51
|
Bellini R, Bonacina F, Norata GD. Crosstalk between dendritic cells and T lymphocytes during atherogenesis: Focus on antigen presentation and break of tolerance. Front Cardiovasc Med 2022; 9:934314. [PMID: 35966516 PMCID: PMC9365967 DOI: 10.3389/fcvm.2022.934314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/05/2022] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is a chronic disease resulting from an impaired lipid and immune homeostasis, where the interaction between innate and adaptive immune cells leads to the promotion of atherosclerosis-associated immune-inflammatory response. Emerging evidence has suggested that this response presents similarities to the reactivity of effector immune cells toward self-epitopes, often as a consequence of a break of tolerance. In this context, dendritic cells, a heterogeneous population of antigen presenting cells, play a key role in instructing effector T cells to react against foreign antigens and T regulatory cells to maintain tolerance against self-antigens and/or to patrol for self-reactive effector T cells. Alterations in this delicate balance appears to contribute to atherogenesis. The aim of this review is to discuss different DC subsets, and their role in atherosclerosis as well as in T cell polarization. Moreover, we will discuss how loss of T cell tolerogenic phenotype participates to the immune-inflammatory response associated to atherosclerosis and how a better understanding of these mechanisms might result in designing immunomodulatory therapies targeting DC-T cell crosstalk for the treatment of atherosclerosis-related inflammation.
Collapse
Affiliation(s)
- Rossella Bellini
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- *Correspondence: Fabrizia Bonacina,
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, Milan, Italy
- Giuseppe Danilo Norata,
| |
Collapse
|
52
|
Sun T, Wang F, He Y, Mao B, Han M, Liu H, Zhao P, Li X, Wang D. Enlarged Pericarotid Lymph Nodes Suggest Recent Ischemic Symptoms in Patients with Carotid Atherosclerosis. Front Immunol 2022; 13:900642. [PMID: 35903093 PMCID: PMC9315152 DOI: 10.3389/fimmu.2022.900642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/23/2022] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease closely associated with immunological activity. Lymph nodes (LNs) are essential secondary lymphoid organs, in which complex immune responses occur. Enlarged LNs are commonly observed around inflamed tissues or tumors; however, their role in atherosclerosis is not well understood. We hypothesized that enlarged pericarotid LNs would be present in symptomatic patients with carotid atherosclerosis. Therefore, we recorded the size of LNs around the carotid artery during surgery in patients undergoing carotid endarterectomy (CEA) for carotid atherosclerotic stenosis. Patients were stratified by enlarged LNs, defined as a diameter ≥ 10mm in the transverse diameters. Demographic and clinical data of participants were measured and analyzed. Hematoxylin and eosin (H&E), Sirius red, DAB-enhanced Perls’ Prussian blue, alizarin red, and immunohistochemistry (IHC) staining were performed for composition identification of plaques or LNs. Symptomatic patients were defined as those presenting with an ipsilateral cerebral ischemic event. Compared with patients with non-enlarged LNs, patients with enlarged LNs were more likely to be symptomatic (22/32, 68.8% versus 9/40, 22.5%, P < 0.001) and use calcium channel blocker drugs (17/32, 53.1% versus 10/40, 25%, P=0.014). In addition, they showed lower body mass index (mean ± SD: 24.00 ± 2.66 versus 25.34 ± 2.56 kg/m2, P=0.034), lower weight (median [interquartile range]: 64 [60.00-76.00] versus 72.5 [65.00-77.50] Kg, P = 0.046) and higher diastolic blood pressure (mean ± SD: 78.94 ± 9.30 versus 73.93 ± 8.84 mmHg, P = 0.022). The plague from patients with enlarged LNs exhibited a lower relative percentage of fibrous tissue (29.49 ± 10.73% versus 34.62 ± 10.33%, P = 0.041). The enlarged LNs remained oval-shaped by visual inspection. Compared to non-enlarged LNs, the predominant changes in enlarged LNs were atrophic lymphatic sinuses and dilated LNs parenchyma. Enlarged LNs contained more germinal centers and lymphocytes. In conclusion, symptomatic patients with carotid atherosclerosis have enlarged pericarotid LNs. The current study supports the conclusion that enlarged LNs with an activated and enhanced adaptive immune response may indicate plaque instability. Pericarotid LNs will be a promising marker of plaque stability and may be a potential therapeutic target in patients with carotid atherosclerosis.
Collapse
Affiliation(s)
- Tao Sun
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fei Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yiming He
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bo Mao
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Mengtao Han
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Han Liu
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peng Zhao
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Xingang Li, ; Donghai Wang,
| | - Donghai Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Xingang Li, ; Donghai Wang,
| |
Collapse
|
53
|
Rossi A, Belmonte B, Carnevale S, Liotti A, De Rosa V, Jaillon S, Piconese S, Tripodo C. Stromal and Immune Cell Dynamics in Tumor Associated Tertiary Lymphoid Structures and Anti-Tumor Immune Responses. Front Cell Dev Biol 2022; 10:933113. [PMID: 35874810 PMCID: PMC9304551 DOI: 10.3389/fcell.2022.933113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are ectopic lymphoid organs that have been observed in chronic inflammatory conditions including cancer, where they are thought to exert a positive effect on prognosis. Both immune and non-immune cells participate in the genesis of TLS by establishing complex cross-talks requiring both soluble factors and cell-to-cell contact. Several immune cell types, including T follicular helper cells (Tfh), regulatory T cells (Tregs), and myeloid cells, may accumulate in TLS, possibly promoting or inhibiting their development. In this manuscript, we propose to review the available evidence regarding specific aspects of the TLS formation in solid cancers, including 1) the role of stromal cell composition and architecture in the recruitment of specific immune subpopulations and the formation of immune cell aggregates; 2) the contribution of the myeloid compartment (macrophages and neutrophils) to the development of antibody responses and the TLS formation; 3) the immunological and metabolic mechanisms dictating recruitment, expansion and plasticity of Tregs into T follicular regulatory cells, which are potentially sensitive to immunotherapeutic strategies directed to costimulatory receptors or checkpoint molecules.
Collapse
Affiliation(s)
- Alessandra Rossi
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | | | - Antonietta Liotti
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche, Naples, Italy
| | - Veronica De Rosa
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche, Naples, Italy
| | - Sebastien Jaillon
- RCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Silvia Piconese
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
- IRCCS Fondazione Santa Lucia, Unità di Neuroimmunologia, Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Rome, Italy
- *Correspondence: Silvia Piconese,
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care “G. D’Alessandro”, University of Palermo, Palermo, Italy
- Histopathology Unit, FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| |
Collapse
|
54
|
Role of tertiary lymphoid organs in the regulation of immune responses in the periphery. Cell Mol Life Sci 2022; 79:359. [PMID: 35689679 PMCID: PMC9188279 DOI: 10.1007/s00018-022-04388-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/28/2022] [Accepted: 05/20/2022] [Indexed: 12/12/2022]
Abstract
Tertiary lymphoid organs (TLOs) are collections of immune cells resembling secondary lymphoid organs (SLOs) that form in peripheral, non-lymphoid tissues in response to local chronic inflammation. While their formation mimics embryologic lymphoid organogenesis, TLOs form after birth at ectopic sites in response to local inflammation resulting in their ability to mount diverse immune responses. The structure of TLOs can vary from clusters of B and T lymphocytes to highly organized structures with B and T lymphocyte compartments, germinal centers, and lymphatic vessels (LVs) and high endothelial venules (HEVs), allowing them to generate robust immune responses at sites of tissue injury. Although our understanding of the formation and function of these structures has improved greatly over the last 30 years, their role as mediators of protective or pathologic immune responses in certain chronic inflammatory diseases remains enigmatic and may differ based on the local tissue microenvironment in which they form. In this review, we highlight the role of TLOs in the regulation of immune responses in chronic infection, chronic inflammatory and autoimmune diseases, cancer, and solid organ transplantation.
Collapse
|
55
|
Gisterå A, Ketelhuth DFJ, Malin SG, Hansson GK. Animal Models of Atherosclerosis-Supportive Notes and Tricks of the Trade. Circ Res 2022; 130:1869-1887. [PMID: 35679358 DOI: 10.1161/circresaha.122.320263] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Atherosclerotic cardiovascular disease is a major cause of death among humans. Animal models have shown that cholesterol and inflammation are causatively involved in the disease process. Apolipoprotein B-containing lipoproteins elicit immune reactions and instigate inflammation in the vessel wall. Still, a treatment that is specific to vascular inflammation is lacking, which motivates continued in vivo investigations of the immune-vascular interactions that drive the disease. In this review, we distill old notions with emerging concepts into a contemporary understanding of vascular disease models. Pros and cons of different models are listed and the complex integrative interplay between cholesterol homeostasis, immune activation, and adaptations of the vascular system is discussed. Key limitations with atherosclerosis models are highlighted, and we suggest improvements that could accelerate progress in the field. However, excessively rigid experimental guidelines or limiting usage to certain animal models can be counterproductive. Continued work in improved models, as well as the development of new models, should be of great value in research and could aid the development of cardiovascular disease diagnostics and therapeutics of the future.
Collapse
Affiliation(s)
- Anton Gisterå
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden (A.G., D.F.J.K., S.G.M., G.K.H.)
| | - Daniel F J Ketelhuth
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden (A.G., D.F.J.K., S.G.M., G.K.H.).,Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark (SDU), Odense, Denmark (D.F.J.K)
| | - Stephen G Malin
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden (A.G., D.F.J.K., S.G.M., G.K.H.)
| | - Göran K Hansson
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden (A.G., D.F.J.K., S.G.M., G.K.H.)
| |
Collapse
|
56
|
Zhao H, Wang H, Zhao Y, Sun Q, Ren X. Tumor-Resident T Cells, Associated With Tertiary Lymphoid Structure Maturity, Improve Survival in Patients With Stage III Lung Adenocarcinoma. Front Immunol 2022; 13:877689. [PMID: 35663939 PMCID: PMC9161276 DOI: 10.3389/fimmu.2022.877689] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/21/2022] [Indexed: 11/24/2022] Open
Abstract
Tertiary lymphoid structure (TLS) and tumor-resident memory T cells (TRM) play crucial roles in the anti-tumor immune response, facilitating a good prognosis in patients with cancer. However, there have been no reports on the relationship between TRM and TLS maturity. In this study, we detected TRM and the maturity of TLS by immunofluorescence staining and analyzed the relationship between their distribution and proportion in patients with lung adenocarcinoma (LUAD). The proportion of TRM within TLSs was significantly higher than that outside and was positively correlated with the survival of patients. In addition, the proportions of CD4+CD103+ TRM and CD8+CD103+ TRM were significantly increased with the gradually maturation of TLSs. We divided the patients into three levels (grade 1, grade 2, and grade 3) according to the presence of increasing maturation of TLSs. The proportion of CD103+ TRM in grade 3 patients was significantly higher than that in grade 1 and grade 2 patients, suggesting a close relationship between CD103+ TRM and TLS maturity. Furthermore, positive prognosis was associated with grade 3 patients that exhibited CD103+T RM High phenotype.
Collapse
Affiliation(s)
- Hua Zhao
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Hao Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Yu Zhao
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Qian Sun
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
57
|
Mohanta SK, Peng L, Li Y, Lu S, Sun T, Carnevale L, Perrotta M, Ma Z, Förstera B, Stanic K, Zhang C, Zhang X, Szczepaniak P, Bianchini M, Saeed BR, Carnevale R, Hu D, Nosalski R, Pallante F, Beer M, Santovito D, Ertürk A, Mettenleiter TC, Klupp BG, Megens RTA, Steffens S, Pelisek J, Eckstein HH, Kleemann R, Habenicht L, Mallat Z, Michel JB, Bernhagen J, Dichgans M, D'Agostino G, Guzik TJ, Olofsson PS, Yin C, Weber C, Lembo G, Carnevale D, Habenicht AJR. Neuroimmune cardiovascular interfaces control atherosclerosis. Nature 2022; 605:152-159. [PMID: 35477759 DOI: 10.1038/s41586-022-04673-6] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 01/31/2022] [Indexed: 02/08/2023]
Abstract
Atherosclerotic plaques develop in the inner intimal layer of arteries and can cause heart attacks and strokes1. As plaques lack innervation, the effects of neuronal control on atherosclerosis remain unclear. However, the immune system responds to plaques by forming leukocyte infiltrates in the outer connective tissue coat of arteries (the adventitia)2-6. Here, because the peripheral nervous system uses the adventitia as its principal conduit to reach distant targets7-9, we postulated that the peripheral nervous system may directly interact with diseased arteries. Unexpectedly, widespread neuroimmune cardiovascular interfaces (NICIs) arose in mouse and human atherosclerosis-diseased adventitia segments showed expanded axon networks, including growth cones at axon endings near immune cells and media smooth muscle cells. Mouse NICIs established a structural artery-brain circuit (ABC): abdominal adventitia nociceptive afferents10-14 entered the central nervous system through spinal cord T6-T13 dorsal root ganglia and were traced to higher brain regions, including the parabrachial and central amygdala neurons; and sympathetic efferent neurons projected from medullary and hypothalamic neurons to the adventitia through spinal intermediolateral neurons and both coeliac and sympathetic chain ganglia. Moreover, ABC peripheral nervous system components were activated: splenic sympathetic and coeliac vagus nerve activities increased in parallel to disease progression, whereas coeliac ganglionectomy led to the disintegration of adventitial NICIs, reduced disease progression and enhanced plaque stability. Thus, the peripheral nervous system uses NICIs to assemble a structural ABC, and therapeutic intervention in the ABC attenuates atherosclerosis.
