | - Nathalie Augé
- Department of Vascular Biology of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse 3, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse, France
| | - Anne Nègre-Salvayre
- Department of Vascular Biology of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse 3, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse, France
| | - Muriel Laffargue
- Department of Vascular Biology of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Université de Toulouse 3, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1048, Toulouse, France
| |
Collapse
|
104
|
Abstract
There is now overwhelming experimental and clinical evidence that atherosclerosis is a chronic inflammatory disease. Lessons from genome-wide association studies, advanced in vivo imaging techniques, transgenic lineage tracing mice, and clinical interventional studies have shown that both innate and adaptive immune mechanisms can accelerate or curb atherosclerosis. Here, we summarize and discuss the pathogenesis of atherosclerosis with a focus on adaptive immunity. We discuss some limitations of animal models and the need for models that are tailored to better translate to human atherosclerosis and ultimately progress in prevention and treatment.
Collapse
Affiliation(s)
- Dennis Wolf
- From the Department of Cardiology and Angiology I, University Heart Center Freiburg, Germany (D.W.).,Faculty of Medicine, University of Freiburg, Germany (D.W.)
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (K.L.).,Department of Bioengineering, University of California San Diego, La Jolla (K.L.)
| |
Collapse
|
105
|
Martini E, Kunderfranco P, Peano C, Carullo P, Cremonesi M, Schorn T, Carriero R, Termanini A, Colombo FS, Jachetti E, Panico C, Faggian G, Fumero A, Torracca L, Molgora M, Cibella J, Pagiatakis C, Brummelman J, Alvisi G, Mazza EMC, Colombo MP, Lugli E, Condorelli G, Kallikourdis M. Single-Cell Sequencing of Mouse Heart Immune Infiltrate in Pressure Overload-Driven Heart Failure Reveals Extent of Immune Activation. Circulation 2019; 140:2089-2107. [PMID: 31661975 DOI: 10.1161/circulationaha.119.041694] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Inflammation is a key component of cardiac disease, with macrophages and T lymphocytes mediating essential roles in the progression to heart failure. Nonetheless, little insight exists on other immune subsets involved in the cardiotoxic response. METHODS Here, we used single-cell RNA sequencing to map the cardiac immune composition in the standard murine nonischemic, pressure-overload heart failure model. By focusing our analysis on CD45+ cells, we obtained a higher resolution identification of the immune cell subsets in the heart, at early and late stages of disease and in controls. We then integrated our findings using multiparameter flow cytometry, immunohistochemistry, and tissue clarification immunofluorescence in mouse and human. RESULTS We found that most major immune cell subpopulations, including macrophages, B cells, T cells and regulatory T cells, dendritic cells, Natural Killer cells, neutrophils, and mast cells are present in both healthy and diseased hearts. Most cell subsets are found within the myocardium, whereas mast cells are found also in the epicardium. Upon induction of pressure overload, immune activation occurs across the entire range of immune cell types. Activation led to upregulation of key subset-specific molecules, such as oncostatin M in proinflammatory macrophages and PD-1 in regulatory T cells, that may help explain clinical findings such as the refractivity of patients with heart failure to anti-tumor necrosis factor therapy and cardiac toxicity during anti-PD-1 cancer immunotherapy, respectively. CONCLUSIONS Despite the absence of infectious agents or an autoimmune trigger, induction of disease leads to immune activation that involves far more cell types than previously thought, including neutrophils, B cells, Natural Killer cells, and mast cells. This opens up the field of cardioimmunology to further investigation by using toolkits that have already been developed to study the aforementioned immune subsets. The subset-specific molecules that mediate their activation may thus become useful targets for the diagnostics or therapy of heart failure.
Collapse
Affiliation(s)
- Elisa Martini
- Adaptive Immunity Laboratory (E.M., M.C., M.K.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Paolo Kunderfranco
- Bioinformatics Unit (P.K., R.C., A.T.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Clelia Peano
- Genomic Unit (C. Peano, J.C.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, Rozzano, Italy (C. Peano, P.C., G.C.)
| | - Pierluigi Carullo
- Department of Cardiovascular Medicine (P.C., C. Panico, C. Pagiatakis, G.C.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, Rozzano, Italy (C. Peano, P.C., G.C.)
| | - Marco Cremonesi
- Adaptive Immunity Laboratory (E.M., M.C., M.K.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Tilo Schorn
- Advanced Imaging Unit (T.S.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Roberta Carriero
- Bioinformatics Unit (P.K., R.C., A.T.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Alberto Termanini
- Bioinformatics Unit (P.K., R.C., A.T.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Federico Simone Colombo
- Flow Cytometry Core (F.S.C., E.L.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Elena Jachetti
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy (E.J., M.P.C.)
| | - Cristina Panico
- Department of Cardiovascular Medicine (P.C., C. Panico, C. Pagiatakis, G.C.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Giuseppe Faggian
- Department of Cardiac Surgery, University of Verona, Italy (G.F.)
| | - Andrea Fumero
- Cardiac Surgery Division, Department of Cardiovascular Medicine (A.F., L.T.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Lucia Torracca
- Cardiac Surgery Division, Department of Cardiovascular Medicine (A.F., L.T.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Martina Molgora
- Laboratory of Experimental Immunopathology (M.M.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Javier Cibella
- Genomic Unit (C. Peano, J.C.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Christina Pagiatakis
- Department of Cardiovascular Medicine (P.C., C. Panico, C. Pagiatakis, G.C.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Jolanda Brummelman
- Laboratory of Translational Immunology (J.B., G.A., E.M.C., E.L.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Giorgia Alvisi
- Laboratory of Translational Immunology (J.B., G.A., E.M.C., E.L.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Emilia Maria Cristina Mazza
- Laboratory of Translational Immunology (J.B., G.A., E.M.C., E.L.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Mario Paolo Colombo
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy (E.J., M.P.C.)
| | - Enrico Lugli
- Flow Cytometry Core (F.S.C., E.L.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Laboratory of Translational Immunology (J.B., G.A., E.M.C., E.L.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Gianluigi Condorelli
- Department of Cardiovascular Medicine (P.C., C. Panico, C. Pagiatakis, G.C.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, Rozzano, Italy (C. Peano, P.C., G.C.).,Humanitas University, Pieve Emanuele, Italy (G.C., M.K.)
| | - Marinos Kallikourdis
- Adaptive Immunity Laboratory (E.M., M.C., M.K.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Humanitas University, Pieve Emanuele, Italy (G.C., M.K.)
| |
Collapse
|
106
|
McArdle S, Buscher K, Ghosheh Y, Pramod AB, Miller J, Winkels H, Wolf D, Ley K. Migratory and Dancing Macrophage Subsets in Atherosclerotic Lesions. Circ Res 2019; 125:1038-1051. [PMID: 31594470 DOI: 10.1161/circresaha.119.315175] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
RATIONALE Macrophages are essential regulators of atherosclerosis. They secrete cytokines, process lipoproteins and cholesterol, and take up apoptotic cells. Multiple subsets of plaque macrophages exist and their differential roles are emerging. OBJECTIVE Here, we explore macrophage heterogeneity in atherosclerosis plaques using transgenic fluorescent mice in which subsets of macrophages are labeled by GFP (green fluorescent protein), YFP (yellow fluorescent protein), neither, or both. The objective was to define migration patterns of the visible subsets and relate them to their phenotypes and transcriptomes. METHODS AND RESULTS Apoe-/- Cx3cr1GFP Cd11cYFP mice have 4 groups of macrophages in their aortas. The 3 visible subsets show varying movement characteristics. GFP and GFP+YFP+ macrophages extend and retract dendritic processes, dancing on the spot with little net movement while YFP macrophages have a more rounded shape and migrate along the arteries. RNA sequencing of sorted cells revealed significant differences in the gene expression patterns of the 4 subsets defined by GFP and YFP expression, especially concerning chemokine and cytokine expression, matrix remodeling, and cell shape dynamics. Gene set enrichment analysis showed that GFP+ cells have similar transcriptomes to cells found in arteries with tertiary lymphoid organs and regressing plaques while YFP+ cells were associated with progressing and stable plaques. CONCLUSIONS The combination of quantitative intravital imaging with deep transcriptomes identified 4 subsets of vascular macrophages in atherosclerosis that have unique transcriptomic profiles. Our data link vascular macrophage transcriptomes to their in vivo migratory function. Future work on the functional significance of the change in gene expression and motility characteristics will be needed to fully understand how these subsets contribute to disease progression.
Collapse
Affiliation(s)
- Sara McArdle
- From the Microscopy Core Facility (S.M.), La Jolla Institute for Immunology, San Diego, CA.,Division of Inflammation Biology (S.M., K.B., Y.G., A.B.P., J.M., H.W., D.W., K.L.), La Jolla Institute for Immunology, San Diego, CA
| | - Konrad Buscher
- Division of Inflammation Biology (S.M., K.B., Y.G., A.B.P., J.M., H.W., D.W., K.L.), La Jolla Institute for Immunology, San Diego, CA.,Department of Nephrology and Rheumatology, University Hospital Muenster, German (K.B.)
| | - Yanal Ghosheh
- Division of Inflammation Biology (S.M., K.B., Y.G., A.B.P., J.M., H.W., D.W., K.L.), La Jolla Institute for Immunology, San Diego, CA
| | - Akula Bala Pramod
- Division of Inflammation Biology (S.M., K.B., Y.G., A.B.P., J.M., H.W., D.W., K.L.), La Jolla Institute for Immunology, San Diego, CA
| | - Jacqueline Miller
- Division of Inflammation Biology (S.M., K.B., Y.G., A.B.P., J.M., H.W., D.W., K.L.), La Jolla Institute for Immunology, San Diego, CA
| | - Holger Winkels
- Division of Inflammation Biology (S.M., K.B., Y.G., A.B.P., J.M., H.W., D.W., K.L.), La Jolla Institute for Immunology, San Diego, CA
| | - Dennis Wolf
- Division of Inflammation Biology (S.M., K.B., Y.G., A.B.P., J.M., H.W., D.W., K.L.), La Jolla Institute for Immunology, San Diego, CA.,University Heart Center and Medical Center, University of Freiburg, Germany (D.W.)
| | - Klaus Ley
- Division of Inflammation Biology (S.M., K.B., Y.G., A.B.P., J.M., H.W., D.W., K.L.), La Jolla Institute for Immunology, San Diego, CA.,Department of Bioengineering, University of California, San Diego (K.L.)
| |
Collapse
|
107
|
Li H, Wang X, Lu X, Zhu H, Li S, Duan S, Zhao X, Zhang F, Alterovitz G, Wang F, Li Q, Tian XL, Xu M. Co-expression network analysis identified hub genes critical to triglyceride and free fatty acid metabolism as key regulators of age-related vascular dysfunction in mice. Aging (Albany NY) 2019; 11:7620-7638. [PMID: 31514170 PMCID: PMC6781998 DOI: 10.18632/aging.102275] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/05/2019] [Indexed: 12/19/2022]
Abstract
Background: Aging has often been linked to age-related vascular disorders. The elucidation of the putative genes and pathways underlying vascular aging likely provides useful insights into vascular diseases at advanced ages. Transcriptional regulatory network analysis is the key to describing genetic interactions between molecular regulators and their target gene transcriptionally changed during vascular aging. Results: A total of 469 differentially expressed genes were parsed into 6 modules. Among the incorporated sample traits, the most significant module related to vascular aging was associated with triglyceride and enriched with biological terms like proteolysis, blood circulation, and circulatory system process. The module associated with triglyceride was preserved in an independent microarray dataset, indicating the robustness of the identified vascular aging-related subnetwork. Additionally, Enpp5, Fez1, Kif1a, F3, H2-Q7, and their interacting miRNAs mmu-miR-449a, mmu-miR-449c, mmu-miR-34c, mmu-miR-34b-5p, mmu-miR-15a, and mmu-let-7, exhibited the most connectivity with external lipid-related traits. Transcriptional alterations of the hub genes Enpp5, Fez1, Kif1a, and F3, and the interacting microRNAs mmu-miR-34c, mmu-miR-34b-5p, mmu-let-7, mmu-miR-449a, and mmu-miR-449c were confirmed. Conclusion: Our findings demonstrate that triglyceride and free fatty acid-related genes are key regulators of age-related vascular dysfunction in mice and show that the hub genes for Enpp5, Fez1, Kif1a, and F3 as well as their interacting miRNAs mmu-miR-34c, mmu-miR-34b-5p, mmu-let-7, mmu-miR-449a, and mmu-miR-449c, could serve as potential biomarkers in vascular aging. Methods: The microarray gene expression profiles of aorta samples from 6-month old mice (n=6) and 20-month old mice (n=6) were processed to identify nominal differentially expressed genes. These nominal differentially expressed genes were subjected to a weighted gene co-expression network analysis. A network-driven integrative analysis with microRNAs and transcription factors was performed to define significant modules and underlying regulatory pathways associated with vascular aging, and module preservation test was conducted to validate the age-related modules based on an independent microarray gene expression dataset in mice aorta samples including three 32-week old wild-type mice (around 6-month old) and three 78-week old wild-type mice (around 20-month old). Gene ontology and protein-protein interaction analyses were conducted to determine the hub genes as potential biomarkers in the progress of vascular aging. The hub genes were further validated with quantitative real-time polymerase chain reaction in aorta samples from 20 young (6-month old) mice and 20 old (20-month old) mice.