Collapse
Affiliation(s)
- Sarajo K Mohanta
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany. .,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| | - Li Peng
- Department of Cardiovascular Internal Medicine, Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yuanfang Li
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Shu Lu
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Ting Sun
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Lorenzo Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Pozzilli, Italy
| | - Marialuisa Perrotta
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Pozzilli, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Zhe Ma
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Benjamin Förstera
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität Munich (LMU), Munich, Germany
| | - Karen Stanic
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität Munich (LMU), Munich, Germany
| | - Chuankai Zhang
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Xi Zhang
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Piotr Szczepaniak
- Department of Internal and Agricultural Medicine, Jagiellonian University Collegium Medicum, Krakow, Poland
| | - Mariaelvy Bianchini
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Borhan R Saeed
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Raimondo Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Pozzilli, Italy
| | - Desheng Hu
- Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Munich, Germany
| | - Ryszard Nosalski
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Fabio Pallante
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Pozzilli, Italy
| | - Michael Beer
- Department for Information Technology, University of Jena, Jena University Hospital, Jena, Germany
| | - Donato Santovito
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Institute for Genetic and Biomedical Research, Unit of Milan, National Research Council, Milan, Italy
| | - Ali Ertürk
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität Munich (LMU), Munich, Germany
| | - Thomas C Mettenleiter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Barbara G Klupp
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Remco T A Megens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Jaroslav Pelisek
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Vascular Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Hans-Henning Eckstein
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands.,Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Livia Habenicht
- II. Medizinische Klinik und Poliklinik, Technische Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Ziad Mallat
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Jean-Baptiste Michel
- Laboratory for Vascular Translational Science, INSERM UMRS 1148, University Paris Diderot (P7), GH Bichat-Claude Bernard, Paris, France
| | - Jürgen Bernhagen
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität Munich (LMU), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität Munich (LMU), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Giuseppe D'Agostino
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Tomasz J Guzik
- Department of Internal and Agricultural Medicine, Jagiellonian University Collegium Medicum, Krakow, Poland.,Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Peder S Olofsson
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Changjun Yin
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Pozzilli, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Pozzilli, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Andreas J R Habenicht
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München (LMU), Munich, Germany. .,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
58
|
Fleig S, Kapanadze T, Bernier-Latmani J, Lill JK, Wyss T, Gamrekelashvili J, Kijas D, Liu B, Hüsing AM, Bovay E, Jirmo AC, Halle S, Ricke-Hoch M, Adams RH, Engel DR, von Vietinghoff S, Förster R, Hilfiker-Kleiner D, Haller H, Petrova TV, Limbourg FP. Loss of vascular endothelial notch signaling promotes spontaneous formation of tertiary lymphoid structures. Nat Commun 2022; 13:2022. [PMID: 35440634 PMCID: PMC9018798 DOI: 10.1038/s41467-022-29701-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 03/21/2022] [Indexed: 12/20/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are lymph node-like immune cell clusters that emerge during chronic inflammation in non-lymphoid organs like the kidney, but their origin remains not well understood. Here we show, using conditional deletion strategies of the canonical Notch signaling mediator Rbpj, that loss of endothelial Notch signaling in adult mice induces the spontaneous formation of bona fide TLS in the kidney, liver and lung, based on molecular, cellular and structural criteria. These TLS form in a stereotypical manner around parenchymal arteries, while secondary lymphoid structures remained largely unchanged. This effect is mediated by endothelium of blood vessels, but not lymphatics, since a lymphatic endothelial-specific targeting strategy did not result in TLS formation, and involves loss of arterial specification and concomitant acquisition of a high endothelial cell phenotype, as shown by transcriptional analysis of kidney endothelial cells. This indicates a so far unrecognized role for vascular endothelial cells and Notch signaling in TLS initiation. Loss of canonical Notch signaling in vascular endothelial cells induces spontaneous formation of proto-typical tertiary lymphoid structures in mouse kidney, liver and lungs, which form around central arteries that acquire a high endothelial cell signature
Collapse
Affiliation(s)
- Susanne Fleig
- Vascular Medicine Research, Hannover Medical School, 30625, Hannover, Germany.,Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany.,Department of Geriatric Medicine (Medical Clinic VI), RWTH Aachen University Hospital, 52074, Aachen, Germany
| | - Tamar Kapanadze
- Vascular Medicine Research, Hannover Medical School, 30625, Hannover, Germany.,Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany
| | - Jeremiah Bernier-Latmani
- Vascular and Tumor Biology Laboratory, Department of Oncology UNIL CHUV and Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Julia K Lill
- Department of Immunodynamics, Institute for Experimental Immunology and Imaging, Medical Research Centre, University Hospital Essen, 45147, Essen, Germany
| | - Tania Wyss
- Vascular and Tumor Biology Laboratory, Department of Oncology UNIL CHUV and Ludwig Institute for Cancer Research, Lausanne, Switzerland.,SIB Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - Jaba Gamrekelashvili
- Vascular Medicine Research, Hannover Medical School, 30625, Hannover, Germany.,Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany
| | - Dustin Kijas
- Vascular Medicine Research, Hannover Medical School, 30625, Hannover, Germany.,Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany
| | - Bin Liu
- Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Anne M Hüsing
- Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany
| | - Esther Bovay
- Max-Planck-Institute for Molecular Biomedicine, 48149, Muenster, Germany
| | - Adan Chari Jirmo
- Hannover Medical School, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.,Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Stephan Halle
- Institute of Immunology, Hannover Medical School, 30625, Hannover, Germany
| | - Melanie Ricke-Hoch
- Department of Cardiology and Angiology, Hannover Medical School, 30625, Hannover, Germany
| | - Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, 48149, Muenster, Germany
| | - Daniel R Engel
- Department of Immunodynamics, Institute for Experimental Immunology and Imaging, Medical Research Centre, University Hospital Essen, 45147, Essen, Germany
| | - Sibylle von Vietinghoff
- Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany.,Division of Medicine I, Nephrology section, UKB Bonn University Hospital, Bonn, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, 30625, Hannover, Germany
| | - Denise Hilfiker-Kleiner
- Department of Cardiology and Angiology, Hannover Medical School, 30625, Hannover, Germany.,Department of Cardiovascular Complications of Oncologic Therapies, Medical Faculty of the Philipps University Marburg, 35037, Marburg, Germany
| | - Hermann Haller
- Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany
| | - Tatiana V Petrova
- Vascular and Tumor Biology Laboratory, Department of Oncology UNIL CHUV and Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Florian P Limbourg
- Vascular Medicine Research, Hannover Medical School, 30625, Hannover, Germany. .,Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
59
|
Liu Y, Zou L, Tang H, Li J, Liu H, Jiang X, Jiang B, Dong Z, Fu W. Single-Cell Sequencing of Immune Cells in Human Aortic Dissection Tissue Provides Insights Into Immune Cell Heterogeneity. Front Cardiovasc Med 2022; 9:791875. [PMID: 35433892 PMCID: PMC9008490 DOI: 10.3389/fcvm.2022.791875] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 03/14/2022] [Indexed: 11/25/2022] Open
Abstract
Background Inflammation plays an important role in the progression of sporadic aortic dissection (AD). Immune cells, especially macrophages, infiltrate the aorta and secrete inflammatory cytokines and matrix metalloproteinases to cause degradation of the extracellular matrix, thereby contributing to the pathogenesis of AD. However, the cellular heterogeneity within these immune cells has not been fully characterized. Methods We used single-cell RNA sequencing to profile the transcriptomes of all immune cells in AD tissue and normal aorta. Using magnetic-activated cell sorting gating on CD45, we obtained a higher resolution identification of the immune cell subsets in the aorta. Results We observed significant differences in the proportion of major immune cell subpopulations between AD and normal aorta tissues. Macrophages accounted for a higher percentage in the normal aorta, while the proportions of T cells, B cells and natural killer (NK) cells were all increased in AD tissues. Macrophage clusters that expanded in AD tissues originated primarily from circulating monocytes and expressed genes encoding proinflammatory cytokines and molecules involved in tissue repair. T and NK cells in AD tissues exhibited enhanced cytotoxic properties. A cluster of CD4+ T cells that had expanded in AD tissues was Th17-like and might contribute to the pathogenesis of AD. Cell–cell interaction analysis highlighted the increased communication between macrophages and T cells, which primarily regulated the costimulation of T cells. Conclusions Our study provides a comprehensive characterization of immune cells in the dissected aorta with an emphasis on the role of macrophages and T cells. The information from our study improves our understanding of immune mechanisms in AD formation and helps to identify additional useful targets for early diagnosis or therapy of AD.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Lingwei Zou
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Hanfei Tang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Jie Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hao Liu
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xiaolang Jiang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Baohong Jiang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Weiguo Fu
| | - Zhihui Dong
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Zhihui Dong
| | - Weiguo Fu
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Baohong Jiang
| |
Collapse
|
60
|
Zhang X, Wang Z, Zhang C, Li Y, Lu S, Steffens S, Mohanta S, Weber C, Habenicht A, Yin C. Laser Capture Microdissection-Based mRNA Expression Microarrays and Single-Cell RNA Sequencing in Atherosclerosis Research. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:715-726. [PMID: 35237997 DOI: 10.1007/978-1-0716-1924-7_43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
A major goal of methodologies related to large scale gene expression analyses is to initiate comprehensive information on transcript signatures in single cells within the tissue's anatomy. Until now, this could be achieved in a stepwise experimental approach: (1) identify the majority of transcripts in a single cell (single cell transcriptome); (2) provide information on transcripts on multiple cell subtypes in a complex sample (cell heterogeneity); and (3) give information on each cell's spatial location within the tissue (zonation transcriptomics). Such genetic information will allow construction of functionally relevant gene expression maps of single cells of a given anatomically defined tissue compartment and thus pave the way for subsequent analyses, including their epigenetic modifications. Until today these aims have not been achieved in the area of cardiovascular disease research though steps toward these goals become apparent: laser capture microdissection (LCM)-based mRNA expression microarrays of atherosclerotic plaques were applied to gain information on local gene expression changes during disease progression, providing limited spatial resolution. Moreover, while LCM-derived tissue RNA extracts have been shown to be highly sensitive and covers a range of 10-16,000 genes per array/small amount of RNA, its original promise to isolate single cells from a tissue section turned out not to be practicable because of the inherent contamination of the cell's RNA of interest with RNA from neighboring cells. Many shortcomings of LCM-based analyses have been overcome using single-cell RNA sequencing (scRNA-seq) technologies though scRNA-seq also has several limitations including low numbers of transcripts/cell and the complete loss of spatial information. Here, we describe a protocol toward combining advantages of both techniques while avoiding their flaws.
Collapse
Affiliation(s)
- Xi Zhang
- Institute for Cardiovascular Prevention (IPEK), Klinikum of the University of Munich (KUM), Ludwig-Maximilians-University (LMU), Munich, Germany.
| | - Zhihua Wang
- Institute for Cardiovascular Prevention (IPEK), Klinikum of the University of Munich (KUM), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Chuankai Zhang
- Institute for Cardiovascular Prevention (IPEK), Klinikum of the University of Munich (KUM), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Yutao Li
- Institute for Cardiovascular Prevention (IPEK), Klinikum of the University of Munich (KUM), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Shu Lu
- Institute for Cardiovascular Prevention (IPEK), Klinikum of the University of Munich (KUM), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Klinikum of the University of Munich (KUM), Ludwig-Maximilians-University (LMU), Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Munich Heart Alliance, Munich, Germany
| | - Sarajo Mohanta
- Institute for Cardiovascular Prevention (IPEK), Klinikum of the University of Munich (KUM), Ludwig-Maximilians-University (LMU), Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Munich Heart Alliance, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Klinikum of the University of Munich (KUM), Ludwig-Maximilians-University (LMU), 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.,Munich Cluster of Systems Neurology (SyNergy), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andreas Habenicht
- Institute for Cardiovascular Prevention (IPEK), Klinikum of the University of Munich (KUM), Ludwig-Maximilians-University (LMU), Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Munich Heart Alliance, Munich, Germany
| | - Changjun Yin
- Institute for Cardiovascular Prevention (IPEK), Klinikum of the University of Munich (KUM), Ludwig-Maximilians-University (LMU), Munich, Germany. .,German Center for Cardiovascular Research (DZHK), Partner site Munich Heart Alliance, Munich, Germany. .,Munich Cluster of Systems Neurology (SyNergy), Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
61
|
Combined Single-Cell RNA and Single-Cell α/β T Cell Receptor Sequencing of the Arterial Wall in Atherosclerosis. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2419:727-746. [PMID: 35237998 DOI: 10.1007/978-1-0716-1924-7_44] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Although various pro- and anti-inflammatory T cell subsets have been observed in murine and human atherosclerosis, principal issues of T cell immunity remain unanswered: Is atherosclerosis progression critically affected by aberrant T cell responses? Are tolerance checkpoints compromised during atherosclerosis progression? Answers to these questions will determine if we are at the cusp of developing T cell-dependent therapeutic strategies. Rapid advances in single cell RNA sequencing (scRNA-seq) and single cell α/β T cell receptor (TCR) (scTCR) sequencing allows to address these issues in unprecedented ways. The majority of T cells recognize peptide antigen-MHC complexes presented by antigen-presenting cells which, in turn, trigger activation and proliferation (clonal expansion) of cognate TCR-carrying T cells. Thus, clonal expansion and their corresponding transcriptome are two similarly important sides of T cell immunity and both will-as hypothesized-affect the outcome of atherosclerosis. Here, we combined scRNA-seq and scTCR-seq in single cells. Moreover, we provide single T cell transcriptomes and TCR maps of three important tissues involved in atherosclerosis This approach is anticipated to address principal questions concerning atherosclerosis autoimmunity that are likely to pave the long sought way to T cell-dependent therapeutic approaches.
Collapse
|
62
|
Funk-Hilsdorf TC, Behrens F, Grune J, Simmons S. Dysregulated Immunity in Pulmonary Hypertension: From Companion to Composer. Front Physiol 2022; 13:819145. [PMID: 35250621 PMCID: PMC8891568 DOI: 10.3389/fphys.2022.819145] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/20/2022] [Indexed: 12/26/2022] Open
Abstract
Pulmonary hypertension (PH) represents a grave condition associated with high morbidity and mortality, emphasizing a desperate need for innovative and targeted therapeutic strategies. Cumulative evidence suggests that inflammation and dysregulated immunity interdependently affect maladaptive organ perfusion and congestion as hemodynamic hallmarks of the pathophysiology of PH. The role of altered cellular and humoral immunity in PH gains increasing attention, especially in pulmonary arterial hypertension (PAH), revealing novel mechanistic insights into the underlying immunopathology. Whether these immunophysiological aspects display a universal character and also hold true for other types of PH (e.g., PH associated with left heart disease, PH-LHD), or whether there are unique immunological signatures depending on the underlying cause of disease are points of consideration and discussion. Inflammatory mediators and cellular immune circuits connect the local inflammatory landscape in the lung and heart through inter-organ communication, involving, e.g., the complement system, sphingosine-1-phosphate (S1P), cytokines and subsets of, e.g., monocytes, macrophages, natural killer (NK) cells, dendritic cells (DCs), and T- and B-lymphocytes with distinct and organ-specific pro- and anti-inflammatory functions in homeostasis and disease. Perivascular macrophage expansion and monocyte recruitment have been proposed as key pathogenic drivers of vascular remodeling, the principal pathological mechanism in PAH, pinpointing toward future directions of anti-inflammatory therapeutic strategies. Moreover, different B- and T-effector cells as well as DCs may play an important role in the pathophysiology of PH as an imbalance of T-helper-17-cells (TH17) activated by monocyte-derived DCs, a potentially protective role of regulatory T-cells (Treg) and autoantibody-producing plasma cells occur in diverse PH animal models and human PH. This article highlights novel aspects of the innate and adaptive immunity and their interaction as disease mediators of PH and its specific subtypes, noticeable inflammatory mediators and summarizes therapeutic targets and strategies arising thereby.
Collapse
Affiliation(s)
- Teresa C. Funk-Hilsdorf
- Junior Research Group “Immunodynamics”, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Felix Behrens
- Junior Research Group “Immunodynamics”, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Jana Grune
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Szandor Simmons
- Junior Research Group “Immunodynamics”, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Laboratory of Lung Vascular Research, Institute of Physiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- *Correspondence: Szandor Simmons,
| |
Collapse
|
63
|
CD169 + macrophages in lymph node and spleen critically depend on dual RANK and LTbetaR signaling. Proc Natl Acad Sci U S A 2022; 119:2108540119. [PMID: 35031565 PMCID: PMC8784161 DOI: 10.1073/pnas.2108540119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2021] [Indexed: 12/16/2022] Open
Abstract
The CD169+ macrophages that play an important role in the fight against infections and cancer are receptive to environmental signals for their differentiation. We show that lymph node and splenic CD169+ macrophages require both LTβR and RANK signaling since the conditional deficiency of either receptor results in their disappearance. Using a reporter mouse, we observe RANKL expression by a splenic mesenchymal cell subset and show that it participates in CD169+ macrophage differentiation. Their absence leads to a reduced viral capture and a greatly attenuated virus-specific CD8+ T cell expansion. Thus, tight control mechanisms operate for the precise positioning of these macrophages at sites where numerous immune-stimulatory forces converge. CD169+ macrophages reside in lymph node (LN) and spleen and play an important role in the immune defense against pathogens. As resident macrophages, they are responsive to environmental cues to shape their tissue-specific identity. We have previously shown that LN CD169+ macrophages require RANKL for formation of their niche and their differentiation. Here, we demonstrate that they are also dependent on direct lymphotoxin beta (LTβ) receptor (R) signaling. In the absence or the reduced expression of either RANK or LTβR, their differentiation is perturbed, generating myeloid cells expressing SIGN-R1 in LNs. Conditions of combined haploinsufficiencies of RANK and LTβR revealed that both receptors contribute equally to LN CD169+ macrophage differentiation. In the spleen, the Cd169-directed ablation of either receptor results in a selective loss of marginal metallophilic macrophages (MMMs). Using a RANKL reporter mouse, we identify splenic marginal zone stromal cells as a source of RANKL and demonstrate that it participates in MMM differentiation. The loss of MMMs had no effect on the splenic B cell compartments but compromised viral capture and the expansion of virus-specific CD8+ T cells. Taken together, the data provide evidence that CD169+ macrophage differentiation in LN and spleen requires dual signals from LTβR and RANK with implications for the immune response.
Collapse
|
64
|
Craig DJ, James AW, Wang Y, Tavian M, Crisan M, Péault BM. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:35-43. [PMID: 35641167 PMCID: PMC8895497 DOI: 10.1093/stcltm/szab001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/30/2020] [Indexed: 11/17/2022] Open
Abstract
The vascular wall is comprised of distinct layers controlling angiogenesis, blood flow, vessel anchorage within organs, and cell and molecule transit between blood and tissues. Moreover, some blood vessels are home to essential stem-like cells, a classic example being the existence in the embryo of hemogenic endothelial cells at the origin of definitive hematopoiesis. In recent years, microvascular pericytes and adventitial perivascular cells were observed to include multi-lineage progenitor cells involved not only in organ turnover and regeneration but also in pathologic remodeling, including fibrosis and atherosclerosis. These perivascular mesodermal elements were identified as native forerunners of mesenchymal stem cells. We have presented in this brief review our current knowledge on vessel wall-associated tissue remodeling cells with respect to discriminating phenotypes, functional diversity in health and disease, and potential therapeutic interest.
Collapse
Affiliation(s)
- David J Craig
- Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Mihaela Crisan
- Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Bruno M Péault
- Center for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA, USA
- Corresponding author: Bruno Péault, PhD, Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California at Los Angeles, 615 Charles E. Young Drive South, Los Angeles, CA 90095-7358, USA.
| |
Collapse
|
65
|
Feng X, Du M, Zhang Y, Ding J, Wang Y, Liu P. The Role of Lymphangiogenesis in Coronary Atherosclerosis. Lymphat Res Biol 2021; 20:290-301. [PMID: 34714136 DOI: 10.1089/lrb.2021.0026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Lymphatic circulation, a one-way channel system independent of blood circulation, collects interstitial fluid in a blind-end way. Existing widely in various organs and tissues, lymphatic vessels play important roles in maintaining tissue fluid homeostasis, regulating immune function, and promoting lipid transport. Recent studies have shown clear evidence that lymphangiogenesis has a strong mutual effect on coronary atherosclerosis (AS). In this study, we focus on this topic, especially in the aspects of relevant ligand/receptor, inflammation, and adipose metabolism. For the moment, however, the role of lymphangiogenesis and remodeling in coronary AS still remains controversial. The studies of our group and accumulating published evidence show that the pathological remodeling of lymphatic vessels in coronary AS may have a negative effect, but normal functional lymphangiogenesis is probably beneficial to the regression of coronary AS. Thus, the conclusion of this review is that lymphatic vessel function rather than its quantity determines its influence in AS, which needs more evidence to support.
Collapse
Affiliation(s)
- Xiaoteng Feng
- Department of Cardiology, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min Du
- Department of Cardiology, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifan Zhang
- Department of Cardiology, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Ding
- Department of Cardiology, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiru Wang
- Department of Cardiology, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Liu
- Department of Cardiology, LongHua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
66
|
Jamaly S, Rakaee M, Abdi R, Tsokos GC, Fenton KA. Interplay of immune and kidney resident cells in the formation of tertiary lymphoid structures in lupus nephritis. Autoimmun Rev 2021; 20:102980. [PMID: 34718163 DOI: 10.1016/j.autrev.2021.102980] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 07/31/2021] [Indexed: 02/07/2023]
Abstract
Kidney involvement confers significant morbidity and mortality in patients with systemic lupus erythematosus (SLE). The pathogenesis of lupus nephritis (LN) involves diverse mechanisms instigated by elements of the autoimmune response which alter the biology of kidney resident cells. Processes in the glomeruli and in the interstitium may proceed independently albeit crosstalk between the two is inevitable. Podocytes, mesangial cells, tubular epithelial cells, kidney resident macrophages and stromal cells with input from cytokines and autoantibodies present in the circulation alter the expression of enzymes, produce cytokines and chemokines which lead to their injury and damage of the kidney. Several of these molecules can be targeted independently to prevent and reverse kidney failure. Tertiary lymphoid structures with true germinal centers are present in the kidneys of patients with lupus nephritis and have been increasingly recognized to associate with poorer renal outcomes. Stromal cells, tubular epithelial cells, high endothelial vessel and lymphatic venule cells produce chemokines which enable the formation of structures composed of a T-cell-rich zone with mature dendritic cells next to a B-cell follicle with the characteristics of a germinal center surrounded by plasma cells. Following an overview on the interaction of the immune cells with kidney resident cells, we discuss the cellular and molecular events which lead to the formation of tertiary lymphoid structures in the interstitium of the kidneys of mice and patients with lupus nephritis. In parallel, molecules and processes that can be targeted therapeutically are presented.
Collapse
Affiliation(s)
- Simin Jamaly
- Department of Medical Biology, Faculty of Health Science, UiT The Arctic University of Norway, N-9037 Tromsø, Norway; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Mehrdad Rakaee
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Reza Abdi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kristin Andreassen Fenton
- Department of Medical Biology, Faculty of Health Science, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| |
Collapse
|
67
|
Herrero-Cervera A, Espinós-Estévez C, Martín-Vañó S, Taberner-Cortés A, Aguilar-Ballester M, Vinué Á, Piqueras L, Martínez-Hervás S, González-Navarro H. Dissecting Abdominal Aortic Aneurysm Is Aggravated by Genetic Inactivation of LIGHT (TNFSF14). Biomedicines 2021; 9:biomedicines9111518. [PMID: 34829747 PMCID: PMC8615201 DOI: 10.3390/biomedicines9111518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022] Open
Abstract
Abdominal aortic aneurysm (AAA), is a complex disorder characterized by vascular vessel wall remodeling. LIGHT (TNFSF14) is a proinflammatory cytokine associated with vascular disease. In the present study, the impact of genetic inactivation of Light was investigated in dissecting AAA induced by angiotensin II (AngII) in the Apolipoprotein E-deficient (Apoe−/−) mice. Studies in aortic human (ah) vascular smooth muscle cells (VSMC) to study potential translation to human pathology were also performed. AngII-treated Apoe−/−Light−/− mice displayed increased abdominal aorta maximum diameter and AAA severity compared with Apoe−/− mice. Notably, reduced smooth muscle α-actin+ area and Acta2 and Col1a1 gene expression were observed in AAA from Apoe−/−Light−/− mice, suggesting a loss of VSMC contractile phenotype compared with controls. Decreased Opn and augmented Sox9 expression, which are associated with detrimental and non-contractile osteochondrogenic VSMC phenotypes, were also seen in AngII-treated Apoe−/−Light−/− mouse AAA. Consistent with a role of LIGHT preserving VSMC contractile characteristics, LIGHT-treatment of ahVSMCs diminished the expression of SOX9 and of the pluripotency marker CKIT. These effects were partly mediated through lymphotoxin β receptor (LTβR) as the silencing of its gene ablated LIGHT effects on ahVSMCs. These studies suggest a protective role of LIGHT through mechanisms that prevent VSMC trans-differentiation in an LTβR-dependent manner.