Collapse
Affiliation(s)
- Huimin Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China.,Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Xinhui Wang
- School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xinyue Lu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Hongxin Zhu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Sheng Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Shiwei Duan
- , Medical Genetics Center, School of Medicine, Ningbo University, Ningbo 315000, China
| | - Xinzhi Zhao
- International Peace Maternity and Child Health Hospital of China Affiliated to Shanghai Jiao Tong University, Shanghai 200030, China
| | | | - Gil Alterovitz
- Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Fudi Wang
- School of Public Health, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qiang Li
- Translational Medical Center for Development and Disease, Institute of Pediatrics, Shanghai Key Laboratory of Birth Defect, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiao-Li Tian
- Department of Human Population Genetics, Human Aging Research Institute and School of Life Science, Nanchang University, Nanchang 330031, China
| | - Mingqing Xu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China.,Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
108
|
Libby P, Buring JE, Badimon L, Hansson GK, Deanfield J, Bittencourt MS, Tokgözoğlu L, Lewis EF. Atherosclerosis. Nat Rev Dis Primers 2019; 5:56. [PMID: 31420554 DOI: 10.1038/s41572-019-0106-z] [Citation(s) in RCA: 1706] [Impact Index Per Article: 284.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2019] [Indexed: 12/12/2022]
Abstract
Atherosclerosis, the formation of fibrofatty lesions in the artery wall, causes much morbidity and mortality worldwide, including most myocardial infarctions and many strokes, as well as disabling peripheral artery disease. Development of atherosclerotic lesions probably requires low-density lipoprotein, a particle that carries cholesterol through the blood. Other risk factors for atherosclerosis and its thrombotic complications include hypertension, cigarette smoking and diabetes mellitus. Increasing evidence also points to a role of the immune system, as emerging risk factors include inflammation and clonal haematopoiesis. Studies of the cell and molecular biology of atherogenesis have provided considerable insight into the mechanisms that link all these risk factors to atheroma development and the clinical manifestations of this disease. An array of diagnostic techniques, both invasive (such as selective coronary arteriography) and noninvasive (such as blood biomarkers, stress testing, CT and nuclear scanning), permit assessment of cardiovascular disease risk and targeting of therapies. An expanding armamentarium of therapies that can modify risk factors and confer clinical benefit is available; however, we face considerable challenge in providing equitable access to these treatments and in maximizing adherence. Yet, the clinical application of the fruits of research has advanced preventive strategies, enhanced clinical outcomes in affected individuals, and improved their quality of life. Rapidly accelerating knowledge and continued research promise to provide further progress in combating this common chronic disease.
Collapse
Affiliation(s)
- Peter Libby
- Department of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Julie E Buring
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Lina Badimon
- Centre d'Investigació Cardiovascular CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Göran K Hansson
- Center for Molecular Medicine, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - John Deanfield
- Institute of Cardiovascular Sciences, University College London, London, UK
| | - Márcio Sommer Bittencourt
- Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, São Paulo, Brazil.,Faculdade Israelita de Ciencias da Saude Albert Einstein, São Paulo, Brazil.,DASA, São Paulo, Brazil
| | | | - Eldrin F Lewis
- Department of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
109
|
Lin L, Hu X, Zhang H, Hu H. Tertiary Lymphoid Organs in Cancer Immunology: Mechanisms and the New Strategy for Immunotherapy. Front Immunol 2019; 10:1398. [PMID: 31281318 PMCID: PMC6596321 DOI: 10.3389/fimmu.2019.01398] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 06/03/2019] [Indexed: 02/05/2023] Open
Abstract
The immune system plays pivotal roles in the occurrence and progression of cancers. As blockade of immune-checkpoint has been proven effective at improving anti-tumor immune response in multiple tumor types, the tumor immunotherapy still faces many challenges. Emerging evidence indicates lymphoid organ-like structures, also called tertiary lymphoid organs (TLOs) or ectopic lymphoid organs (ELOs), have been identified in cancers, as the result of lymphoid neoorganogenesis. The prognostic value of TLOs in cancer patients has been evaluated with debates, however, such well-organized lymphoid structures in the site of cancer indicate TLOs are the important modulators of cancer immunological microenvironment. TLOs have attracted remarkable efforts to investigate their neoorganogenesis and function in immune responses, aiming to develop new strategies for cancer immunotherapy. In this review, we summarize the current understandings about the molecular and cellular mechanisms governing the formation and function of TLOs in immune responses against cancer.
Collapse
Affiliation(s)
- Liangbin Lin
- The State Key Laboratory of Biotherapy, Department of Rheumatology and Immunology, Collaboration and Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Hu
- The State Key Laboratory of Biotherapy, Department of Rheumatology and Immunology, Collaboration and Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Huiyuan Zhang
- The State Key Laboratory of Biotherapy, Department of Rheumatology and Immunology, Collaboration and Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongbo Hu
- The State Key Laboratory of Biotherapy, Department of Rheumatology and Immunology, Collaboration and Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
110
|
Arakawa K, Ishigami T, Nakai-Sugiyama M, Chen L, Doi H, Kino T, Minegishi S, Saigoh-Teranaka S, Sasaki-Nakashima R, Hibi K, Kimura K, Tamura K. Lubiprostone as a potential therapeutic agent to improve intestinal permeability and prevent the development of atherosclerosis in apolipoprotein E-deficient mice. PLoS One 2019; 14:e0218096. [PMID: 31206525 PMCID: PMC6576757 DOI: 10.1371/journal.pone.0218096] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/24/2019] [Indexed: 12/17/2022] Open
Abstract
The interaction between atherosclerosis and commensal microbes through leaky gut syndrome (LGS), which is characterized by impaired intestinal permeability and the introduction of undesired pathogens into the body, has not been fully elucidated. Our aim was to investigate the potential role of a ClC-2 chloride channel activator, lubiprostone, which is reported to have beneficial effects on LGS, in the development of atherosclerosis in apolipoprotein E–deficient (ApoE-/-) mice. After a 15-week feeding period of a Western diet (WD), ApoE-/- mice were treated with a Western-type diet (WD) alone or WD with oral supplementation of lubiprostone for 10 weeks. This feeding protocol was followed by experimental evaluation of LGS and atherosclerotic lesions in the aorta. In mice with lubiprostone, in vivo translocation of orally administered 4-kDa FITC-dextran was significantly improved, and RNA expression of the epithelial tight junction proteins, Zo-1 and occludin, was significantly up-regulated in the ileum, compared to the WD alone group, suggesting a possible reversal of WD-induced intestinal barrier dysfunction. As a result, WD-induced exacerbation of atherosclerotic lesion formation was reduced by 69% in longitudinally opened aortas and 26% in aortic root regions. In addition, there was a significant decrease in circulating immunoglobulin level, followed by an attenuation of inflammatory responses in the perivascular adipose tissue, as evidenced by reduced expression of pro-inflammatory cytokines and chemokines. Lubiprostone attenuates atherosclerosis by ameliorating LGS-induced inflammation through the restoration of the intestinal barrier. These findings raise the possibility of targeting LGS for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Kentaro Arakawa
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University, School of Medicine, Kanagawa, Japan
| | - Tomoaki Ishigami
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University, School of Medicine, Kanagawa, Japan
- * E-mail:
| | - Michiko Nakai-Sugiyama
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University, School of Medicine, Kanagawa, Japan
| | - Lin Chen
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University, School of Medicine, Kanagawa, Japan
| | - Hiroshi Doi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University, School of Medicine, Kanagawa, Japan
| | - Tabito Kino
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University, School of Medicine, Kanagawa, Japan
| | - Shintaro Minegishi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University, School of Medicine, Kanagawa, Japan
| | - Sae Saigoh-Teranaka
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University, School of Medicine, Kanagawa, Japan
| | - Rie Sasaki-Nakashima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University, School of Medicine, Kanagawa, Japan
| | - Kiyoshi Hibi
- Division of Cardiology, Yokohama City University Medical Center, Kanagawa, Japan
| | - Kazuo Kimura
- Division of Cardiology, Yokohama City University Medical Center, Kanagawa, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University, School of Medicine, Kanagawa, Japan
| |
Collapse
|
111
|
Stock AD, Der E, Gelb S, Huang M, Weidenheim K, Ben-Zvi A, Putterman C. Tertiary lymphoid structures in the choroid plexus in neuropsychiatric lupus. JCI Insight 2019; 4:124203. [PMID: 31167973 PMCID: PMC6629135 DOI: 10.1172/jci.insight.124203] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 04/23/2019] [Indexed: 12/20/2022] Open
Abstract
The central nervous system manifestations of systemic lupus erythematosus (SLE) remain poorly understood. Given the well-defined role of autoantibodies in other lupus manifestations, extensive work has gone into the identification of neuropathic autoantibodies. However, attempts to translate these findings to patients with SLE have yielded mixed results. We used the MRL/MpJ-Faslpr/lpr mouse, a well-established, spontaneous model of SLE, to establish the immune effectors responsible for brain disease. Transcriptomic analysis of the MRL/MpJ-Faslpr/lpr choroid plexus revealed an expression signature driving tertiary lymphoid structure formation, including chemokines related to stromal reorganization and lymphocyte compartmentalization. Additionally, transcriptional profiles indicated various stages of lymphocyte activation and germinal center formation. The extensive choroid plexus infiltrate present in MRL/MpJ-Faslpr/lpr mice with overt neurobehavioral deficits included locally proliferating B and T cells, intercellular interactions between lymphocytes and antigen-presenting cells, as well as evidence for in situ somatic hypermutation and class switch recombination. Furthermore, the choroid plexus was a site for trafficking lymphocytes into the brain. Finally, histological evaluation in human lupus patients with neuropsychiatric manifestations revealed increased leukocyte migration through the choroid plexus. These studies identify a potential new pathway underlying neuropsychiatric lupus and support tertiary lymphoid structure formation in the choroid plexus as a novel mechanism of brain-immune interfacing.