Collapse
Affiliation(s)
- Andrea Herrero-Cervera
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
| | | | - Susana Martín-Vañó
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
| | - Alida Taberner-Cortés
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
| | - María Aguilar-Ballester
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
| | - Ángela Vinué
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
| | - Laura Piqueras
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
- Department of Pharmacology, University of Valencia, 46010 Valencia, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Sergio Martínez-Hervás
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Endocrinology and Nutrition Service, Clinic Hospital of Valencia, 46010 Valencia, Spain
- Department of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Herminia González-Navarro
- INCLIVA, Institute of Health Research, 46010 Valencia, Spain; (A.H.-C.); (S.M.-V.); (A.T.-C.); (M.A.-B.); (Á.V.); (L.P.); (S.M.-H.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Biochemistry and Molecular Biology Department, University of Valencia, 46010 Valencia, Spain
- Correspondence: ; Tel.: +34-96-386-44-03; Fax: +34-96-398-78-60
| |
Collapse
|
68
|
Merlin J, Ivanov S, Dumont A, Sergushichev A, Gall J, Stunault M, Ayrault M, Vaillant N, Castiglione A, Swain A, Orange F, Gallerand A, Berton T, Martin JC, Carobbio S, Masson J, Gaisler-Salomon I, Maechler P, Rayport S, Sluimer JC, Biessen EAL, Guinamard RR, Gautier EL, Thorp EB, Artyomov MN, Yvan-Charvet L. Non-canonical glutamine transamination sustains efferocytosis by coupling redox buffering to oxidative phosphorylation. Nat Metab 2021; 3:1313-1326. [PMID: 34650273 PMCID: PMC7611882 DOI: 10.1038/s42255-021-00471-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 09/06/2021] [Indexed: 12/04/2022]
Abstract
Macrophages rely on tightly integrated metabolic rewiring to clear dying neighboring cells by efferocytosis during homeostasis and disease. Here we reveal that glutaminase-1-mediated glutaminolysis is critical to promote apoptotic cell clearance by macrophages during homeostasis in mice. In addition, impaired macrophage glutaminolysis exacerbates atherosclerosis, a condition during which, efficient apoptotic cell debris clearance is critical to limit disease progression. Glutaminase-1 expression strongly correlates with atherosclerotic plaque necrosis in patients with cardiovascular diseases. High-throughput transcriptional and metabolic profiling reveals that macrophage efferocytic capacity relies on a non-canonical transaminase pathway, independent from the traditional requirement of glutamate dehydrogenase to fuel ɑ-ketoglutarate-dependent immunometabolism. This pathway is necessary to meet the unique requirements of efferocytosis for cellular detoxification and high-energy cytoskeletal rearrangements. Thus, we uncover a role for non-canonical glutamine metabolism for efficient clearance of dying cells and maintenance of tissue homeostasis during health and disease in mouse and humans.
Collapse
Affiliation(s)
- Johanna Merlin
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Stoyan Ivanov
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Adélie Dumont
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | | | - Julie Gall
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Marion Stunault
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Marion Ayrault
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Nathalie Vaillant
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Alexia Castiglione
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Amanda Swain
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Francois Orange
- Université Côte d'Azur, Centre Commun de Microscopie Appliquée (CCMA), Nice, France
| | - Alexandre Gallerand
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Thierry Berton
- Centre de Recherche Cardiovasculaire et Nutritionnelle (C2VN), INSERM, Institut National de la Recherche Agricole (INRA), BioMet, Aix-Marseille University, Marseille, France
| | - Jean-Charles Martin
- Centre de Recherche Cardiovasculaire et Nutritionnelle (C2VN), INSERM, Institut National de la Recherche Agricole (INRA), BioMet, Aix-Marseille University, Marseille, France
| | - Stefania Carobbio
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
- Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Justine Masson
- Inserm UMR-S1270, Institut du Fer à Moulin, Sorbonne Université, Paris, France
- Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY, USA
| | - Inna Gaisler-Salomon
- Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
- SPC-IBBR, University of Haifa, Haifa, Israel
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
| | - Stephen Rayport
- Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - Judith C Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
- Institute for Molecular Cardiovascular Research, RWTH Klinikum Aachen, Aachen, Germany
| | - Rodolphe R Guinamard
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | | | - Edward B Thorp
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France.
| |
Collapse
|
69
|
Márquez AB, van der Vorst EPC, Maas SL. Key Chemokine Pathways in Atherosclerosis and Their Therapeutic Potential. J Clin Med 2021; 10:3825. [PMID: 34501271 PMCID: PMC8432216 DOI: 10.3390/jcm10173825] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/20/2021] [Accepted: 08/20/2021] [Indexed: 12/24/2022] Open
Abstract
The search to improve therapies to prevent or treat cardiovascular diseases (CVDs) rages on, as CVDs remain a leading cause of death worldwide. Here, the main cause of CVDs, atherosclerosis, and its prevention, take center stage. Chemokines and their receptors have long been known to play an important role in the pathophysiological development of atherosclerosis. Their role extends from the initiation to the progression, and even the potential regression of atherosclerotic lesions. These important regulators in atherosclerosis are therefore an obvious target in the development of therapeutic strategies. A plethora of preclinical studies have assessed various possibilities for targeting chemokine signaling via various approaches, including competitive ligands and microRNAs, which have shown promising results in ameliorating atherosclerosis. Developments in the field also include detailed imaging with tracers that target specific chemokine receptors. Lastly, clinical trials revealed the potential of various therapies but still require further investigation before commencing clinical use. Although there is still a lot to be learned and investigated, it is clear that chemokines and their receptors present attractive yet extremely complex therapeutic targets. Therefore, this review will serve to provide a general overview of the connection between various chemokines and their receptors with atherosclerosis. The different developments, including mouse models and clinical trials that tackle this complex interplay will also be explored.
Collapse
Affiliation(s)
- Andrea Bonnin Márquez
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Sanne L. Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
70
|
Rodriguez AB, Peske JD, Woods AN, Leick KM, Mauldin IS, Meneveau MO, Young SJ, Lindsay RS, Melssen MM, Cyranowski S, Parriott G, Conaway MR, Fu YX, Slingluff CL, Engelhard VH. Immune mechanisms orchestrate tertiary lymphoid structures in tumors via cancer-associated fibroblasts. Cell Rep 2021; 36:109422. [PMID: 34289373 PMCID: PMC8362934 DOI: 10.1016/j.celrep.2021.109422] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 02/26/2021] [Accepted: 06/28/2021] [Indexed: 12/21/2022] Open
Abstract
Tumor-associated tertiary lymphoid structures (TA-TLS) are associated with enhanced patient survival and responsiveness to cancer therapies, but the mechanisms underlying their development are unknown. We show here that TA-TLS development in murine melanoma is orchestrated by cancer-associated fibroblasts (CAF) with characteristics of lymphoid tissue organizer cells that are induced by tumor necrosis factor receptor signaling. CAF organization into reticular networks is mediated by CD8 T cells, while CAF accumulation and TA-TLS expansion depend on CXCL13-mediated recruitment of B cells expressing lymphotoxin-α1β2. Some of these elements are also overrepresented in human TA-TLS. Additionally, we demonstrate that immunotherapy induces more and larger TA-TLS that are more often organized with discrete T and B cell zones, and that TA-TLS presence, number, and size are correlated with reduced tumor size and overall response to checkpoint immunotherapy. This work provides a platform for manipulating TA-TLS development as a cancer immunotherapy strategy.
Collapse
Affiliation(s)
- Anthony B Rodriguez
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - J David Peske
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Amber N Woods
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Katie M Leick
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Ileana S Mauldin
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Max O Meneveau
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Samuel J Young
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Robin S Lindsay
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Marit M Melssen
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Salwador Cyranowski
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Geoffrey Parriott
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Mark R Conaway
- Division of Translational Research & Applied Statistics, Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Craig L Slingluff
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Victor H Engelhard
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| |
Collapse
|
71
|
Dieudé M, Kaci I, Hébert MJ. The Impact of Programmed Cell Death on the Formation of Tertiary Lymphoid Structures. Front Immunol 2021; 12:696311. [PMID: 34335608 PMCID: PMC8320843 DOI: 10.3389/fimmu.2021.696311] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/28/2021] [Indexed: 01/14/2023] Open
Abstract
Tertiary lymphoid structures are clusters of lymphoid tissue that develop post-natally at sites of chronic inflammation. They have been described in association with infection, autoimmune disorders, cancer, and allograft rejection. In their mature stage, TLS function as ectopic germinal centers, favoring the local production of autoantibodies and cytokines. TLS formation tends to parallel the severity of tissue injury and they are usually indicative of locally active immune responses. The presence of TLS in patients with solid tumors is usually associated with a better prognosis whereas their presence predicts increased maladaptive immunologic activity in patients with autoimmune disorders or allograft transplantation. Recent data highlight a correlation between active cell death and TLS formation and maturation. Our group recently identified apoptotic exosome-like vesicles, released by apoptotic cells, as novel inducers of TLS formation. Here, we review mechanisms of TLS formation and maturation with a specific focus on the emerging importance of tissue injury, programmed cell death and extracellular vesicles in TLS biogenesis.
Collapse
Affiliation(s)
- Mélanie Dieudé
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.,Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.,Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
| | - Imane Kaci
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.,Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Molecular Biology Programs, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Marie-Josée Hébert
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.,Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
72
|
Abstract
Atherosclerosis is the leading cause of acute cardiovascular events, and vascular calcification is an important pathological phenomenon in atherosclerosis. Recently, many studies have shown that immune cells are closely associated with the development of atherosclerosis and calcification, but there are many conflicting viewpoints because of immune system complications, such as the pro-atherosclerotic and atheroprotective effects of regulatory B cells (Bregs), T helper type 2 (Th2) cells and T helper type 17 (Th17) cells. In this review, we summarize the studies on the roles of immune cells, especially lymphocytes and macrophages, in atherosclerotic calcification. Furthermore, we prepared graphs showing the relationship between T cells, B cells and macrophages and atherosclerotic calcification. Finally, we highlight some potential issues that are closely associated with the function of immune cells in atherosclerotic calcification. Based on current research results, this review summarizes the relationship between immune cells and atherosclerotic calcification, and it will be beneficial to understand the relationship of immune cells and atherosclerotic calcification.
Collapse
Affiliation(s)
- Jingsong Cao
- Clinical Medicine Research Center, 574417The First Affiliated Hospital of University of South China, Hengyang, China.,Department of Endocrinology and Metabolism, 574417The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xuyu Zu
- Clinical Medicine Research Center, 574417The First Affiliated Hospital of University of South China, Hengyang, China
| | - Jianghua Liu
- Clinical Medicine Research Center, 574417The First Affiliated Hospital of University of South China, Hengyang, China.,Department of Endocrinology and Metabolism, 574417The First Affiliated Hospital of University of South China, Hengyang, Hunan, China.,Department of Metabolism and Endocrinology, 574417The First Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
73
|
Zhao Y, Zhang J, Zhang W, Xu Y. A myriad of roles of dendritic cells in atherosclerosis. Clin Exp Immunol 2021; 206:12-27. [PMID: 34109619 DOI: 10.1111/cei.13634] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/17/2021] [Accepted: 05/28/2021] [Indexed: 11/29/2022] Open
Abstract
Atherosclerosis is an inflammatory disease with break-down of homeostatic immune regulation of vascular tissues. As a critical initiator of host immunity, dendritic cells (DCs) have also been identified in the aorta of healthy individuals and atherosclerotic patients, whose roles in regulating arterial inflammation aroused great interest. Accumulating evidence has now pointed to the fundamental roles for DCs in every developmental stage of atherosclerosis due to their myriad of functions in immunity and tolerance induction, ranging from lipid uptake, efferocytosis and antigen presentation to pro- and anti-inflammatory cytokine or chemokine secretion. In this study we provide a timely summary of the published works in this field, and comprehensively discuss both the direct and indirect roles of DCs in atherogenesis. Understanding the pathogenic roles of DCs during the development of atherosclerosis in vascular tissues would certainly help to open therapeutic avenue to the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yanfang Zhao
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, China
| | - Jing Zhang
- Department of Thoracic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenjie Zhang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, China
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, China
| |
Collapse
|
74
|
Singh H, Rai V, Agrawal DK. Discerning the promising binding sites of S100/calgranulins and their therapeutic potential in atherosclerosis. Expert Opin Ther Pat 2021; 31:1045-1057. [PMID: 34056993 DOI: 10.1080/13543776.2021.1937122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Atherosclerosis is a chronic inflammatory disease in which the members of S100 family proteins (calgranulins) bind with their receptors, particularly receptor for advanced glycation end products (RAGE) and toll-like receptor-4 (TLR-4) and play a key role in the pathogenesis and progression of disease. Thus, these proteins could be considered as potential biomarkers and therapeutic targets in the treatment of atherosclerotic inflammation. AREAS COVERED This review summarizes the pathology of S100A8, S100A9, and S100A12 in the development of atherosclerosis and reveals key structural features of these proteins which are potentially critical in their pathological effects. This article focuses on the translational significance of antagonizing these proteins by using small molecules in patent literature, clinical and preclinical studies and also discusses future approaches that could be employed to block these proteins in the treatment of atherosclerosis. EXPERT OPINION Based on the critical role of S100/calgranulins in the regulation of atherosclerosis, these proteins are potential targets to develop better therapeutic options in the treatment of inflammatory diseases. However, further research is still needed to clarify their exact molecular mechanism by analyzing their detailed structural features that can expedite future research to develop novel therapeutics against these proteins to treat atherosclerotic inflammation.