Collapse
Affiliation(s)
- Ariel D. Stock
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, New York, USA
| | - Evan Der
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, New York, USA
| | - Sivan Gelb
- Department of Developmental Biology and Cancer Research, Faculty of Medicine, The Hebrew University, Ein-Kerem, Jerusalem, Israel
| | - Michelle Huang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, New York, USA
| | | | - Ayal Ben-Zvi
- Department of Developmental Biology and Cancer Research, Faculty of Medicine, The Hebrew University, Ein-Kerem, Jerusalem, Israel
| | - Chaim Putterman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, New York, USA
- Division of Rheumatology, Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
112
|
Guedj K, Abitbol Y, Cazals-Hatem D, Morvan M, Maggiori L, Panis Y, Bouhnik Y, Caligiuri G, Corcos O, Nicoletti A. Adipocytes orchestrate the formation of tertiary lymphoid organs in the creeping fat of Crohn's disease affected mesentery. J Autoimmun 2019; 103:102281. [PMID: 31171476 DOI: 10.1016/j.jaut.2019.05.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/14/2019] [Accepted: 05/14/2019] [Indexed: 12/21/2022]
Abstract
The formation of tertiary lymphoid organs (TLOs) is orchestrated by the stromal cells of tissues chronically submitted to inflammatory stimuli, in order to uphold specific adaptive immune responses. We have recently shown that the smooth muscle cells of the arterial wall orchestrate the formation of the TLOs associated with atherosclerosis in response to the local release of TNF-α. Observational studies have recently documented the presence of structures resembling TLOs the creeping fat that develops in the mesentery of patients with Crohn's disease (CD), an inflammatory condition combining a complex and as yet not elucidated infectious and autoimmune responses. We have performed a comprehensive analysis of the TLO structures in order to decipher the mechanism leading to their formation in the mesentery of CD patients, and assessed the effect of infectious and/or inflammatory inducers on the potential TLO-organizer functions of adipocytes. Quantitative analysis showed that both T and B memory cells, as well as plasma cells, are enriched in the CD-affected mesentery, as compared with tissue from control subjects. Immunohistochemistry revealed that these cells are concentrated within the creeping fat of CD patients, in the vicinity of transmural lesions; that T and B cells are compartmentalized in clearly distinct areas; that they are supplied by post-capillary high endothelial venules and drained by lymphatic vessels indicating that these nodules are fully mature TLOs. Organ culture showed that mesenteric tissue samples from CD patients contained greater amounts of adipocyte-derived chemokines and the use of the conditioned medium from these cultures in functional assays was able to actively recruit T and B lymphocytes. Finally, the production of chemokines involved in TLO formation by 3T3-L1 adipocytes was directly elicited by a combination of TNF-α and LPS in vitro. We therefore propose a mechanism in which mesenteric adipocyte, through their production of key chemokines in response to inflammatory/bacterial stimuli, may orchestrate the formation of functional TLOs developing in CD-affected mesentery.
Collapse
Affiliation(s)
- Kevin Guedj
- Université de Paris, UMRS1148, INSERM, DHU Fire, Hôpital Bichat, 46 rue Henri Huchard, 75018, Paris, France; APHP, Department of Gastroenterology, IBD and Intestinal Failure, Hôpital Beaujon, 100 Boulevard du Général Leclerc, 92110, Clichy, France
| | - Yaël Abitbol
- Université de Paris, UMRS1148, INSERM, DHU Fire, Hôpital Bichat, 46 rue Henri Huchard, 75018, Paris, France; APHP, Department of Gastroenterology, IBD and Intestinal Failure, Hôpital Beaujon, 100 Boulevard du Général Leclerc, 92110, Clichy, France
| | - Dominique Cazals-Hatem
- APHP, Department of Pathology, Hôpital Beaujon, 100 Boulevard du Général Leclerc, 92110, Clichy, France
| | - Marion Morvan
- Université de Paris, UMRS1148, INSERM, DHU Fire, Hôpital Bichat, 46 rue Henri Huchard, 75018, Paris, France
| | - Léon Maggiori
- APHP, Department of Colorectal Surgery, Hôpital Beaujon, 100 Boulevard du Général Leclerc, 92110, Clichy, France; DHU Unity - Paris 7 Diderot University, 5 rue Thomas Mann, 75013, Paris, France
| | - Yves Panis
- APHP, Department of Colorectal Surgery, Hôpital Beaujon, 100 Boulevard du Général Leclerc, 92110, Clichy, France; DHU Unity - Paris 7 Diderot University, 5 rue Thomas Mann, 75013, Paris, France
| | - Yoram Bouhnik
- DHU Unity - Paris 7 Diderot University, 5 rue Thomas Mann, 75013, Paris, France; APHP, Department of Gastroenterology, IBD and Intestinal Failure, Hôpital Beaujon, 100 Boulevard du Général Leclerc, 92110, Clichy, France
| | - Giuseppina Caligiuri
- Université de Paris, UMRS1148, INSERM, DHU Fire, Hôpital Bichat, 46 rue Henri Huchard, 75018, Paris, France
| | - Olivier Corcos
- Université de Paris, UMRS1148, INSERM, DHU Fire, Hôpital Bichat, 46 rue Henri Huchard, 75018, Paris, France; DHU Unity - Paris 7 Diderot University, 5 rue Thomas Mann, 75013, Paris, France; APHP, Department of Gastroenterology, IBD and Intestinal Failure, Hôpital Beaujon, 100 Boulevard du Général Leclerc, 92110, Clichy, France
| | - Antonino Nicoletti
- Université de Paris, UMRS1148, INSERM, DHU Fire, Hôpital Bichat, 46 rue Henri Huchard, 75018, Paris, France.
| |
Collapse
|
113
|
Hu D, Yin C, Luo S, Habenicht AJR, Mohanta SK. Vascular Smooth Muscle Cells Contribute to Atherosclerosis Immunity. Front Immunol 2019; 10:1101. [PMID: 31164888 PMCID: PMC6534067 DOI: 10.3389/fimmu.2019.01101] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/30/2019] [Indexed: 11/13/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) constitute the major cells in the media layer of arteries, and are critical to maintain the integrity of the arterial wall. They participate in arterial wall remodeling, and play important roles in atherosclerosis throughout all stages of the disease. Studies demonstrate that VSMCs can adopt numerous phenotypes depending on inputs from endothelial cells (ECs) of the intima, resident cells of the adventitia, circulating immune cells, hormones, and plasma lipoproteins. This plasticity allows them to perform multiple tasks in physiology and disease. In this minireview, we focus on a previously underappreciated activity of VSMCs, i.e., their impact on atherosclerosis immunity via formation of artery tertiary lymphoid organs (ATLOs).
Collapse
Affiliation(s)
- Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changjun Yin
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Shanshan Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Andreas J R Habenicht
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sarajo K Mohanta
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
114
|
Williams JW, Huang LH, Randolph GJ. Cytokine Circuits in Cardiovascular Disease. Immunity 2019; 50:941-954. [PMID: 30995508 PMCID: PMC6924925 DOI: 10.1016/j.immuni.2019.03.007] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/08/2019] [Accepted: 03/12/2019] [Indexed: 02/06/2023]
Abstract
Arterial inflammation is a hallmark of atherosclerosis, and appropriate management of this inflammation represents a major unmet therapeutic need for cardiovascular disease patients. Here, we review the diverse contributions of immune cells to atherosclerosis, the mechanisms of immune cell activation in this context, and the cytokine circuits that underlie disease progression. We discuss the recent application of these insights in the form of immunotherapy to treat cardiovascular disease and highlight how studies on the cardiovascular co-morbidity that arises in autoimmunity might reveal additional roles for cytokines in atherosclerosis. Currently, data point to interleukin-1β (IL-1β), tumor necrosis factor (TNF), and IL-17 as cytokines that, at least in some settings, are effective targets to reduce cardiovascular disease progression.
Collapse
Affiliation(s)
- Jesse W Williams
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63139, USA
| | - Li-Hao Huang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63139, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63139, USA.
| |
Collapse
|
115
|
Abstract
There is now overwhelming experimental and clinical evidence that arteriosclerosis is a chronic inflammatory disease. Lessons learned from genome-wide association studies, advanced in vivo imaging techniques, transgenic lineage tracing mice models and clinical interventional studies have shown that both innate and adaptive immune mechanisms can accelerate or curb arteriosclerosis. This article summarizes and discusses the pathogenesis of arteriosclerosis with a focus on the role of the adaptive immune system. Some limitations of animal models are discussed and the need for models that are tailored to better translate to human atherosclerosis and ultimately progress in prevention and treatment are emphasized.
Collapse
Affiliation(s)
- D Wolf
- Abteilung für Kardiologie und Angiologie I, Universitäts-Herzzentrum Freiburg, Freiburg, Deutschland
- Medizinische Fakultät, Universität Freiburg, Freiburg, Deutschland
| | - K Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Cir, 92037, La Jolla, CA, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
116
|
Workel HH, Lubbers JM, Arnold R, Prins TM, van der Vlies P, de Lange K, Bosse T, van Gool IC, Eggink FA, Wouters MCA, Komdeur FL, van der Slikke EC, Creutzberg CL, Kol A, Plat A, Glaire M, Church DN, Nijman HW, de Bruyn M. A Transcriptionally Distinct CXCL13 +CD103 +CD8 + T-cell Population Is Associated with B-cell Recruitment and Neoantigen Load in Human Cancer. Cancer Immunol Res 2019; 7:784-796. [PMID: 30872264 DOI: 10.1158/2326-6066.cir-18-0517] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/03/2018] [Accepted: 03/06/2019] [Indexed: 11/16/2022]
Abstract
The chemokine CXCL13 mediates recruitment of B cells to tumors and is essential for the formation of tertiary lymphoid structures (TLSs). TLSs are thought to support antitumor immunity and are associated with improved prognosis. However, it remains unknown whether TLSs are formed in response to the general inflammatory character of the tumor microenvironment, or rather, are induced by (neo)antigen-specific adaptive immunity. We here report on the finding that the TGFβ-dependent CD103+CD8+ tumor-infiltrating T-cell (TIL) subpopulation expressed and produced CXCL13. Accordingly, CD8+ T cells from peripheral blood activated in the presence of TGFβ upregulated CD103 and secreted CXCL13. Conversely, inhibition of TGFβ receptor signaling abrogated CXCL13 production. CXCL13+CD103+CD8+ TILs correlated with B-cell recruitment, TLSs, and neoantigen burden in six cohorts of human tumors. Altogether, our findings indicated that TGFβ plays a noncanonical role in coordinating immune responses against human tumors and suggest a potential role for CXCL13+CD103+CD8+ TILs in mediating B-cell recruitment and TLS formation in human tumors.
Collapse
Affiliation(s)
- Hagma H Workel
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Joyce M Lubbers
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Roland Arnold
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Thalina M Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Pieter van der Vlies
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kim de Lange
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Inge C van Gool
- Department of Pathology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Florine A Eggink
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Maartje C A Wouters
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | - Fenne L Komdeur
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Elisabeth C van der Slikke
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Carien L Creutzberg
- Department of Radiation Oncology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Arjan Kol
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Annechien Plat
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mark Glaire
- Molecular and Population Genetics Laboratory, The Wellcome Trust Centre for Human Genetics and Oxford Cancer Centre, University of Oxford, Oxford, United Kingdom
| | - David N Church
- Molecular and Population Genetics Laboratory, The Wellcome Trust Centre for Human Genetics and Oxford Cancer Centre, University of Oxford, Oxford, United Kingdom.,NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust and John Radcliffe Hospital, Oxford, United Kingdom
| | - Hans W Nijman
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marco de Bruyn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
117
|
Nandkeolyar S, Naqvi A, Fan W, Sharma A, Rana JS, Rozanski A, Shaw L, Friedman JD, Hayes S, Dey D, Wong ND, Berman DS. Utility of novel serum biomarkers to predict subclinical atherosclerosis: A sub-analysis of the EISNER study. Atherosclerosis 2019; 282:80-84. [DOI: 10.1016/j.atherosclerosis.2019.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/28/2018] [Accepted: 01/10/2019] [Indexed: 11/27/2022]
|
118
|
Zhang X, Liu F, Bai P, Dong N, Chu C. Identification of key genes and pathways contributing to artery tertiary lymphoid organ development in advanced mouse atherosclerosis. Mol Med Rep 2019; 19:3071-3086. [PMID: 30816519 PMCID: PMC6423582 DOI: 10.3892/mmr.2019.9961] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/12/2019] [Indexed: 01/13/2023] Open
Abstract
Atherosclerosis is a leading cause of mortality worldwide. Artery tertiary lymphoid organ (ATLO) neogenesis is affected by abdominal aorta atherosclerosis, which may lead to an immune response. The present study obtained microarray data to investigate the gene expression differences underlying the potential pathogenesis of atherosclerosis and to elucidate the mechanisms underlying ATLO development. Microarray studies of the aorta, plaques, adventitia, blood, spleen, renal lymph nodes and ATLO were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified in aorta clusters and ATLO clusters. Kyoto Encyclopedia of Genes and Genomes enrichment and Gene Ontology (GO) analyses were conducted to predict the biological functions of DEGs. The results demonstrated that interleukin 7 receptor (Il7r), C‑X‑C motif chemokine ligand (Cxcl)16, Cxcl13, Cxcl12, C‑C motif chemokine receptor 2, C‑C motif chemokine ligand (Ccl)8, Ccl5 and Ccl12 may function through pathways associated with 'cytokine‑cytokine receptor interaction' and 'chemokine signaling pathway' in ATLO. Gene expression alterations were validated by reverse transcription‑quantitative polymerase chain reaction. Il7r appeared to be the central gene involved in these events, and chemokines and/or chemokine receptors were visualized by GO enrichment. A protein‑protein interaction network was constructed, which suggested that Il7r had a core function in all clusters. Taken together, the results indicated that Il7r upregulation may serve an important role in ATLO development via 'cytokine‑cytokine receptor interaction' and 'chemokine signaling pathway'. This may provide novel perspectives for understanding ATLO development and the regulation of the immune response in atherosclerosis.
Collapse
Affiliation(s)
- Xi Zhang
- Institute for Cardiovascular Prevention, Ludwig‑Maximilians University Munich, D‑80336 Munich, Germany
| | - Fayuan Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Peng Bai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Chong Chu
- Institute for Cardiovascular Prevention, Ludwig‑Maximilians University Munich, D‑80336 Munich, Germany
| |
Collapse
|
119
|
Yin C, Ackermann S, Ma Z, Mohanta SK, Zhang C, Li Y, Nietzsche S, Westermann M, Peng L, Hu D, Bontha SV, Srikakulapu P, Beer M, Megens RTA, Steffens S, Hildner M, Halder LD, Eckstein HH, Pelisek J, Herms J, Roeber S, Arzberger T, Borodovsky A, Habenicht L, Binder CJ, Weber C, Zipfel PF, Skerka C, Habenicht AJR. ApoE attenuates unresolvable inflammation by complex formation with activated C1q. Nat Med 2019; 25:496-506. [PMID: 30692699 PMCID: PMC6420126 DOI: 10.1038/s41591-018-0336-8] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/13/2018] [Indexed: 01/17/2023]
Abstract
ApoE has been implicated in Alzheimer´s disease, atherosclerosis,
and other unresolvable inflammatory conditions but a common mechanism of action
remains elusive. We found in ApoE-deficient mice that oxidized lipids activated
the classical complement cascade (CCC) resulting in leukocyte infiltration of
the choroid plexus (ChP). All human ApoE isoforms attenuated CCC activity via
high-affinity binding to the activated CCC-initiating C1q protein
(KD~140-580 pM) in vitro; and C1q-ApoE
complexes emerged as markers for ongoing complement activity of diseased ChPs,
Aβ plaques, and atherosclerosis in vivo. C1q-ApoE
complexes in human ChPs, Aβ plaques, and arteries correlated with
cognitive decline and atherosclerosis, respectively. Treatment with siRNA
against C5 which is formed by all complement pathways, attenuated murine ChP
inflammation, Aβ-associated microglia accumulation, and atherosclerosis.
Thus, ApoE is a direct checkpoint inhibitor of unresolvable inflammation and
reducing C5 attenuates disease burden.
Collapse
Affiliation(s)
- Changjun Yin
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany. .,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.
| | - Susanne Ackermann
- Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Zhe Ma
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Sarajo K Mohanta
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Chuankai Zhang
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Yuanfang Li
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| | - Sandor Nietzsche
- Centre for Electron Microscopy, Jena University Hospital, Friedrich-Schiller-University of Jena, Jena, Germany
| | - Martin Westermann
- Centre for Electron Microscopy, Jena University Hospital, Friedrich-Schiller-University of Jena, Jena, Germany
| | - Li Peng
- Department of Cardiovascular Medicine of Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | | | - Prasad Srikakulapu
- Cardiovascular Research Center (CVRC), University of Virginia, Charlottesville, VA, USA
| | - Michael Beer
- Department of Information Technology, University Clinic Jena, Jena, Germany
| | - Remco T A Megens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Markus Hildner
- Institute for Anatomy II, University Clinic Jena, Jena, Germany
| | - Luke D Halder
- Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
| | - Hans-Henning Eckstein
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jaroslav Pelisek
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Ludwig-Maximilians-University, Munich, Germany
| | - Sigrun Roeber
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany
| | - Thomas Arzberger
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University, Munich, Germany.,Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, Munich, Germany
| | | | - Livia Habenicht
- II. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Peter F Zipfel
- Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany.,Friedrich-Schiller-University, Faculty of Biological Sciences, Jena, Germany
| | - Christine Skerka
- Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany.
| | - Andreas J R Habenicht
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
120
|
Abstract
PURPOSE OF REVIEW The immune system plays a critical role in the development and modulation of atherosclerosis. New high-parameter technologies, including mass cytometry (CyTOF) and single-cell RNA sequencing (scRNAseq), allow for an encompassing analysis of immune cells. Unexplored marker combinations and transcriptomes can define new immune cell subsets and suggest their functions. Here, we review recent advances describing the immune cells in the artery wall of mice with and without atherosclerosis. We compare technologies and discuss limitations and advantages. RECENT FINDINGS Both CyTOF and scRNAseq on leukocytes from digested aortae show 10-30 immune cell subsets. Myeloid, T, B and natural killer cells were confirmed. Although cellular functions can be inferred from RNA-Seq data, some subsets cannot be identified based on current knowledge, suggesting they may be new cell types. CyTOF and scRNAseq each identified four B-cell subsets and three macrophage subsets in the atherosclerotic aorta. Limitations include cell death caused by enzymatic digestion and the limited depth of the scRNAseq transcriptomes. SUMMARY High-parameter methods are powerful tools for uncovering leukocyte diversity. CyTOF is currently more powerful at discerning leukocyte subsets in the atherosclerotic aorta, whereas scRNAseq provides more insight into their likely functions.
Collapse
Affiliation(s)
- Holger Winkels
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Erik Ehinger
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Yanal Ghosheh
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Dennis Wolf
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
- Department of Cardiology and Angiology I, University Heart Center Freiburg
- Faculty of Medicine, University of Freiburg, Freiburg, Germany and
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
121
|
Pipi E, Nayar S, Gardner DH, Colafrancesco S, Smith C, Barone F. Tertiary Lymphoid Structures: Autoimmunity Goes Local. Front Immunol 2018; 9:1952. [PMID: 30258435 PMCID: PMC6143705 DOI: 10.3389/fimmu.2018.01952] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 08/07/2018] [Indexed: 12/18/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are frequently observed in target organs of autoimmune diseases. TLS present features of secondary lymphoid organs such as segregated T and B cell zones, presence of follicular dendritic cell networks, high endothelial venules and specialized lymphoid fibroblasts and display the mechanisms to support local adaptive immune responses toward locally displayed antigens. TLS detection in the tissue is often associated with poor prognosis of disease, auto-antibody production and malignancy development. This review focuses on the contribution of TLS toward the persistence of the inflammatory drive, the survival of autoreactive lymphocyte clones and post-translational modifications, responsible for the pathogenicity of locally formed autoantibodies, during autoimmune disease development.
Collapse
Affiliation(s)
- Elena Pipi
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,Experimental Medicine Unit, Immuno-Inflammation Therapeutic Area, GSK Medicines Research Centre, Stevenage, United Kingdom
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - David H Gardner
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | | | - Charlotte Smith
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Francesca Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
122
|
Williams JW, Martel C, Potteaux S, Esaulova E, Ingersoll MA, Elvington A, Saunders BT, Huang LH, Habenicht AJ, Zinselmeyer BH, Randolph GJ. Limited Macrophage Positional Dynamics in Progressing or Regressing Murine Atherosclerotic Plaques-Brief Report. Arterioscler Thromb Vasc Biol 2018; 38:1702-1710. [PMID: 29903736 PMCID: PMC6202234 DOI: 10.1161/atvbaha.118.311319] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 05/22/2018] [Indexed: 01/19/2023]
Abstract
Objective- Macrophages play important roles in the pathogenesis of atherosclerosis, but their dynamics within plaques remain obscure. We aimed to quantify macrophage positional dynamics within progressing and regressing atherosclerotic plaques. Approach and Results- In a stable intravital preparation, large asymmetrical foamy macrophages in the intima of carotid artery plaques were sessile, but smaller rounded cells nearer plaque margins, possibly newly recruited monocytes, mobilized laterally along plaque borders. Thus, to test macrophage dynamics in plaques over a longer period of time in progressing and regressing disease, we quantified displacement of nondegradable phagocytic particles within macrophages for up to 6 weeks. In progressing plaques, macrophage-associated particles appeared to mobilize to deeper layers in plaque, whereas in regressing plaques, the label was persistently located near the lumen. By measuring the distance of the particles from the floor of the plaque, we discovered that particles remained at the same distance from the floor regardless of plaque progression or regression. The apparent deeper penetration of labeled cells in progressing conditions could be attributed to monocyte recruitment that generated new superficial layers of macrophages over the labeled phagocytes. Conclusions- Although there may be individual exceptions, as a population, newly differentiated macrophages fail to penetrate significantly deeper than the limited depth they reside on initial entry, regardless of plaque progression, or regression. These limited dynamics may prevent macrophages from escaping areas with unfavorable conditions (such as hypoxia) and pose a challenge for newly recruited macrophages to clear debris through efferocytosis deep within plaque.
Collapse
Affiliation(s)
- Jesse W. Williams
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO 63110 USA
| | - Catherine Martel
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO 63110 USA
| | - Stephane Potteaux
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, NY 10029 USA
| | - Ekaterina Esaulova
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO 63110 USA
| | - Molly A. Ingersoll
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, NY 10029 USA
| | - Andrew Elvington
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO 63110 USA
| | - Brian T. Saunders
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO 63110 USA
| | - Li-Hao Huang
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO 63110 USA
| | - Andreas J. Habenicht
- Institute of Vascular Prevention; Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Bernd H. Zinselmeyer
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO 63110 USA
| | - Gwendalyn J. Randolph
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO 63110 USA
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, NY 10029 USA
| |
Collapse
|
123
|
Getz GS, Reardon CA. T Cells in Atherosclerosis in Ldlr-/- and Apoe-/- Mice. JOURNAL OF IMMUNOLOGICAL SCIENCES 2018; 2:69-76. [PMID: 30854522 PMCID: PMC6404748 DOI: 10.29245/2578-3009/2018/3.1144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Atherosclerosis is the underlying basis for most cardiovascular diseases. It is a chronic inflammation affecting the arterial intima and is promoted by hypercholesterolemia. Cells of both the innate and adaptive immune systems contribute to this inflammation with macrophages and T cells being the most abundant immune cells in the atherosclerotic plaques. In this review, we discuss the studies that examined the role of T cells and T cell subsets in Apoe-/- and Ldlr-/- murine models of atherosclerosis. While there is a general consensus that Th1 cells are pro-atherogenic and regulatory T cells are atheroprotective, the role of other subsets is more ambiguous. In addition, the results in the two models of atherosclerosis do not always yield similar results. Additional studies in the two murine models using cell specific gene manipulations are needed.