Collapse
Affiliation(s)
- Harbinder Singh
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
75
|
Yeo KP, Lim HY, Angeli V. Leukocyte Trafficking via Lymphatic Vessels in Atherosclerosis. Cells 2021; 10:cells10061344. [PMID: 34072313 PMCID: PMC8229118 DOI: 10.3390/cells10061344] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 02/03/2023] Open
Abstract
In recent years, lymphatic vessels have received increasing attention and our understanding of their development and functional roles in health and diseases has greatly improved. It has become clear that lymphatic vessels are critically involved in acute and chronic inflammation and its resolution by supporting the transport of immune cells, fluid, and macromolecules. As we will discuss in this review, the involvement of lymphatic vessels has been uncovered in atherosclerosis, a chronic inflammatory disease of medium- and large-sized arteries causing deadly cardiovascular complications worldwide. The progression of atherosclerosis is associated with morphological and functional alterations in lymphatic vessels draining the diseased artery. These defects in the lymphatic vasculature impact the inflammatory response in atherosclerosis by affecting immune cell trafficking, lymphoid neogenesis, and clearance of macromolecules in the arterial wall. Based on these new findings, we propose that targeting lymphatic function could be considered in conjunction with existing drugs as a treatment option for atherosclerosis.
Collapse
|
76
|
Johansson-Percival A, Ganss R. Therapeutic Induction of Tertiary Lymphoid Structures in Cancer Through Stromal Remodeling. Front Immunol 2021; 12:674375. [PMID: 34122434 PMCID: PMC8191417 DOI: 10.3389/fimmu.2021.674375] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/04/2021] [Indexed: 01/01/2023] Open
Abstract
Improving the effectiveness of anti-cancer immunotherapy remains a major clinical challenge. Cytotoxic T cell infiltration is crucial for immune-mediated tumor rejection, however, the suppressive tumor microenvironment impedes their recruitment, activation, maturation and function. Nevertheless, solid tumors can harbor specialized lymph node vasculature and immune cell clusters that are organized into tertiary lymphoid structures (TLS). These TLS support naïve T cell infiltration and intratumoral priming. In many human cancers, their presence is a positive prognostic factor, and importantly, predictive for responsiveness to immune checkpoint blockade. Thus, therapeutic induction of TLS is an attractive concept to boost anti-cancer immunotherapy. However, our understanding of how cancer-associated TLS could be initiated is rudimentary. Exciting new reagents which induce TLS in preclinical cancer models provide mechanistic insights into the exquisite stromal orchestration of TLS formation, a process often associated with a more functional or "normalized" tumor vasculature and fueled by LIGHT/LTα/LTβ, TNFα and CC/CXC chemokine signaling. These emerging insights provide innovative opportunities to induce and shape TLS in the tumor microenvironment to improve immunotherapies.
Collapse
Affiliation(s)
- Anna Johansson-Percival
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia
- Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia
| | - Ruth Ganss
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia
- Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
77
|
Aoyama S, Nakagawa R, Mulé JJ, Mailloux AW. Inducible Tertiary Lymphoid Structures: Promise and Challenges for Translating a New Class of Immunotherapy. Front Immunol 2021; 12:675538. [PMID: 34054863 PMCID: PMC8160316 DOI: 10.3389/fimmu.2021.675538] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are ectopically formed aggregates of organized lymphocytes and antigen-presenting cells that occur in solid tissues as part of a chronic inflammation response. Sharing structural and functional characteristics with conventional secondary lymphoid organs (SLO) including discrete T cell zones, B cell zones, marginal zones with antigen presenting cells, reticular stromal networks, and high endothelial venues (HEV), TLS are prominent centers of antigen presentation and adaptive immune activation within the periphery. TLS share many signaling axes and leukocyte recruitment schemes with SLO regarding their formation and function. In cancer, their presence confers positive prognostic value across a wide spectrum of indications, spurring interest in their artificial induction as either a new form of immunotherapy, or as a means to augment other cell or immunotherapies. Here, we review approaches for inducible (iTLS) that utilize chemokines, inflammatory factors, or cellular analogues vital to TLS formation and that often mirror conventional SLO organogenesis. This review also addresses biomaterials that have been or might be suitable for iTLS, and discusses remaining challenges facing iTLS manufacturing approaches for clinical translation.
Collapse
Affiliation(s)
- Shota Aoyama
- Department of Surgery, Institute of Gastroenterology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Ryosuke Nakagawa
- Department of Surgery, Institute of Gastroenterology, Tokyo Women’s Medical University, Tokyo, Japan
| | - James J. Mulé
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
- Cutaneous Oncology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Adam W. Mailloux
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
78
|
Gago da Graça C, van Baarsen LGM, Mebius RE. Tertiary Lymphoid Structures: Diversity in Their Development, Composition, and Role. THE JOURNAL OF IMMUNOLOGY 2021; 206:273-281. [PMID: 33397741 DOI: 10.4049/jimmunol.2000873] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Lymph node stromal cells coordinate the adaptive immune response in secondary lymphoid organs, providing both a structural matrix and soluble factors that regulate survival and migration of immune cells, ultimately promoting Ag encounter. In several inflamed tissues, resident fibroblasts can acquire lymphoid-stroma properties and drive the formation of ectopic aggregates of immune cells, named tertiary lymphoid structures (TLSs). Mature TLSs are functional sites for the development of adaptive responses and, consequently, when present, can have an impact in both autoimmunity and cancer conditions. In this review, we go over recent findings concerning both lymph node stromal cells and TLSs function and formation and further describe what is currently known about their role in disease, particularly their potential in tolerance.
Collapse
Affiliation(s)
- Catarina Gago da Graça
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, Vrije Universiteit, 1081HZ Amsterdam, the Netherlands
| | - Lisa G M van Baarsen
- Department of Rheumatology and Clinical Immunology, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands.,Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, the Netherlands; and.,Amsterdam Rheumatology and Immunology Center, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, Vrije Universiteit, 1081HZ Amsterdam, the Netherlands;
| |
Collapse
|
79
|
Blanchard L, Girard JP. High endothelial venules (HEVs) in immunity, inflammation and cancer. Angiogenesis 2021; 24:719-753. [PMID: 33956259 PMCID: PMC8487881 DOI: 10.1007/s10456-021-09792-8] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022]
Abstract
High endothelial venules (HEVs) are specialized blood vessels mediating lymphocyte trafficking to lymph nodes (LNs) and other secondary lymphoid organs. By supporting high levels of lymphocyte extravasation from the blood, HEVs play an essential role in lymphocyte recirculation and immune surveillance for foreign invaders (bacterial and viral infections) and alterations in the body’s own cells (neoantigens in cancer). The HEV network expands during inflammation in immune-stimulated LNs and is profoundly remodeled in metastatic and tumor-draining LNs. HEV-like blood vessels expressing high levels of the HEV-specific sulfated MECA-79 antigens are induced in non-lymphoid tissues at sites of chronic inflammation in many human inflammatory and allergic diseases, including rheumatoid arthritis, Crohn’s disease, allergic rhinitis and asthma. Such vessels are believed to contribute to the amplification and maintenance of chronic inflammation. MECA-79+ tumor-associated HEVs (TA-HEVs) are frequently found in human tumors in CD3+ T cell-rich areas or CD20+ B-cell rich tertiary lymphoid structures (TLSs). TA-HEVs have been proposed to play important roles in lymphocyte entry into tumors, a process essential for successful antitumor immunity and lymphocyte-mediated cancer immunotherapy with immune checkpoint inhibitors, vaccines or adoptive T cell therapy. In this review, we highlight the phenotype and function of HEVs in homeostatic, inflamed and tumor-draining lymph nodes, and those of HEV-like blood vessels in chronic inflammatory diseases. Furthermore, we discuss the role and regulation of TA-HEVs in human cancer and mouse tumor models.
Collapse
Affiliation(s)
- Lucas Blanchard
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
80
|
Salem D, Chelvanambi M, Storkus WJ, Fecek RJ. Cutaneous Melanoma: Mutational Status and Potential Links to Tertiary Lymphoid Structure Formation. Front Immunol 2021; 12:629519. [PMID: 33746966 PMCID: PMC7970117 DOI: 10.3389/fimmu.2021.629519] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 02/11/2021] [Indexed: 12/21/2022] Open
Abstract
Recent advances in immunotherapy have enabled rapid evolution of novel interventional approaches designed to reinvigorate and expand patient immune responses against cancer. An emerging approach in cancer immunology involves the conditional induction of tertiary lymphoid structures (TLS), which are non-encapsulated ectopic lymphoid structures forming at sites of chronic, pathologic inflammation. Cutaneous melanoma (CM), a highly-immunogenic form of solid cancer, continues to rise in both incidence and mortality rate, with recent reports supporting a positive correlation between the presence of TLS in melanoma and beneficial treatment outcomes amongst advanced-stage patients. In this context, TLS in CM are postulated to serve as dynamic centers for the initiation of robust anti-tumor responses within affected regions of active disease. Given their potential importance to patient outcome, significant effort has been recently devoted to gaining a better understanding of TLS neogenesis and the influence these lymphoid organs exert within the tumor microenvironment. Here, we briefly review TLS structure, function, and response to treatment in the setting of CM. To uncover potential tumor-intrinsic mechanisms that regulate TLS formation, we have taken the novel perspective of evaluating TLS induction in melanomas impacted by common driver mutations in BRAF, PTEN, NRAS, KIT, PRDM1, and MITF. Through analysis of The Cancer Genome Atlas (TCGA), we show expression of DNA repair proteins (DRPs) including BRCA1, PAXIP, ERCC1, ERCC2, ERCC3, MSH2, and PMS2 to be negatively correlated with expression of pro-TLS genes, suggesting DRP loss may favor TLS development in support of improved patient outcome and patient response to interventional immunotherapy.
Collapse
Affiliation(s)
- Deepak Salem
- Department of Microbiology, Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA, United States
| | - Manoj Chelvanambi
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ronald J Fecek
- Department of Microbiology, Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA, United States
| |
Collapse
|
81
|
Li W, Gauthier JM, Tong AY, Terada Y, Higashikubo R, Frye CC, Harrison MS, Hashimoto K, Bery AI, Ritter JH, Nava RG, Puri V, Wong BW, Lavine KJ, Bharat A, Krupnick AS, Gelman AE, Kreisel D. Lymphatic drainage from bronchus-associated lymphoid tissue in tolerant lung allografts promotes peripheral tolerance. J Clin Invest 2021; 130:6718-6727. [PMID: 33196461 DOI: 10.1172/jci136057] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 09/03/2020] [Indexed: 12/29/2022] Open
Abstract
Tertiary lymphoid organs are aggregates of immune and stromal cells including high endothelial venules and lymphatic vessels that resemble secondary lymphoid organs and can be induced at nonlymphoid sites during inflammation. The function of lymphatic vessels within tertiary lymphoid organs remains poorly understood. During lung transplant tolerance, Foxp3+ cells accumulate in tertiary lymphoid organs that are induced within the pulmonary grafts and are critical for the local downregulation of alloimmune responses. Here, we showed that tolerant lung allografts could induce and maintain tolerance of heterotopic donor-matched hearts through pathways that were dependent on the continued presence of the transplanted lung. Using lung retransplantation, we showed that Foxp3+ cells egressed from tolerant lung allografts via lymphatics and were recruited into donor-matched heart allografts. Indeed, survival of the heart allografts was dependent on lymphatic drainage from the tolerant lung allograft to the periphery. Thus, our work indicates that cellular trafficking from tertiary lymphoid organs regulates immune responses in the periphery. We propose that these findings have important implications for a variety of disease processes that are associated with the induction of tertiary lymphoid organs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jon H Ritter
- Pathology & Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | | | | | | | - Ankit Bharat
- Department of Surgery, Northwestern University, Chicago, Illinois, USA
| | | | - Andrew E Gelman
- Departments of Surgery.,Pathology & Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Daniel Kreisel
- Departments of Surgery.,Pathology & Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
82
|
Busnelli M, Manzini S, Chiara M, Colombo A, Fontana F, Oleari R, Potì F, Horner D, Bellosta S, Chiesa G. Aortic Gene Expression Profiles Show How ApoA-I Levels Modulate Inflammation, Lysosomal Activity, and Sphingolipid Metabolism in Murine Atherosclerosis. Arterioscler Thromb Vasc Biol 2021; 41:651-667. [PMID: 33327742 PMCID: PMC7837693 DOI: 10.1161/atvbaha.120.315669] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 12/01/2020] [Indexed: 01/16/2023]
Abstract
OBJECTIVE HDL (high-density lipoprotein) particles are known to possess several antiatherogenic properties that include the removal of excess cholesterol from peripheral tissues, the maintenance of endothelial integrity, antioxidant, and anti-inflammatory activities. ApoA-I overexpression in apoE-deficient (EKO) mice has been shown to increase HDL levels and to strongly reduce atherosclerosis development. The aim of the study was to investigate gene expression patterns associated with atherosclerosis development in the aorta of EKO mice and how HDL plasma levels relate to gene expression patterns at different stages of atherosclerosis development and with different dietary treatments. Approach and Results: Eight-week-old EKO mice, EKO mice overexpressing human apoA-I, and wild-type mice as controls were fed either normal laboratory or Western diet for 6 or 22 weeks. Cholesterol distribution among lipoproteins was evaluated, and atherosclerosis of the aorta was quantified. High-throughput sequencing technologies were used to analyze the transcriptome of the aorta of the 3 genotypes in each experimental condition. In addition to the well-known activation of inflammation and immune response, the impairment of sphingolipid metabolism, phagosome-lysosome system, and osteoclast differentiation emerged as relevant players in atherosclerosis development. The reduced atherosclerotic burden in the aorta of EKO mice expressing high levels of apoA-I was accompanied by a reduced activation of immune system markers, as well as reduced perturbation of lysosomal activity and a better regulation of the sphingolipid synthesis pathway. CONCLUSIONS ApoA-I modulates atherosclerosis development in the aorta of EKO mice affecting the expression of pathways additional to those associated with inflammation and immune response.
Collapse
Affiliation(s)
- Marco Busnelli
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., F.F., R.O., S.B., G.C.), Università degli Studi di Milano, Italy
| | - Stefano Manzini
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., F.F., R.O., S.B., G.C.), Università degli Studi di Milano, Italy
| | - Matteo Chiara
- Department of Biosciences (M.C., D.H.), Università degli Studi di Milano, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy (M.C., D.H.)
| | - Alice Colombo
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., F.F., R.O., S.B., G.C.), Università degli Studi di Milano, Italy
| | - Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., F.F., R.O., S.B., G.C.), Università degli Studi di Milano, Italy
| | - Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., F.F., R.O., S.B., G.C.), Università degli Studi di Milano, Italy
| | - Francesco Potì
- Department of Medicine and Surgery—Unit of Neurosciences, University of Parma, Italy (F.P.)
| | - David Horner
- Department of Biosciences (M.C., D.H.), Università degli Studi di Milano, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy (M.C., D.H.)
| | - Stefano Bellosta
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., F.F., R.O., S.B., G.C.), Università degli Studi di Milano, Italy
| | - Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., F.F., R.O., S.B., G.C.), Università degli Studi di Milano, Italy
| |
Collapse
|
83
|
Functional Role of B Cells in Atherosclerosis. Cells 2021; 10:cells10020270. [PMID: 33572939 PMCID: PMC7911276 DOI: 10.3390/cells10020270] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis is a lipid-driven inflammatory disease of blood vessels, and both innate and adaptive immune responses are involved in its development. The impact of B cells on atherosclerosis has been demonstrated in numerous studies and B cells have been found in close proximity to atherosclerotic plaques in humans and mice. B cells exert both atheroprotective and pro-atherogenic functions, which have been associated with their B cell subset attribution. While B1 cells and marginal zone B cells are considered to protect against atherosclerosis, follicular B cells and innate response activator B cells have been shown to promote atherosclerosis. In this review, we shed light on the role of B cells from a different, functional perspective and focus on the three major B cell functions: antibody production, antigen presentation/T cell interaction, and the release of cytokines. All of these functions have the potential to affect atherosclerosis by multiple ways and are dependent on the cellular milieu and the activation status of the B cell. Moreover, we discuss B cell receptor signaling and the mechanism of B cell activation under atherosclerosis-prone conditions. By summarizing current knowledge of B cells in and beyond atherosclerosis, we are pointing out open questions and enabling new perspectives.
Collapse
|
84
|
Mohmmad‐Rezaei M, Arefnezhad R, Ahmadi R, Abdollahpour‐Alitappeh M, Mirzaei Y, Arjmand M, Ferns GA, Bashash D, Bagheri N. An overview of the innate and adaptive immune system in atherosclerosis. IUBMB Life 2020. [DOI: 10.1002/iub.2425] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Mina Mohmmad‐Rezaei
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| | - Reza Arefnezhad
- Halal Research Center of IRI, FDA Tehran Iran
- Department of Anatomy, School of Medicine Shiraz University of Medical Sciences Shiraz Iran
| | - Reza Ahmadi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| | | | - Yousef Mirzaei
- Department of Biogeosciences, Scientific Research Center Soran University Soran Iraq
| | - Mohammad‐Hassan Arjmand
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
- Cancer Research Center Shahrekord University of Medical Sciences Shahrekord Iran
| | - Gordon A. Ferns
- Brighton & Sussex Medical School, Division of Medical Education Sussex United Kingdom
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| |
Collapse
|
85
|
Cohen M, Giladi A, Raposo C, Zada M, Li B, Ruckh J, Deczkowska A, Mohar B, Shechter R, Lichtenstein RG, Amit I, Schwartz M. Meningeal lymphoid structures are activated under acute and chronic spinal cord pathologies. Life Sci Alliance 2020; 4:4/1/e202000907. [PMID: 33277355 PMCID: PMC7723261 DOI: 10.26508/lsa.202000907] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/01/2022] Open
Abstract
We found that acute insult to the central nervous system induces the formation of lymphocyte aggregates reminiscent of tertiary lymphoid structures within the spinal cord meninges. Unlike draining CNS-cervical lymph nodes, meningeal lymphocytes are locally activated during neuro-inflammtion and neurodegeneration. Tertiary lymphoid structures (TLS) are organized aggregates of B and T cells formed ectopically during different stages of life in response to inflammation, infection, or cancer. Here, we describe formation of structures reminiscent of TLS in the spinal cord meninges under several central nervous system (CNS) pathologies. After acute spinal cord injury, B and T lymphocytes locally aggregate within the meninges to form TLS-like structures, and continue to accumulate during the late phase of the response to the injury, with a negative impact on subsequent pathological conditions, such as experimental autoimmune encephalomyelitis. Using a chronic model of spinal cord pathology, the mSOD1 mouse model of amyotrophic lateral sclerosis, we further showed by single-cell RNA-sequencing that a meningeal lymphocyte niche forms, with a unique organization and activation state, including accumulation of pre-B cells in the spinal cord meninges. Such a response was not found in the CNS-draining cervical lymph nodes. The present findings suggest that a special immune response develops in the meninges during various neurological pathologies in the CNS, a possible reflection of its immune privileged nature.
Collapse
Affiliation(s)
- Merav Cohen
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Amir Giladi
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Catarina Raposo
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Mor Zada
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Baoguo Li
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Julia Ruckh
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Boaz Mohar
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Ravid Shechter
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Rachel G Lichtenstein
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel .,Klarman Cell Observatory, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| |
Collapse
|
86
|
Steines L, Poth H, Herrmann M, Schuster A, Banas B, Bergler T. B Cell Activating Factor (BAFF) Is Required for the Development of Intra-Renal Tertiary Lymphoid Organs in Experimental Kidney Transplantation in Rats. Int J Mol Sci 2020; 21:ijms21218045. [PMID: 33126753 PMCID: PMC7662293 DOI: 10.3390/ijms21218045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/22/2022] Open
Abstract
Intra-renal tertiary lymphoid organs (TLOs) are associated with worsened outcome in kidney transplantation (Ktx). We used an anti-BAFF (B cell activating factor) intervention to investigate whether BAFF is required for TLO formation in a full MHC-mismatch Ktx model in rats. Rats received either therapeutic immunosuppression (no rejection, NR) or subtherapeutic immunosuppression (chronic rejection, CR) and were sacrificed on d56. One group additionally received an anti-BAFF antibody (CR + AB). Intra-renal T (CD3+) and B (CD20+) cells, their proliferation (Ki67+), and IgG+ plasma cells were analyzed by immunofluorescence microscopy. Formation of T and B cell zones and TLOs was assessed. Intra-renal expression of TLO-promoting factors, molecules of T:B crosstalk, and B cell differentiation was analyzed by qPCR. Intra-renal B and T cell zones and TLOs were detected in CR and were associated with elevated intra-renal mRNA expression of TLO-promoting factors, including CXCL13, CCL19, lymphotoxin-β, and BAFF. Intra-renal plasma cells were also elevated in CR. Anti-BAFF treatment significantly decreased intra-renal B cell zones and TLO, as well as intra-renal B cell-derived TLO-promoting factors and B cell differentiation markers. We conclude that BAFF-dependent intra-renal B cells promote TLO formation and advance local adaptive alloimmune responses in chronic rejection.
Collapse
Affiliation(s)
- Louisa Steines
- Correspondence: ; Tel.: +49-941-9447301; Fax: +49-941-9447302
| | | | | | | | | | | |
Collapse
|
87
|
MacRitchie N, Grassia G, Noonan J, Cole JE, Hughes CE, Schroeder J, Benson RA, Cochain C, Zernecke A, Guzik TJ, Garside P, Monaco C, Maffia P. The aorta can act as a site of naïve CD4+ T-cell priming. Cardiovasc Res 2020; 116:306-316. [PMID: 30980670 DOI: 10.1093/cvr/cvz102] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 02/20/2019] [Accepted: 04/09/2019] [Indexed: 12/18/2022] Open
Abstract
AIMS Aortic adaptive immunity plays a role in atherosclerosis; however, the precise mechanisms leading to T-cell activation in the arterial wall remain poorly understood. METHODS AND RESULTS Here, we have identified naïve T cells in the aorta of wild-type and T-cell receptor transgenic mice and we demonstrate that naïve T cells can be primed directly in the vessel wall with both kinetics and frequency of T-cell activation found to be similar to splenic and lymphoid T cells. Aortic homing of naïve T cells is regulated at least in part by the P-selectin glycosylated ligand-1 receptor. In experimental atherosclerosis the aorta supports CD4+ T-cell activation selectively driving Th1 polarization. By contrast, secondary lymphoid organs display Treg expansion. CONCLUSION Our results demonstrate that the aorta can support T-cell priming and that naïve T cells traffic between the circulation and vessel wall. These data underpin the paradigm that local priming of T cells specific for plaque antigens contributes to atherosclerosis progression.
Collapse
Affiliation(s)
- Neil MacRitchie
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Gianluca Grassia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Jonathan Noonan
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Jennifer E Cole
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Catherine E Hughes
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Juliane Schroeder
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Robert A Benson
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Clement Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK.,Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Kraków, Poland
| | - Paul Garside
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Claudia Monaco
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK.,Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK.,Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
88
|
Clément M, Lareyre F, Loste A, Sannier A, Burel-Vandenbos F, Massiot N, Carboni J, Jean-Baptiste E, Caligiuri G, Nicoletti A, Raffort J. Vascular Remodeling and Immune Cell Infiltration in Splenic Artery Aneurysms. Angiology 2020; 72:539-549. [PMID: 32851875 DOI: 10.1177/0003319720952290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rupture of splenic artery aneurysms (SAAs) is associated with a high mortality rate. The aim of this study was to identify the features of SAAs. Tissue sections from SAAs were compared to nonaneurysmal splenic arteries using various stains. The presence of intraluminal thrombus (ILT), vascular smooth muscle cells (VSMCs), cluster of differentiation (CD)-68+ phagocytes, myeloperoxidase+ neutrophils, CD3+, and CD20+ adaptive immune cells were studied using immunofluorescence microscopy. Analysis of SAAs revealed the presence of atherosclerotic lesions, calcifications, and ILT. Splenic artery aneurysms were characterized by a profound vascular remodeling with a dramatic loss of VSMCs, elastin degradation, adventitial fibrosis associated with enhanced apoptosis, and increased matrix metalloproteinase 9 expression. We observed an infiltration of immune cells comprising macrophages, neutrophils, T, and B cells. The T and B cells were found in the adventitial layer of SAAs, but their organization into tertiary lymphoid organs was halted. We failed to detect germinal centers even in the most organized T/B cell follicles and these lymphoid clusters lacked lymphoid stromal cells. This detailed histopathological characterization of the vascular remodeling during SAA showed that lymphoid neogenesis was incomplete, suggesting that critical mediators of their development must be missing.