Collapse
Affiliation(s)
- Godfrey S. Getz
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Catherine A. Reardon
- Ben May Institute for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
124
|
Trajkovski G, Ognjenovic L, Jota G, Hadzi-Manchev D, Trajkovska V, Volcevski G, Nikolova D, Petrushevska G, Janevska V, Janevski V. Tumour Lymphocytic Infiltration, Its Structure and Influence in Colorectal Cancer Progression. Open Access Maced J Med Sci 2018; 6:1003-1009. [PMID: 29983792 PMCID: PMC6026406 DOI: 10.3889/oamjms.2018.279] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND: The role of the immune system in the control of tumour progression has been stressed, recently. Many studies indicate the fact that the immune system can prevent tumour progression in several types of human malignant neoplasms including colorectal cancer. According to some authors, a higher density of “tumour-associated lymphocytes” (TAL), in malignant neoplasms, correlate with prolonged survival of patients. AIM: This study aims to determine the structure and the influence of the immune cells, TAL, in the progression of colorectal cancer (CRC). PATIENTS AND METHODS: The study included 103 patients with CRC operated at the University Clinic of Digestive Surgery in Skopje, whose operative material was analysed at the Institute of Pathology, Medical Faculty in Skopje. The structure of tumor-associated cells and their density were determined and were correlated with neoplasm’s grade, local growth (T), positive lymph nodes, lymphatic invasion and stage of the disease. RESULTS: CD4+, CD8+ and CD20+ lymphocytes (Ly) were found in TAL. The density of TAL was significantly different in neoplasms with different T status, lymphatic invasion, patients with and without nodal metastasis and patients with a different stage of the disease. The density of CD4+, CD8+, and CD20+ cells were significantly different in neoplasms with different T. The density of CD8+ and CD20+ lymphocytes was lower in patients with nodal metastasis and higher stage. CONCLUSION: The density of tumor-associated lymphocytes can anticipate the disease progression in patients with colorectal cancer, and the density of TAL influences the control of tumour progression.
Collapse
Affiliation(s)
- Gjorgji Trajkovski
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Ljubomir Ognjenovic
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Gjorgji Jota
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Dragan Hadzi-Manchev
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Vanja Trajkovska
- University Clinic of TOARILUC, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Goce Volcevski
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Dafina Nikolova
- University Clinic of Gastroenterohepatology, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Gordana Petrushevska
- Institute of Pathology, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Vesna Janevska
- Institute of Pathology, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - Vlado Janevski
- University Clinic of Abdominal Surgery, Faculty of Medicine, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| |
Collapse
|
125
|
Meier LA, Binstadt BA. The Contribution of Autoantibodies to Inflammatory Cardiovascular Pathology. Front Immunol 2018; 9:911. [PMID: 29755478 PMCID: PMC5934424 DOI: 10.3389/fimmu.2018.00911] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/12/2018] [Indexed: 12/19/2022] Open
Abstract
Chronic inflammation and resulting tissue damage underlie the vast majority of acquired cardiovascular disease (CVD), a general term encompassing a widely diverse array of conditions. Both innate and adaptive immune mechanisms contribute to chronic inflammation in CVD. Although maladies, such as atherosclerosis and cardiac fibrosis, are commonly conceptualized as disorders of inflammation, the cellular and molecular mechanisms that promote inflammation during the natural history of these diseases in human patients are not fully defined. Autoantibodies (AAbs) with specificity to self-derived epitopes accompany many forms of CVD in humans. Both adaptive/induced iAAbs (generated following cognate antigen encounter) and also autoantigen-reactive natural antibodies (produced independently of infection and in the absence of T cell help) have been demonstrated to modulate the natural history of multiple forms of CVD including atherosclerosis (atherosclerotic cardiovascular disease), dilated cardiomyopathy, and valvular heart disease. Despite the breadth of experimental evidence for the role of AAbs in CVD, there is a lack of consensus regarding their specific functions, primarily due to disparate conclusions reached, even when similar approaches and experimental models are used. In this review, we seek to summarize the current understanding of AAb function in CVD through critical assessment of the clinical and experimental evidence in this field. We additionally highlight the difficulty in translating observations made in animal models to human physiology and disease and provide a summary of unresolved questions that are critical to address in future studies.
Collapse
Affiliation(s)
- Lee A Meier
- Center for Immunology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Bryce A Binstadt
- Center for Immunology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
126
|
Sedding DG, Boyle EC, Demandt JAF, Sluimer JC, Dutzmann J, Haverich A, Bauersachs J. Vasa Vasorum Angiogenesis: Key Player in the Initiation and Progression of Atherosclerosis and Potential Target for the Treatment of Cardiovascular Disease. Front Immunol 2018; 9:706. [PMID: 29719532 PMCID: PMC5913371 DOI: 10.3389/fimmu.2018.00706] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/22/2018] [Indexed: 01/08/2023] Open
Abstract
Plaque microvascularization and increased endothelial permeability are key players in the development of atherosclerosis, from the initial stages of plaque formation to the occurrence of acute cardiovascular events. First, endothelial dysfunction and increased permeability facilitate the entry of diverse inflammation-triggering molecules and particles such as low-density lipoproteins into the artery wall from the arterial lumen and vasa vasorum (VV). Recognition of entering particles by resident phagocytes in the vessel wall triggers a maladaptive inflammatory response that initiates the process of local plaque formation. The recruitment and accumulation of inflammatory cells and the subsequent release of several cytokines, especially from resident macrophages, stimulate the expansion of existing VV and the formation of new highly permeable microvessels. This, in turn, exacerbates the deposition of pro-inflammatory particles and results in the recruitment of even more inflammatory cells. The progressive accumulation of leukocytes in the intima, which trigger proliferation of smooth muscle cells in the media, results in vessel wall thickening and hypoxia, which further stimulates neoangiogenesis of VV. Ultimately, this highly inflammatory environment damages the fragile plaque microvasculature leading to intraplaque hemorrhage, plaque instability, and eventually, acute cardiovascular events. This review will focus on the pivotal roles of endothelial permeability, neoangiogenesis, and plaque microvascularization by VV during plaque initiation, progression, and rupture. Special emphasis will be given to the underlying molecular mechanisms and potential therapeutic strategies to selectively target these processes.
Collapse
Affiliation(s)
- Daniel G Sedding
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Erin C Boyle
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Jasper A F Demandt
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Judith C Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands.,BHF Centre for Cardiovascular Science, Edinburgh University, Edinburgh, United Kingdom
| | - Jochen Dutzmann
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Axel Haverich
- Department of Cardiothoracic, Transplantation, and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
127
|
Wen Z, Shen Y, Berry G, Shahram F, Li Y, Watanabe R, Liao YJ, Goronzy JJ, Weyand CM. The microvascular niche instructs T cells in large vessel vasculitis via the VEGF-Jagged1-Notch pathway. Sci Transl Med 2018; 9:9/399/eaal3322. [PMID: 28724574 DOI: 10.1126/scitranslmed.aal3322] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 03/07/2017] [Accepted: 05/31/2017] [Indexed: 12/11/2022]
Abstract
Microvascular networks in the adventitia of large arteries control access of inflammatory cells to the inner wall layers (media and intima) and thus protect the immune privilege of the aorta and its major branches. In autoimmune vasculitis giant cell arteritis (GCA), CD4 T helper 1 (TH1) and TH17 cells invade into the wall of the aorta and large elastic arteries to form tissue-destructive granulomas. Whether the disease microenvironment provides instructive cues for vasculitogenic T cells is unknown. We report that adventitial microvascular endothelial cells (mvECs) perform immunoregulatory functions by up-regulating the expression of the Notch ligand Jagged1. Vascular endothelial growth factor (VEGF), abundantly present in GCA patients' blood, induced Jagged1 expression, allowing mvECs to regulate effector T cell induction via the Notch-mTORC1 (mammalian target of rapamycin complex 1) pathway. We found that circulating CD4 T cells in GCA patients have left the quiescent state, actively signal through the Notch pathway, and differentiate into TH1 and TH17 effector cells. In an in vivo model of large vessel vasculitis, exogenous VEGF functioned as an effective amplifier to recruit and activate vasculitogenic T cells. Thus, systemic VEGF co-opts endothelial Jagged1 to trigger aberrant Notch signaling, biases responsiveness of CD4 T cells, and induces pathogenic effector functions. Adventitial microvascular networks function as an instructive tissue niche, which can be exploited to target vasculitogenic immunity in large vessel vasculitis.
Collapse
Affiliation(s)
- Zhenke Wen
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yi Shen
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gerald Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Farhad Shahram
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yinyin Li
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ryu Watanabe
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yaping Joyce Liao
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jörg J Goronzy
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
128
|
Winkels H, Ehinger E, Vassallo M, Buscher K, Dinh HQ, Kobiyama K, Hamers AAJ, Cochain C, Vafadarnejad E, Saliba AE, Zernecke A, Pramod AB, Ghosh AK, Anto Michel N, Hoppe N, Hilgendorf I, Zirlik A, Hedrick CC, Ley K, Wolf D. Atlas of the Immune Cell Repertoire in Mouse Atherosclerosis Defined by Single-Cell RNA-Sequencing and Mass Cytometry. Circ Res 2018; 122:1675-1688. [PMID: 29545366 DOI: 10.1161/circresaha.117.312513] [Citation(s) in RCA: 383] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/04/2018] [Accepted: 03/14/2018] [Indexed: 12/24/2022]
Abstract
RATIONALE Atherosclerosis is a chronic inflammatory disease that is driven by the interplay of pro- and anti-inflammatory leukocytes in the aorta. Yet, the phenotypic and transcriptional diversity of aortic leukocytes is poorly understood. OBJECTIVE We characterized leukocytes from healthy and atherosclerotic mouse aortas in-depth by single-cell RNA-sequencing and mass cytometry (cytometry by time of flight) to define an atlas of the immune cell landscape in atherosclerosis. METHODS AND RESULTS Using single-cell RNA-sequencing of aortic leukocytes from chow diet- and Western diet-fed Apoe-/- and Ldlr-/- mice, we detected 11 principal leukocyte clusters with distinct phenotypic and spatial characteristics while the cellular repertoire in healthy aortas was less diverse. Gene set enrichment analysis on the single-cell level established that multiple pathways, such as for lipid metabolism, proliferation, and cytokine secretion, were confined to particular leukocyte clusters. Leukocyte populations were differentially regulated in atherosclerotic Apoe-/- and Ldlr-/- mice. We confirmed the phenotypic diversity of these clusters with a novel mass cytometry 35-marker panel with metal-labeled antibodies and conventional flow cytometry. Cell populations retrieved by these protein-based approaches were highly correlated to transcriptionally defined clusters. In an integrated screening strategy of single-cell RNA-sequencing, mass cytometry, and fluorescence-activated cell sorting, we detected 3 principal B-cell subsets with alterations in surface markers, functional pathways, and in vitro cytokine secretion. Leukocyte cluster gene signatures revealed leukocyte frequencies in 126 human plaques by a genetic deconvolution strategy. This approach revealed that human carotid plaques and microdissected mouse plaques were mostly populated by macrophages, T-cells, and monocytes. In addition, the frequency of genetically defined leukocyte populations in carotid plaques predicted cardiovascular events in patients. CONCLUSIONS The definition of leukocyte diversity by high-dimensional analyses enables a fine-grained analysis of aortic leukocyte subsets, reveals new immunologic mechanisms and cell-type-specific pathways, and establishes a functional relevance for lesional leukocytes in human atherosclerosis.