Collapse
Affiliation(s)
- Marc Clément
- Université de Paris, LVTS, 121283INSERM U1148, Paris, France
| | - Fabien Lareyre
- Department of Vascular Surgery, 26992University Hospital of Nice, France.,Department of Vascular Surgery, University Hospital of Antibes-Juan-les-Pins, France.,439710Université Côte d'Azur, CHU, INSERM U1065, C3M, Nice, France
| | - Alexia Loste
- Université de Paris, LVTS, 121283INSERM U1148, Paris, France
| | - Aurélie Sannier
- Université de Paris, LVTS, 121283INSERM U1148, Paris, France
| | | | - Nicolas Massiot
- Department of Vascular Surgery, 26992University Hospital of Nice, France
| | - Joseph Carboni
- Department of Vascular Surgery, 26992University Hospital of Nice, France
| | - Elixène Jean-Baptiste
- Department of Vascular Surgery, 26992University Hospital of Nice, France.,439710Université Côte d'Azur, CHU, INSERM U1065, C3M, Nice, France
| | | | | | - Juliette Raffort
- 439710Université Côte d'Azur, CHU, INSERM U1065, C3M, Nice, France.,Clinical Chemistry Laboratory, 121283University Hospital of Nice, France
| |
Collapse
|
89
|
Chen L, Ishigami T, Doi H, Arakawa K, Tamura K. Gut microbiota and atherosclerosis: role of B cell for atherosclerosis focusing on the gut-immune-B2 cell axis. J Mol Med (Berl) 2020; 98:1235-1244. [PMID: 32737524 PMCID: PMC7447622 DOI: 10.1007/s00109-020-01936-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 05/13/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023]
Abstract
Atherosclerosis is the leading cause of cardiovascular mortality and morbidity worldwide and is described as a complex disease involving several different cell types and their molecular products. Recent studies have revealed that atherosclerosis arises from a systemic inflammatory process, including the accumulation and activities of various immune cells. However, the immune system is a complicated network made up of many cell types, hundreds of bioactive cytokines, and millions of different antigens, making it challenging to readily define the associated mechanism of atherosclerosis. Nevertheless, we previously reported a potential persistent inflammatory process underlying atherosclerosis development, centered on a pathological humoral immune response between commensal microbes and activated subpopulations of substantial B cells in the vicinity of the arterial adventitia. Accumulating evidence has indicated the importance of gut microbiota in atherosclerosis development. Commensal microbiota are considered important regulators of immunity and metabolism and also to be possible antigenic sources for atherosclerosis development. However, the interplay between gut microbiota and metabolism with regard to the modulation of atherosclerosis-associated immune responses remains poorly understood. Here, we review the mechanisms by which the gut microbiota may influence atherogenesis, with particular focus on humoral immunity and B cells, especially the gut-immune-B2 cell axis. Under high-fat and high-calorie conditions, signals driven by the intestinal microbiota via the TLR signaling pathway cause B2 cells in the spleen to become functionally active and activated B2 cells then modify responses such as antibody production (generation of active antibodies IgG and IgG3), thereby contributing to the development of atherosclerosis. On the other hand, intestinal microbiota also resulted in recruitment and ectopic activation of B2 cells via the TLR signaling pathway in perivascular adipose tissue (PVAT), and, subsequently, an increase in circulating IgG and IgG3 led to the enhanced disease development. This is a potential link between microbiota alterations and B cells in the context of atherosclerosis. ![]()
Collapse
Affiliation(s)
- Lin Chen
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan.,Department of Cardiology, Sir Run Run Hospital, Nanjing Medical University, Long Mian Avenue 109 Jiangning, Nanjing, Jiangsu, China
| | - Tomoaki Ishigami
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan.
| | - Hiroshi Doi
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Kentaro Arakawa
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-ku, Yokohama, Kanagawa, Japan
| |
Collapse
|
90
|
Zernecke A, Winkels H, Cochain C, Williams JW, Wolf D, Soehnlein O, Robbins CS, Monaco C, Park I, McNamara CA, Binder CJ, Cybulsky MI, Scipione CA, Hedrick CC, Galkina EV, Kyaw T, Ghosheh Y, Dinh HQ, Ley K. Meta-Analysis of Leukocyte Diversity in Atherosclerotic Mouse Aortas. Circ Res 2020; 127:402-426. [PMID: 32673538 PMCID: PMC7371244 DOI: 10.1161/circresaha.120.316903] [Citation(s) in RCA: 231] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The diverse leukocyte infiltrate in atherosclerotic mouse aortas was recently analyzed in 9 single-cell RNA sequencing and 2 mass cytometry studies. In a comprehensive meta-analysis, we confirm 4 known macrophage subsets-resident, inflammatory, interferon-inducible cell, and Trem2 (triggering receptor expressed on myeloid cells-2) foamy macrophages-and identify a new macrophage subset resembling cavity macrophages. We also find that monocytes, neutrophils, dendritic cells, natural killer cells, innate lymphoid cells-2, and CD (cluster of differentiation)-8 T cells form prominent and separate immune cell populations in atherosclerotic aortas. Many CD4 T cells express IL (interleukin)-17 and the chemokine receptor CXCR (C-X-C chemokine receptor)-6. A small number of regulatory T cells and T helper 1 cells is also identified. Immature and naive T cells are present in both healthy and atherosclerotic aortas. Our meta-analysis overcomes limitations of individual studies that, because of their experimental approach, over- or underrepresent certain cell populations. Mass cytometry studies demonstrate that cell surface phenotype provides valuable information beyond the cell transcriptomes. The present analysis helps resolve some long-standing controversies in the field. First, Trem2+ foamy macrophages are not proinflammatory but interferon-inducible cell and inflammatory macrophages are. Second, about half of all foam cells are smooth muscle cell-derived, retaining smooth muscle cell transcripts rather than transdifferentiating to macrophages. Third, Pf4, which had been considered specific for platelets and megakaryocytes, is also prominently expressed in the main population of resident vascular macrophages. Fourth, a new type of resident macrophage shares transcripts with cavity macrophages. Finally, the discovery of a prominent innate lymphoid cell-2 cluster links the single-cell RNA sequencing work to recent flow cytometry data suggesting a strong atheroprotective role of innate lymphoid cells-2. This resolves apparent discrepancies regarding the role of T helper 2 cells in atherosclerosis based on studies that predated the discovery of innate lymphoid cells-2 cells.
Collapse
Affiliation(s)
- Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Holger Winkels
- Heart Center, University Hospital Cologne, Cologne, Germany
- Clinic III for Internal Medicine, Department of Cardiology, University of Cologne, Cologne, Germany
| | - Clément Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Würzburg, Wüzburg, Germany
| | - Jesse W. Williams
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN USA
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN USA
| | - Dennis Wolf
- Department of Cardiology and Angiology I, University Heart Center, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), Klinikum LMU Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Physiology and Pharmacology (FyFa), Karolinska Institute, Stockholm, Sweden
| | - Clint S. Robbins
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S1A1, Canada
- Department of Immunology, University of Toronto, Toronto, ON M5S1A1, Canada
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, Toronto, ON M5G1L7, Canada
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Inhye Park
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Coleen A. McNamara
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, USA
- Division of Cardioascular Medicine, University of Virginia School of Medicine, Charlottesville, USA
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Myron I. Cybulsky
- Toronto General Research Institute, University Health Network, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Corey A. Scipione
- Toronto General Research Institute, University Health Network, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Elena V. Galkina
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Road, Norfolk, VA USA
| | - Tin Kyaw
- Vascular Biology and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Centre for Inflammatory Diseases, Department of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | | | - Huy Q. Dinh
- La Jolla Institute for Immunology, La Jolla, CA USA
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA USA
- Department of Bioengineering, University of California San Diego, CA, USA
| |
Collapse
|
91
|
Marinkovic T, Marinkovic D. Biological mechanisms of ectopic lymphoid structure formation and their pathophysiological significance. Int Rev Immunol 2020; 40:255-267. [PMID: 32631119 DOI: 10.1080/08830185.2020.1789620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Ectopic lymphoid structures (ELS) or tertiary lymphoid organs are structures with the organization similar to the one of secondary lymphoid organs, formed in non-lymphoid tissues. They are considered to be an important site for the lymphocytic physiological and pathological role in conditions such are chronic infections, autoimmune diseases, cancer, and allograft rejection. Although similar to the secondary lymphoid tissues, the initiation of ELS formation is not preprogramed and requires chronic inflammation, expression of homeostatic chemokines, and lymphotoxin beta receptor activation. Importantly, while ELS formation may be considered beneficiary in antimicrobial and antitumor immunity, the persistence of these active lymphoid structures within the tissue increase the chance for development of autoimmunity and lymphoma. This paper is providing an overview of biological mechanisms involved in ELS formation, as well as the overview of the pathophysiological role of these structures. In addition, the paper discusses the possibility to therapeutically target ELS formation, bearing in mind their bivalent nature and role in different pathophysiological conditions.
Collapse
Affiliation(s)
- Tatjana Marinkovic
- Department of Medical Sciences, Western Serbia Academy of Applied Sciences, Uzice, Serbia
| | - Dragan Marinkovic
- Faculty of Special Education and Rehabilitation, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
92
|
Man AWC, Zhou Y, Xia N, Li H. Perivascular Adipose Tissue as a Target for Antioxidant Therapy for Cardiovascular Complications. Antioxidants (Basel) 2020; 9:E574. [PMID: 32630640 PMCID: PMC7402161 DOI: 10.3390/antiox9070574] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/23/2020] [Accepted: 06/27/2020] [Indexed: 12/12/2022] Open
Abstract
Perivascular adipose tissue (PVAT) is the connective tissue surrounding most of the systemic blood vessels. PVAT is now recognized as an important endocrine tissue that maintains vascular homeostasis. Healthy PVAT has anticontractile, anti-inflammatory, and antioxidative roles. Vascular oxidative stress is an important pathophysiological event in cardiometabolic complications of obesity, type 2 diabetes, and hypertension. Accumulating data from both humans and experimental animal models suggests that PVAT dysfunction is potentially linked to cardiovascular diseases, and associated with augmented vascular inflammation, oxidative stress, and arterial remodeling. Reactive oxygen species produced from PVAT can be originated from mitochondria, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, and uncoupled endothelial nitric oxide synthase. PVAT can also sense vascular paracrine signals and response by secreting vasoactive adipokines. Therefore, PVAT may constitute a novel therapeutic target for the prevention and treatment of cardiovascular diseases. In this review, we summarize recent findings on PVAT functions, ROS production, and oxidative stress in different pathophysiological settings and discuss the potential antioxidant therapies for cardiovascular diseases by targeting PVAT.
Collapse
Affiliation(s)
| | | | | | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, 55131 Mainz, Germany; (A.W.C.M.); (Y.Z.); (N.X.)
| |
Collapse
|
93
|
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall and the primary underlying cause of cardiovascular disease. Data from in vivo imaging, cell-lineage tracing and knockout studies in mice, as well as clinical interventional studies and advanced mRNA sequencing techniques, have drawn attention to the role of T cells as critical drivers and modifiers of the pathogenesis of atherosclerosis. CD4+ T cells are commonly found in atherosclerotic plaques. A large body of evidence indicates that T helper 1 (TH1) cells have pro-atherogenic roles and regulatory T (Treg) cells have anti-atherogenic roles. However, Treg cells can become pro-atherogenic. The roles in atherosclerosis of other TH cell subsets such as TH2, TH9, TH17, TH22, follicular helper T cells and CD28null T cells, as well as other T cell subsets including CD8+ T cells and γδ T cells, are less well understood. Moreover, some T cells seem to have both pro-atherogenic and anti-atherogenic functions. In this Review, we summarize the knowledge on T cell subsets, their functions in atherosclerosis and the process of T cell homing to atherosclerotic plaques. Much of our understanding of the roles of T cells in atherosclerosis is based on findings from experimental models. Translating these findings into human disease is challenging but much needed. T cells and their specific cytokines are attractive targets for developing new preventive and therapeutic approaches including potential T cell-related therapies for atherosclerosis.
Collapse
Affiliation(s)
- Ryosuke Saigusa
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Holger Winkels
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
94
|
Antonioli L, Fornai M, Pellegrini C, Masi S, Puxeddu I, Blandizzi C. Ectopic Lymphoid Organs and Immune-Mediated Diseases: Molecular Basis for Pharmacological Approaches. Trends Mol Med 2020; 26:1021-1033. [PMID: 32600794 DOI: 10.1016/j.molmed.2020.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/21/2020] [Accepted: 06/04/2020] [Indexed: 12/15/2022]
Abstract
Chronic inflammation is the result a persistent increase in the expression of several proinflammatory pathways with impaired inflammatory resolution. Ectopic lymphoid organs (ELOs), untypical lymphoid annexes, emerge during chronic inflammation and contribute to the physiopathology of chronic inflammatory disorders. This review discusses the pathophysiological role of ELOs in the progression of immune-mediated inflammatory diseases (IMIDs), including multiple sclerosis (MS), rheumatoid arthritis (RA), inflammatory bowel disease (IBD), atherosclerosis, and Sjögren syndrome (SSj). The molecular pathways underlying the emergence of ELOs are of interest for the development of novel pharmacological approaches for the management of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy.