Collapse
Affiliation(s)
- Holger Winkels
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Erik Ehinger
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Melanie Vassallo
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Konrad Buscher
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Huy Q Dinh
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Kouji Kobiyama
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Anouk A J Hamers
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Clément Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany (C.C., A.Z.)
| | - Ehsan Vafadarnejad
- Helmholtz Institute for RNA-based Infection Research, Würzburg, Germany (E.V., A.-E.S.)
| | | | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany (C.C., A.Z.)
| | - Akula Bala Pramod
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Amlan K Ghosh
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Nathaly Anto Michel
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.).,the Faculty of Medicine, University of Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.)
| | - Natalie Hoppe
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.).,the Faculty of Medicine, University of Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.)
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.).,the Faculty of Medicine, University of Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.)
| | - Andreas Zirlik
- Department of Cardiology and Angiology I, University Heart Center Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.).,the Faculty of Medicine, University of Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.)
| | - Catherine C Hedrick
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.)
| | - Klaus Ley
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.).,Department of Bioengineering, University of California, San Diego (K.L.)
| | - Dennis Wolf
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany (C.C., A.Z.) .,From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA (H.W., E.E., M.V., K.B., H.Q.D., K.K., A.A.J.H., A.B.P., A.K.G., C.C.H., K.L., D.W.).,Department of Cardiology and Angiology I, University Heart Center Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.).,the Faculty of Medicine, University of Freiburg, Germany (N.A.M., N.H., I.H., A.Z., D.W.)
| |
Collapse
|
129
|
Engelhard VH, Rodriguez AB, Mauldin IS, Woods AN, Peske JD, Slingluff CL. Immune Cell Infiltration and Tertiary Lymphoid Structures as Determinants of Antitumor Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:432-442. [PMID: 29311385 PMCID: PMC5777336 DOI: 10.4049/jimmunol.1701269] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/19/2017] [Indexed: 12/22/2022]
Abstract
Limited representation of intratumoral immune cells is a major barrier to tumor control. However, simply enhancing immune responses in tumor-draining lymph nodes or through adoptive transfer may not overcome the limited ability of tumor vasculature to support effector infiltration. An alternative is to promote a sustained immune response intratumorally. This idea has gained traction with the observation that many tumors are associated with tertiary lymphoid structures (TLS), which organizationally resemble lymph nodes. These peri- and intratumoral structures are usually, but not always, associated with positive prognoses in patients. Preclinical and clinical data support a role for TLS in modulating immunity in the tumor microenvironment. However, there appear to be varied functions of TLS, potentially based on their structure or location in relation to the tumor or the origin or location of the tumor itself. Understanding more about TLS development, composition, and function may offer new therapeutic opportunities to modulate antitumor immunity.
Collapse
Affiliation(s)
- Victor H Engelhard
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908;
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908; and
| | - Anthony B Rodriguez
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908; and
| | - Ileana S Mauldin
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Amber N Woods
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908; and
| | - J David Peske
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908; and
| | - Craig L Slingluff
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908
| |
Collapse
|
130
|
Di Carlo SE, Peduto L. The perivascular origin of pathological fibroblasts. J Clin Invest 2018; 128:54-63. [PMID: 29293094 DOI: 10.1172/jci93558] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The ability to repair tissues is essential for the survival of organisms. In chronic settings, the failure of the repair process to terminate results in overproduction of collagen, a pathology known as fibrosis, which compromises organ recovery and impairs function. The origin of the collagen-overproducing cell has been debated for years. Here we review recent insights gained from the use of lineage tracing approaches in several organs. The resulting evidence points toward specific subsets of tissue-resident mesenchymal cells, mainly localized in a perivascular position, as the major source for collagen-producing cells after injury. We discuss these findings in view of the functional heterogeneity of mesenchymal cells of the perivascular niche, which have essential vascular, immune, and regenerative functions that need to be preserved for efficient repair.
Collapse
|
131
|
Mueller CG, Nayar S, Campos J, Barone F. Molecular and Cellular Requirements for the Assembly of Tertiary Lymphoid Structures. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:55-72. [PMID: 30155622 DOI: 10.1007/978-3-319-78127-3_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
At sites of chronic inflammation, recruited immune cells form structures that resemble secondary lymphoid organs (SLOs). Those are characterized by segregated areas of prevalent T- or B-cell aggregation, differentiation of high endothelial venules (HEVs) and local activation of resident stromal cells. B-cell proliferation and affinity maturation towards locally displayed autoantigens have been demonstrated at those sites, known as tertiary lymphoid structures (TLSs). TLS formation has been associated with local disease persistence and progression as well as increased systemic manifestations. While bearing a similar histological structure to SLO, the signals that regulate TLS and SLO formation can diverge, and a series of pro-inflammatory cytokines has been ascribed as responsible for TLS formation at different anatomical sites. Here we review the structural elements as well as the signals responsible for TLS aggregation, aiming to provide an overview to this complex immunological phenomenon.
Collapse
Affiliation(s)
- C G Mueller
- CNRS UPR 3572, Laboratory of Immunopathology and Therapeutic Chemistry/Laboratory of Excellence MEDALIS, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France
| | - S Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham, UK
| | - J Campos
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham, UK
| | - F Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham, UK.
| |
Collapse
|
132
|
Mueller CG, Nayar S, Gardner D, Barone F. Cellular and Vascular Components of Tertiary Lymphoid Structures. Methods Mol Biol 2018; 1845:17-30. [PMID: 30141005 DOI: 10.1007/978-1-4939-8709-2_2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Inflammatory immune cells recruited at the site of chronic inflammation form structures that resemble secondary lymphoid organs (SLO). These are characterized by segregated areas of prevalent T- or B-cell aggregation, differentiation of high endothelial venules, and local activation of resident stromal cells, including lymphatic endothelial cells. B-cell proliferation and affinity maturation toward locally displayed autoantigens have been demonstrated at these sites, known as tertiary lymphoid structures (TLS). TLS formation during chronic inflammation has been associated with local disease persistence and progression, as well as increased systemic manifestations. While bearing a similar histological structure to SLO, the signals that regulate TLS and SLO formation can diverge and a series of pro-inflammatory cytokines have been ascribed as responsible for TLS formation at different anatomical sites. Moreover, for a long time the structural compartment that regulates TLS homeostasis, including survival and recirculation of leucocytes has been neglected. In this chapter, we summarize the novel data available on TLS formation, structural organization, and the functional and anatomical links connecting TLS and SLOs.
Collapse
Affiliation(s)
- Christopher George Mueller
- Laboratoire d'Immunologie, Immunopathologie et Chimie Thérapeutique, Institut de Biologie Moléculaire et Cellulaire (IBMC), CNRS UPR 3572, University of Strasbourg, Strasbourg, France
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - David Gardner
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Francesca Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK.
| |
Collapse
|
133
|
Abstract
Atherosclerosis is initiated by cholesterol entry into arteries that triggers chronic immune-inflammatory lesions in the vessels. Early lesions are clinically insignificant but advanced complex lesions and vulnerable rupture prone lesions impact on quality of life and can be life threatening. Rupture of vulnerable atherosclerotic lesions initiates thrombotic occlusion of vital arteries precipitating heart attacks and strokes that remain major killers globally despite therapeutic use of statins to lower blood cholesterol levels. Conventional B2 cells are proatherogenic whereas peritoneal Bla cells are atheroprotective. Depletion of B2 cells by administration of mAb to CD20 or to BAFF receptor or in BAFF receptor-deficient mice ameliorates atherosclerosis. B2 cells may promote atherosclerosis by production of IgG, secretion of proinflammatory cytokine TNFα and activation of CD4 T cells. Together these B2 cell mechanisms contribute to generation of rupture-prone vulnerable atherosclerotic plaques characterised by large necrotic cores. In contrast, peritoneal Bla cells protect against atherosclerosis by secretion of natural IgM that scavenges apoptotic cells and oxidised LDL and reduces necrotic cores in atherosclerotic lesions. These atheroprotective effects can be further increased by stimulating Bla cells by administration of apoptotic cells, liposomes of phosphatidylserine abundant on surfaces of apoptotic cell, by mAb to TIM1, a phosphatidylserine receptor expressed by B1a cells and by TLR4-MyD88 activation. Experimental studies of atherosclerosis in mouse models indicate that reductions in atherogenic B2 cells and/or activation of atheroprotective B1a cells protects against atherosclerosis development, findings which have potential for clinical translation to reduce risks of deaths from heart attacks and strokes.
Collapse
Affiliation(s)
- Tin Kyaw
- a Australia and Baker IDI Heart and Diabetes Institute , Victoria , Australia.,b Department of Medicine , Southern Clinical School, Monash University , Victoria , Australia , and
| | - Peter Tipping
- b Department of Medicine , Southern Clinical School, Monash University , Victoria , Australia , and
| | - Alex Bobik
- a Australia and Baker IDI Heart and Diabetes Institute , Victoria , Australia.,c Department of Immunology , Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University , Victoria , Australia
| | - Ban-Hock Toh
- b Department of Medicine , Southern Clinical School, Monash University , Victoria , Australia , and
| |
Collapse
|
134
|
Colbeck EJ, Ager A, Gallimore A, Jones GW. Tertiary Lymphoid Structures in Cancer: Drivers of Antitumor Immunity, Immunosuppression, or Bystander Sentinels in Disease? Front Immunol 2017; 8:1830. [PMID: 29312327 PMCID: PMC5742143 DOI: 10.3389/fimmu.2017.01830] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/04/2017] [Indexed: 12/16/2022] Open
Abstract
Secondary lymphoid organs are integral to initiation and execution of adaptive immune responses. These organs provide a setting for interactions between antigen-specific lymphocytes and antigen-presenting cells recruited from local infected or inflamed tissues. Secondary lymphoid organs develop as a part of a genetically preprogrammed process during embryogenesis. However, organogenesis of secondary lymphoid tissues can also be recapitulated in adulthood during de novo lymphoid neogenesis of tertiary lymphoid structures (TLSs). These ectopic lymphoid-like structures form in the inflamed tissues afflicted by various pathological conditions, including cancer, autoimmunity, infection, or allograft rejection. Studies are beginning to shed light on the function of such structures in different disease settings, raising important questions regarding their contribution to progression or resolution of disease. Data show an association between the tumor-associated TLSs and a favorable prognosis in various types of human cancer, attracting the speculation that TLSs support effective local antitumor immune responses. However, definitive evidence for the role for TLSs in fostering immune responses in vivo are lacking, with current data remaining largely correlative by nature. In fact, some more recent studies have even demonstrated an immunosuppressive, tumor-promoting role for cancer-associated TLSs. In this review, we will discuss what is known about the development of cancer-associated TLSs and the current understanding of their potential role in the antitumor immune response.
Collapse
Affiliation(s)
| | - Ann Ager
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Awen Gallimore
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Gareth Wyn Jones
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
135
|
Alsughayyir J, Pettigrew GJ, Motallebzadeh R. Spoiling for a Fight: B Lymphocytes As Initiator and Effector Populations within Tertiary Lymphoid Organs in Autoimmunity and Transplantation. Front Immunol 2017; 8:1639. [PMID: 29218052 PMCID: PMC5703719 DOI: 10.3389/fimmu.2017.01639] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 11/09/2017] [Indexed: 12/13/2022] Open
Abstract
Tertiary lymphoid organs (TLOs) develop at ectopic sites within chronically inflamed tissues, such as in autoimmunity and rejecting organ allografts. TLOs differ structurally from canonical secondary lymphoid organs (SLOs), in that they lack a mantle zone and are not encapsulated, suggesting that they may provide unique immune function. A notable feature of TLOs is the frequent presence of structures typical of germinal centers (GCs). However, little is known about the role of such GCs, and in particular, it is not clear if the B cell response within is autonomous, or whether it synergizes with concurrent responses in SLOs. This review will discuss ectopic lymphoneogenesis and the role of the B cell in TLO formation and subsequent effector output in the context of autoimmunity and transplantation, with particular focus on the contribution of ectopic GCs to affinity maturation in humoral immune responses and to the potential breakdown of self-tolerance and development of humoral autoimmunity.
Collapse
Affiliation(s)
- Jawaher Alsughayyir
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Gavin J Pettigrew
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Reza Motallebzadeh
- Division of Surgery and Interventional Science, University College London, London, United Kingdom.,Institute of Immunity and Transplantation, University College London, London, United Kingdom.,Department of Nephrology, Urology and Transplantation, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
136
|
Manthey H, Zernecke A. Dendritic cells in atherosclerosis: Functions in immune regulation and beyond. Thromb Haemost 2017; 106:772-8. [DOI: 10.1160/th11-05-0296] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 08/02/2011] [Indexed: 12/15/2022]
Abstract
SummaryChronic inflammation drives the development of atherosclerosis. Dendritic cells (DCs) are known as central mediators of adaptive immune responses and the development of immunological memory and tolerance. DCs are present in non-diseased arteries, and accumulate within atherosclerotic lesions where they can be localised in close vicinity to T cells. Recent work has revealed important functions of DCs in regulating immune mechanisms in atherogenesis, and vaccination strategies using DCs have been explored for treatment of disease. However, in line with a phenotypical and functional overlap with plaque macrophages vascular DCs were also identified to engulf lipids, thus contributing to lipid burden in the vessel wall and initiation of lesion growth. Furthermore, a function of DCs in regulating cholesterol homeostasis has been revealed. Finally, phenotypically distinct plasmacytoid dendritic cells (pDCs) have been identified within atherosclerotic lesions. This review will dissect the multifaceted contribution of DCs and pDCs to the initiation and progression of atherosclerosis and the experimental approaches utilising DCs in therapeutic vaccination strategies.
Collapse
|
137
|
Yang JG, Sun YF, He KF, Ren JG, Liu ZJ, Liu B, Zhang W, Zhao YF. Lymphotoxins Promote the Progression of Human Lymphatic Malformation by Enhancing Lymphatic Endothelial Cell Proliferation. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2602-2615. [DOI: 10.1016/j.ajpath.2017.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/24/2017] [Accepted: 07/26/2017] [Indexed: 12/31/2022]
|
138
|
Migrating into the Tumor: a Roadmap for T Cells. Trends Cancer 2017; 3:797-808. [DOI: 10.1016/j.trecan.2017.09.006] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/20/2017] [Accepted: 09/21/2017] [Indexed: 12/17/2022]
|
139
|
Wang Y, Feng X, Shen B, Ma J, Zhao W. Is Vascular Amyloidosis Intertwined with Arterial Aging, Hypertension and Atherosclerosis? Front Genet 2017; 8:126. [PMID: 29085385 PMCID: PMC5649204 DOI: 10.3389/fgene.2017.00126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/04/2017] [Indexed: 12/21/2022] Open
Abstract
Vascular amyloidosis (VA) is a component of aging, but both VA and aging move forward together. Although, not all age-related molecules are involved with VA, some molecules are involved in a crosstalk between both of them. However, the cellular mechanism by which, vascular cells are phenotypically shifted to arterial remodeling, is not only involved in aging but also linked to VA. Additionally, patients with hypertension and atherosclerosis are susceptible to VA, while amyloidosis alone may provide fertile soil for the initiation and progression of subsequent hypertension and atherosclerosis. It is known that hypertension, atherosclerosis and amyloidosis can be viewed as accelerated aging. This review summarizes the available experimental and clinical evidence to help the reader to understand the advance and underlying mechanisms for VA involvement in and interaction with aging. Taken together, it is clear that VA, hypertension and atherosclerosis are closely intertwined with arterial aging as equal partners.
Collapse
Affiliation(s)
- Yushi Wang
- Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaoxing Feng
- Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | - Botao Shen
- Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | - Jing Ma
- Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| | - Waiou Zhao
- Cardiovascular Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
140
|
Xu X, Wang B, Ren C, Hu J, Greenberg DA, Chen T, Xie L, Jin K. Age-related Impairment of Vascular Structure and Functions. Aging Dis 2017; 8:590-610. [PMID: 28966804 PMCID: PMC5614324 DOI: 10.14336/ad.2017.0430] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 04/30/2017] [Indexed: 12/12/2022] Open
Abstract
Among age-related diseases, cardiovascular and cerebrovascular diseases are major causes of death. Vascular dysfunction is a key characteristic of these diseases wherein age is an independent and essential risk factor. The present work will review morphological alterations of aging vessels in-depth, which includes the discussion of age-related microvessel loss and changes to vasculature involving the capillary basement membrane, intima, media, and adventitia as well as the accompanying vascular dysfunctions arising from these alterations.
Collapse
Affiliation(s)
- Xianglai Xu
- 1Zhongshan Hospital, Fudan University, Shanghai 200032, China.,2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Brian Wang
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | - Changhong Ren
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA.,4Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University. Beijing, China
| | - Jiangnan Hu
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| | | | - Tianxiang Chen
- 6Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Liping Xie
- 3Department of Urology, the First Affiliated Hospital, Zhejiang University, Zhejiang Province, China
| | - Kunlin Jin
- 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, TX 76107, USA
| |
Collapse
|
141
|
Tang H, Zhu M, Qiao J, Fu YX. Lymphotoxin signalling in tertiary lymphoid structures and immunotherapy. Cell Mol Immunol 2017; 14:809-818. [PMID: 28413217 PMCID: PMC5649108 DOI: 10.1038/cmi.2017.13] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/24/2017] [Accepted: 01/24/2017] [Indexed: 12/16/2022] Open
Abstract
Tertiary lymphoid structures (TLS) often develop at sites of persistent inflammation, including cancers and autoimmune diseases. In most cases, the presence of TLS correlates with active immune responses. Because of their proximity to pathological loci, TLS are an intriguing target for the manipulation of immune responses. For several years, it has become clear that lymphotoxin (LT) signalling plays critical roles in lymphoid tissue organogenesis and maintenance. In the current review, we will discuss the role of LT signalling in the development of TLS. With a focus on cancers and autoimmune diseases, we will highlight the correlations between TLS and disease progression. We will also discuss the current efforts and potential directions for manipulating TLS for immunotherapies.
Collapse
Affiliation(s)
- Haidong Tang
- Department of Pathology, University of Texas, Southwestern Medical Center, Dallas, TX 75235, USA
| | - Mingzhao Zhu
- IBP-UTSW Joint Immunotherapy Group, Chinese Academy of Science, Key Laboratory for Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jian Qiao
- Department of Pathology, University of Texas, Southwestern Medical Center, Dallas, TX 75235, USA
| | - Yang-Xin Fu
- Department of Pathology, University of Texas, Southwestern Medical Center, Dallas, TX 75235, USA
- IBP-UTSW Joint Immunotherapy Group, Chinese Academy of Science, Key Laboratory for Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
142
|
Sakamoto A, Tanaka T, Hirano K, Koike K, Komuro I. Immunoglobulin G4-related Coronary Periarteritis and Luminal Stenosis in a Patient with a History of Autoimmune Pancreatitis. Intern Med 2017; 56:2445-2450. [PMID: 28824056 PMCID: PMC5643172 DOI: 10.2169/internalmedicine.8259-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Immunoglobulin G4 (IgG4)-related disease is a systemic inflammatory disorder that was first described in patients with autoimmune pancreatitis. Although IgG4-related disease is thought to involve the cardiovascular system, case reports describing coronary artery involvement are relatively rare. We describe a patient who was previously diagnosed with autoimmune pancreatitis and found to have coronary periarteritis and luminal narrowing. After the initiation of steroid treatment, the patient's coronary periarteritis and luminal stenosis were both ameliorated with an improvement in the serum IgG4 concentration. The present findings collectively suggest that IgG4-related immuno-inflammation may have a role in the development of coronary periarteritis and luminal atherosclerosis.
Collapse
Affiliation(s)
- Aiko Sakamoto
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, Japan
- Division for Health Service Promotion, University of Tokyo, Japan
| | - Tomofumi Tanaka
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, Japan
- Department of Cardiology, Sakakibara Heart Institute, Japan
| | - Kenji Hirano
- Department of Gastroenterology, University of Tokyo Graduate School of Medicine, Japan
- Department of Gastroenterology, Tokyo Takanawa Hospital, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, University of Tokyo Graduate School of Medicine, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, Japan
| |
Collapse
|
143
|
Akhavanpoor M, Gleissner CA, Akhavanpoor H, Lasitschka F, Doesch AO, Katus HA, Erbel C. Adventitial tertiary lymphoid organ classification in human atherosclerosis. Cardiovasc Pathol 2017; 32:8-14. [PMID: 29078120 DOI: 10.1016/j.carpath.2017.08.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/25/2017] [Accepted: 08/26/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease of the arterial wall. Adjacent to lamina intima lesion progression, a cellular compound develops in the lamina adventitia, defined as tertiary lymphoid organs (TLO) in mice. But in human system, it remains unknown whether these adventitial cellular accumulations represent these highly organized immunological structures. PATIENTS AND METHODS In this study, we investigated whether the adventitial cellular compounds represent TLOs in 72 human coronary artery samples by immunoenzyme staining. RESULTS The study showed that the adventitial cellular compound partly represented TLOs in human coronary arteries affected by atherogenesis in patients suffering from ischemic heart disease (56%) or a fatal myocardial infarction (100%), but not dilated cardiomyopathy. In addition, we established a classification for human TLOs, stage I-III, and showed that all stages were present in diseased coronary arteries. The stage of TLOs highly correlated with lesion size as well as plaque instability and rupture, and all patients with a myocardial infarction had stage III. Additionally, there were cellular infiltration and destruction of the lamina media, which were restricted to TLOs next to ruptured plaques in patients with a fatal myocardial infarction. CONCLUSIONS TLOs are present in patients with a coronary artery disease and highly correlated with lesion size, plaque instability, and rupture. Further studies are needed to investigate whether TLOs might be a specific diagnostic and drug target to modify plaque instability/rupture.
Collapse
Affiliation(s)
- Mohammadreza Akhavanpoor
- Department of Cardiology, University of Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg, Mannheim, Germany
| | - Christian A Gleissner
- Department of Cardiology, University of Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg, Mannheim, Germany
| | - Hamidreza Akhavanpoor
- Department of Cardiology, University of Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg, Mannheim, Germany
| | | | - Andreas O Doesch
- Department of Cardiology, University of Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg, Mannheim, Germany
| | - Hugo A Katus
- Department of Cardiology, University of Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg, Mannheim, Germany
| | - Christian Erbel
- Department of Cardiology, University of Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg, Mannheim, Germany.
| |
Collapse
|
144
|
Tammaro A, Derive M, Gibot S, Leemans JC, Florquin S, Dessing MC. TREM-1 and its potential ligands in non-infectious diseases: from biology to clinical perspectives. Pharmacol Ther 2017; 177:81-95. [PMID: 28245991 DOI: 10.1016/j.pharmthera.2017.02.043] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Triggering receptor expressed on myeloid cells-1 (TREM-1) is expressed on the majority of innate immune cells and to a lesser extent on parenchymal cells. Upon activation, TREM-1 can directly amplify an inflammatory response. Although it was initially demonstrated that TREM-1 was predominantly associated with infectious diseases, recent evidences shed new light into its role in sterile inflammatory diseases. Indeed, TREM-1 receptor and its signaling pathways contribute to the pathology of several non-infectious acute and chronic inflammatory diseases, including atherosclerosis, ischemia reperfusion-induced tissue injury, colitis, fibrosis and cancer. This review, aims to give an extensive overview of TREM-1 in non-infectious diseases, with the focus on the therapeutic potential of TREM-1 intervention strategies herein. In addition, we provide the reader with a functional enrichment analysis of TREM-1 signaling pathway and potential TREM-1 ligands in these diseases, obtained via in silico approach. We discuss pre-clinical studies which show that TREM-1 inhibition, via synthetic soluble TREM-1 protein mimickers, is effective in treating (preventing) specific inflammatory disorders, without significant effects on antibacterial response. Further research aimed at identifying specific TREM-1 ligands, in different inflammatory disorders, is required to further unravel the role of this receptor, and explore new avenues to modulate its function.