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | | | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Ilaria Puxeddu
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
95
|
Padgett LE, Araujo DJ, Hedrick CC, Olingy CE. Functional crosstalk between T cells and monocytes in cancer and atherosclerosis. J Leukoc Biol 2020; 108:297-308. [PMID: 32531833 DOI: 10.1002/jlb.1mir0420-076r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/03/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022] Open
Abstract
Monocytes and monocyte-derived cells, including Mϕs and dendritic cells, exhibit a diverse array of phenotypic states that are dictated by their surrounding microenvironment. These cells direct T cell activation and function via cues that range from being immunosuppressive to immunostimulatory. Solid tumors and atherosclerotic plaques represent two pathological niches with distinct immune microenvironments. While monocytes and their progeny possess a phenotypic spectrum found within both disease contexts, most within tumors are pro-tumoral and support evasion of host immune responses by tumor cells. In contrast, monocyte-derived cells within atherosclerotic plaques are usually pro-atherogenic, pro-inflammatory, and predominantly directed against self-antigens. Consequently, cancer immunotherapies strive to enhance the immune response against tumor antigens, whereas atherosclerosis treatments seek to dampen the immune response against lipid antigens. Insights into monocyte-T cell interactions within these niches could thus inform therapeutic strategies for two immunologically distinct diseases. Here, we review monocyte diversity, interactions between monocytes and T cells within tumor and plaque microenvironments, how certain therapies have leveraged these interactions, and novel strategies to assay such associations.
Collapse
Affiliation(s)
- Lindsey E Padgett
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Daniel J Araujo
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Claire E Olingy
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, California, USA
| |
Collapse
|
96
|
Perivascular adipose tissue in age-related vascular disease. Ageing Res Rev 2020; 59:101040. [PMID: 32112889 DOI: 10.1016/j.arr.2020.101040] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/31/2020] [Accepted: 02/23/2020] [Indexed: 12/16/2022]
Abstract
Perivascular adipose tissue (PVAT), a crucial regulator of vascular homeostasis, is actively involved in vascular dysfunction during aging. PVAT releases various adipocytokines, chemokines and growth factors. In an endocrine and paracrine manner PVAT-derived factors regulate vascular signalling and inflammation modulating functions of adjacent layers of the vasculature. Pathophysiological conditions such as obesity, type 2 diabetes, vascular injury and aging can cause PVAT dysfunction, leading to vascular endothelial and smooth muscle cell dysfunctions. We and others have suggested that PVAT is involved in the inflammatory response of the vascular wall in diet induced obesity animal models leading to vascular dysfunction due to disappearance of the physiological anticontractile effect. Previous studies confirm a crucial role for pinpointed PVAT inflammation in promoting vascular oxidative stress and inflammation in aging, enhancing the risk for development of cardiovascular disease. In this review, we discuss several studies and mechanisms linking PVAT to age-related vascular diseases. An overview of the suggested roles played by PVAT in different disorders associated with the vasculature such as endothelial dysfunction, neointimal formation, aneurysm, vascular contractility and stiffness will be performed. PVAT may be considered a potential target for therapeutic intervention in age-related vascular disease.
Collapse
|
97
|
Tanaka S, Gauthier JM, Fuchs A, Li W, Tong AY, Harrison MS, Higashikubo R, Terada Y, Hachem RR, Ruiz-Perez D, Ritter JH, Cella M, Colonna M, Turnbull IR, Krupnick AS, Gelman AE, Kreisel D. IL-22 is required for the induction of bronchus-associated lymphoid tissue in tolerant lung allografts. Am J Transplant 2020; 20:1251-1261. [PMID: 31721409 PMCID: PMC7183893 DOI: 10.1111/ajt.15701] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 10/28/2019] [Accepted: 11/11/2019] [Indexed: 01/25/2023]
Abstract
Long-term survival after lung transplantation remains profoundly limited by graft rejection. Recent work has shown that bronchus-associated lymphoid tissue (BALT), characterized by the development of peripheral nodal addressin (PNAd)-expressing high endothelial venules and enriched in B and Foxp3+ T cells, is important for the maintenance of allograft tolerance. Mechanisms underlying BALT induction in tolerant pulmonary allografts, however, remain poorly understood. Here, we show that the development of PNAd-expressing high endothelial venules within intragraft lymphoid follicles and the recruitment of B cells, but not Foxp3+ cells depends on IL-22. We identify graft-infiltrating gamma-delta (γδ) T cells and Type 3 innate lymphoid cells (ILC3s) as important producers of IL-22. Reconstitution of IL-22 at late time points through retransplantation into wildtype hosts mediates B cell recruitment into lymphoid follicles within the allograft, resulting in a significant increase in their size, but does not induce PNAd expression. Our work has identified cellular and molecular requirements for the induction of BALT in pulmonary allografts during tolerance induction and may provide a platform for the development of new therapies for lung transplant patients.
Collapse
Affiliation(s)
- Satona Tanaka
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - Jason M. Gauthier
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - Anja Fuchs
- Section of Acute and Critical Care Surgery, Department of Surgery, Washington University, Saint Louis, Missouri
| | - Wenjun Li
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - Alice Y. Tong
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - M. Shea Harrison
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - Ryuji Higashikubo
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - Yuriko Terada
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - Ramsey R. Hachem
- Department of Medicine, Washington University, Saint Louis, Missouri
| | - Daniel Ruiz-Perez
- Department of Pathology & Immunology, Washington University, Saint Louis, Missouri
| | - Jon H. Ritter
- Division of Experimental Surgery, La Paz University Hospital, Madrid, Spain
| | - Marina Cella
- Division of Experimental Surgery, La Paz University Hospital, Madrid, Spain
| | - Marco Colonna
- Division of Experimental Surgery, La Paz University Hospital, Madrid, Spain
| | - Isaiah R. Turnbull
- Section of Acute and Critical Care Surgery, Department of Surgery, Washington University, Saint Louis, Missouri
| | - Alexander S. Krupnick
- Division of Thoracic Surgery, Department of Surgery, University of Virginia, Charlottesville, Virginia
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia
| | - Andrew E. Gelman
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
- Division of Experimental Surgery, La Paz University Hospital, Madrid, Spain
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
- Division of Experimental Surgery, La Paz University Hospital, Madrid, Spain
| |
Collapse
|
98
|
Milutinović A, Šuput D, Zorc-Pleskovič R. Pathogenesis of atherosclerosis in the tunica intima, media, and adventitia of coronary arteries: An updated review. Bosn J Basic Med Sci 2020; 20:21-30. [PMID: 31465719 PMCID: PMC7029210 DOI: 10.17305/bjbms.2019.4320] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of arteries and it affects the structure and function of all three layers of the coronary artery wall. Current theories suggest that the dysfunction of endothelial cells is one of the initial steps in the development of atherosclerosis. The view that the tunica intima normally consists of a single layer of endothelial cells attached to the subendothelial layer and internal elastic membrane has been questioned in recent years. The structure of intima changes with age and it becomes multilayered due to migration of smooth muscle cells from the media to intima. At this stage, the migration and proliferation of smooth muscle cells do not cause pathological changes in the intima. The multilayering of intima is classically considered to be an important stage in the development of atherosclerosis, but in fact atherosclerotic plaques develop only focally due to the interplay of various processes that involve the resident and invading inflammatory cells. The tunica media consists of multiple layers of smooth muscle cells that produce the extracellular matrix, and this layer normally does not contain microvessels. During the development of atherosclerosis, the microvessels from the tunica adventitia or from the lumen may penetrate thickened media to provide nutrition and oxygenation. According to some theories, the endothelial dysfunction of these nutritive vessels may significantly contribute to the atherosclerosis of coronary arteries. The adventitia contains fibroblasts, progenitor cells, immune cells, microvessels, and adrenergic nerves. The degree of inflammatory cell infiltration into the adventitia, which can lead to the formation of tertiary lymphoid organs, correlates with the severity of atherosclerotic plaques. Coronary arteries are surrounded by perivascular adipose tissue that also participates in the atherosclerotic process.
Collapse
Affiliation(s)
- Aleksandra Milutinović
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| | - Dušan Šuput
- Institute of Pathophysiology, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia.
| | - Ruda Zorc-Pleskovič
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; International Center for Cardiovascular Diseases MC Medicor d.d., Izola, Slovenia.
| |
Collapse
|
99
|
Upadhye A, Sturek JM, McNamara CA. 2019 Russell Ross Memorial Lecture in Vascular Biology: B Lymphocyte-Mediated Protective Immunity in Atherosclerosis. Arterioscler Thromb Vasc Biol 2020; 40:309-322. [PMID: 31852222 PMCID: PMC7398219 DOI: 10.1161/atvbaha.119.313064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/30/2019] [Indexed: 12/11/2022]
Abstract
Atherosclerosis-the major underlying pathology of cardiovascular disease-is characterized by accumulation and subsequent oxidative modification of lipoproteins within the artery wall, leading to inflammatory cell infiltration and lesion formation that can over time result in arterial stenosis, ischemia, and downstream adverse events. The contribution of innate and adaptive immunity to atherosclerosis development is well established, and B cells have emerged as important modulators of both pro- and anti-inflammatory effects in atherosclerosis. Murine B cells can broadly be divided into 2 subsets: (1) B-2 cells, which are bone marrow derived and include conventional follicular and marginal zone B cells, and (2) B-1 cells, which are largely fetal liver derived and persist in adults through self-renewal. B-cell subsets are developmentally, functionally, and phenotypically distinct with unique subset-specific contributions to atherosclerosis development. Mechanisms whereby B cells regulate vascular inflammation and atherosclerosis will be discussed with a particular emphasis on B-1 cells. B-1 cells have a protective role in atherosclerosis that is mediated in large part by IgM antibody production. Accumulating evidence over the last several years has pointed to a previously underappreciated heterogeneity in B-1 cell populations, which may have important implications for understanding atherosclerosis development and potential targeted therapeutic approaches. This heterogeneity within atheroprotective innate B-cell subsets will be highlighted.
Collapse
Affiliation(s)
- Aditi Upadhye
- From the Robert M. Berne Cardiovascular Research Center (A.U., C.A.M.), University of Virginia School of Medicine, Charlottesville
| | - Jeffrey M Sturek
- Division of Pulmonary and Critical Care Medicine, Department of Medicine (J.M.S.), University of Virginia School of Medicine, Charlottesville
| | - Coleen A McNamara
- From the Robert M. Berne Cardiovascular Research Center (A.U., C.A.M.), University of Virginia School of Medicine, Charlottesville
- Division of Cardiovascular Medicine (C.A.M.), University of Virginia School of Medicine, Charlottesville
| |
Collapse
|
100
|
Caravaca AS, Centa M, Gallina AL, Tarnawski L, Olofsson PS. Neural reflex control of vascular inflammation. Bioelectron Med 2020; 6:3. [PMID: 32232111 PMCID: PMC7065709 DOI: 10.1186/s42234-020-0038-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/14/2020] [Indexed: 12/26/2022] Open
Abstract
Atherosclerosis is a multifactorial chronic inflammatory disease that underlies myocardial infarction and stroke. Efficacious treatment for hyperlipidemia and hypertension has significantly reduced morbidity and mortality in cardiovascular disease. However, atherosclerosis still confers a considerable risk of adverse cardiovascular events. In the current mechanistic understanding of the pathogenesis of atherosclerosis, inflammation is pivotal both in disease development and progression. Recent clinical data provided support for this notion and treatment targeting inflammation is currently being explored. Interestingly, neural reflexes regulate cytokine production and inflammation. Hence, new technology utilizing implantable devices to deliver electrical impulses to activate neural circuits are currently being investigated in treatment of inflammation. Hopefully, it may become possible to target vascular inflammation in cardiovascular disease using bioelectronic medicine. In this review, we discuss neural control of inflammation and the potential implications of new therapeutic strategies to treat cardiovascular disease.
Collapse
Affiliation(s)
- A. S. Caravaca
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - M. Centa
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Biomedical Science and Bioelectronic Medicine, The Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| | - A. L. Gallina
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - L. Tarnawski
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - P. S. Olofsson
- Laboratory of Immunobiology, Center for Bioelectronic Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Biomedical Science and Bioelectronic Medicine, The Feinstein Institute for Medical Research, Manhasset, NY 11030 USA
| |
Collapse
|