Collapse
Affiliation(s)
- Alessandra Tammaro
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | - Sebastien Gibot
- Medical Intensive Care Unit, Hôpital Central, CHU Nancy, Nancy, France; Inserm UMR_S1116, Faculté de Médecine, Université de Lorraine, Nancy, France
| | - Jaklien C Leemans
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sandrine Florquin
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pathology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Mark C Dessing
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
145
|
Newland SA, Mohanta S, Clément M, Taleb S, Walker JA, Nus M, Sage AP, Yin C, Hu D, Kitt LL, Finigan AJ, Rodewald HR, Binder CJ, McKenzie ANJ, Habenicht AJ, Mallat Z. Type-2 innate lymphoid cells control the development of atherosclerosis in mice. Nat Commun 2017; 8:15781. [PMID: 28589929 PMCID: PMC5467269 DOI: 10.1038/ncomms15781] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 04/27/2017] [Indexed: 01/06/2023] Open
Abstract
Type-2 innate lymphoid cells (ILC2) are a prominent source of type II cytokines and are found constitutively at mucosal surfaces and in visceral adipose tissue. Despite their role in limiting obesity, how ILC2s respond to high fat feeding is poorly understood, and their direct influence on the development of atherosclerosis has not been explored. Here, we show that ILC2 are present in para-aortic adipose tissue and lymph nodes and display an inflammatory-like phenotype atypical of adipose resident ILC2. High fat feeding alters both the number of ILC2 and their type II cytokine production. Selective genetic ablation of ILC2 in Ldlr−/− mice accelerates the development of atherosclerosis, which is prevented by reconstitution with wild type but not Il5−/− or Il13−/− ILC2. We conclude that ILC2 represent a major innate cell source of IL-5 and IL-13 required for mounting atheroprotective immunity, which can be altered by high fat diet. Type-2 innate lymphoid cells (ILC2) affect adipose tissue metabolism and function. Here the authors show that the ILC2 are present in para-aortic adipose tissue and represent a major source of IL-5 and IL-13 required for mounting atheroprotective immunity, which can be altered by high fat diet.
Collapse
Affiliation(s)
- Stephen A Newland
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Sarajo Mohanta
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Marc Clément
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Soraya Taleb
- Institut National de la Santé et de la Recherche Médicale, U970 Paris, France
| | - Jennifer A Walker
- Division of Protein and Nucleic Acid Chemistry, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Meritxell Nus
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Andrew P Sage
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Changjun Yin
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Desheng Hu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, Fujian 361102, China
| | - Lauren L Kitt
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Alison J Finigan
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Andrew N J McKenzie
- Division of Protein and Nucleic Acid Chemistry, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Andreas J Habenicht
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Ziad Mallat
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK.,Institut National de la Santé et de la Recherche Médicale, U970 Paris, France
| |
Collapse
|
146
|
Kohlgrüber S, Upadhye A, Dyballa-Rukes N, McNamara CA, Altschmied J. Regulation of Transcription Factors by Reactive Oxygen Species and Nitric Oxide in Vascular Physiology and Pathology. Antioxid Redox Signal 2017; 26:679-699. [PMID: 27841660 PMCID: PMC5421514 DOI: 10.1089/ars.2016.6946] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SIGNIFICANCE Cardiovascular diseases are the main cause of death worldwide and pose an immense economical burden. In most cases, the underlying problem is vascular occlusion by atherosclerotic plaques. Importantly, different cell types of the vascular wall and the immune system play crucial roles in atherosclerosis at different stages of the disease. Furthermore, atherosclerosis and conditions recognized as risk factors are characterized by a reduced availability of the vasoprotective molecule nitric oxide and an increase in reactive oxygen species, so-called oxidative stress. Transcription factors function as intracellular signal integrators and relays and thus, play a central role in cellular responses to changing conditions. Recent Advances: Work on specific transcriptional regulators has uncovered many of their functions and the upstream pathways modulating their activity in response to reactive oxygen and nitrogen species. Here, we have reviewed for a few selected examples how this can contribute not only to protection against atherosclerosis development but also to disease progression and the occurrence of clinical manifestations, such as plaque rupture. CRITICAL ISSUES Transcription factors have pleiotropic outputs and often also divergent functions in different cell types and tissues. Thus, in light of potential severe adverse side effects, a global activation or inhibition of particular transcriptions factors does not seem a feasible therapeutic option. FUTURE DIRECTIONS A further in-depth characterization of the cell- and stage-specific actions and regulation of transcription factors in atherosclerosis with respect to protein-protein interactions and target genes could open up new avenues for prevention or therapeutic interventions in this vascular disease. Antioxid. Redox Signal. 26, 679-699.
Collapse
Affiliation(s)
- Stefanie Kohlgrüber
- 1 IUF-Leibniz Research Institute for Environmental Medicine , Düsseldorf, Germany
| | - Aditi Upadhye
- 2 Department of Microbiology, Immunology, Cancer Biology, University of Virginia , Charlottesville, Virginia
| | - Nadine Dyballa-Rukes
- 1 IUF-Leibniz Research Institute for Environmental Medicine , Düsseldorf, Germany
| | - Coleen A McNamara
- 3 Cardiovascular Division, Department of Medicine and Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine , Charlottesville, Virginia
| | - Joachim Altschmied
- 1 IUF-Leibniz Research Institute for Environmental Medicine , Düsseldorf, Germany
| |
Collapse
|
147
|
Abstract
Cardiovascular disease is the leading cause of death worldwide, both in the general population and among patients with chronic kidney disease (CKD). In most cases, the underlying cause of the cardiovascular event is atherosclerosis - a chronic inflammatory disease. CKD accelerates atherosclerosis via augmentation of inflammation, perturbation of lipid metabolism, and other mechanisms. In the artery wall, subendothelial retention of plasma lipoproteins triggers monocyte-derived macrophages and T helper type 1 (TH1) cells to form atherosclerotic plaques. Inflammation is initiated by innate immune reactions to modified lipoproteins and is perpetuated by TH1 cells that react to autoantigens from the apolipoprotein B100 protein of LDL. Other T cells are also active in atherosclerotic lesions; regulatory T cells inhibit pathological inflammation, whereas TH17 cells can promote plaque fibrosis. The slow build-up of atherosclerotic plaques is asymptomatic, but plaque rupture or endothelial erosion can induce thrombus formation, leading to myocardial infarction or ischaemic stroke. Targeting risk factors for atherosclerosis has reduced mortality, but a need exists for novel therapies to stabilize plaques and to treat arterial inflammation. Patients with CKD would likely benefit from such preventive measures.
Collapse
Affiliation(s)
- Anton Gisterå
- Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Göran K Hansson
- Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, SE-17176 Stockholm, Sweden
| |
Collapse
|
148
|
Srikakulapu P, McNamara CA. B cells and atherosclerosis. Am J Physiol Heart Circ Physiol 2017; 312:H1060-H1067. [PMID: 28314764 DOI: 10.1152/ajpheart.00859.2016] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/08/2017] [Accepted: 03/13/2017] [Indexed: 12/14/2022]
Abstract
B cells have emerged as important immune cells in cardiovascular disease. Initial studies have suggested that B cells protect against atherosclerosis development. However, subsequent studies demonstrating aggravation of atherosclerosis by B-2 cells have shed light on the subset-dependent effects of B cells. Here, we review the literature that has led to our current understanding of B cell regulation of atherosclerosis, touching on the importance of subsets, local regulation, human translation, and therapeutic potential.
Collapse
Affiliation(s)
| | - Coleen A McNamara
- Cardiovascular Research Center, Charlottesville, Virginia; and.,Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
149
|
The vascular adventitia: An endogenous, omnipresent source of stem cells in the body. Pharmacol Ther 2017; 171:13-29. [DOI: 10.1016/j.pharmthera.2016.07.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 07/12/2016] [Indexed: 12/22/2022]
|
150
|
Ciccia F, Rizzo A, Maugeri R, Alessandro R, Croci S, Guggino G, Cavazza A, Raimondo S, Cannizzaro A, Iacopino DG, Salvarani C, Triolo G. Ectopic expression of CXCL13, BAFF, APRIL and LT-β is associated with artery tertiary lymphoid organs in giant cell arteritis. Ann Rheum Dis 2017; 76:235-243. [PMID: 27098405 DOI: 10.1136/annrheumdis-2016-209217] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/29/2016] [Accepted: 03/31/2016] [Indexed: 12/26/2022]
Abstract
OBJECTIVES To investigate whether artery tertiary lymphoid organs (ATLOs) are present in giant cell arteritis (GCA) and that their formation is associated with the ectopic expression of constitutive lymphoid tissue-homing chemokines. METHODS Reverse transcriptase PCR, immunohistochemical and immunofluorescence analysis were used to determine the presence of ectopic ATLOs in GCA and the expression of chemokines/chemokine receptors and cytokines involved in lymphoneogenesis in the temporal artery samples obtained from 50 patients with GCA and 30 controls. The presence of lymphatic conduits, of follicular dendritic cells (FDCs) precursors and lymphoid tissue inducer cells was also investigated. Finally, expression of CXCL13, B cell activating factor (BAFF), a proliferation-inducing ligand (APRIL) and CCL21 by isolated myofibroblasts was evaluated before and after stimulation with Toll-like receptors (TLRs) agonists and cytokines. RESULTS ATLOs were observed in the media layer of 60% of patients with GCA in close proximity to high endothelial venules and independently by the age of patients and the presence of atherosclerosis. ATLO formation was also accompanied by the expression of CXCL13, BAFF, a proliferation-inducing ligand (APRIL), lymphotoxin (LT)-β, interleukin (IL)-17 and IL-7, the presence of FDC precursors and of lymphoid conduits. Stimulation of myofibroblasts with TLR agonists and cytokines resulted in the upregulation of BAFF and CXCL13. CONCLUSIONS ATLOs occur in the inflamed arteries of patients with GCA possibly representing the immune sites where immune responses towards unknown arterial wall-derived antigens may be organised.
Collapse
Affiliation(s)
- Francesco Ciccia
- Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
| | - Aroldo Rizzo
- Dipartimento di Oncoematologia, Sezione di Anatomia Patologica, Azienda Ospedaliera Ospedali riuniti Villa Sofia Cervello, Palermo, Italy
| | - Rosario Maugeri
- Dipartimento di Emergenze, Urgenze e Neuroscienze Cliniche, Università di Palermo, Palermo, Italy
| | - Riccardo Alessandro
- Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di Palermo, Palermo, Italy
| | - Stefania Croci
- Clinical Immunology, Allergy and Advanced Biotechnologies Unit, Arcispedale Santa Maria Nuova, IRCCS, Reggio Emilia, Italy
| | - Giuliana Guggino
- Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
| | - Alberto Cavazza
- Pathology Unit, Arcispedale Santa Maria Nuova, IRCCS, Reggio Emilia, Italy
| | - Stefania Raimondo
- Dipartimento di Biopatologia e Biotecnologie Mediche e Forensi, Università di Palermo, Palermo, Italy
| | - Alessandra Cannizzaro
- Dipartimento di Oncoematologia, Sezione di Anatomia Patologica, Azienda Ospedaliera Ospedali riuniti Villa Sofia Cervello, Palermo, Italy
| | | | - Carlo Salvarani
- Unità operativa di Reumatologia, Arcispedale S. Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - Giovanni Triolo
- Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
| |
Collapse
|