1
|
Shu DH, Sidiropoulos DN. Maturation of Tertiary Lymphoid Structures. Methods Mol Biol 2025; 2864:43-55. [PMID: 39527216 DOI: 10.1007/978-1-0716-4184-2_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Tertiary lymphoid structures (TLS) are organized collections of B and T lymphocytes that arise in nonlymphoid tissue in response to chronic, unresolved inflammation. TLS have structural and functional similarities to germinal centers found in lymph nodes and are believed to support the establishment of lymph node-like adaptive immune responses at local sites of inflammation. However, understanding of the underlying biology of these structures remains limited, particularly the different stages of TLS life cycle and the signals governing the initiation, maturation, and termination of TLS. Here, we review current understanding of the maturation of TLS and the signals and cell types involved in various stages of development with particular emphasis on recent studies of TLS in cancer, where evidence suggests that TLS may play an important role in supporting antitumor immune responses in solid tumors.
Collapse
Affiliation(s)
- Daniel H Shu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Convergence Institute, Johns Hopkins University, Baltimore, MD, USA.
- Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, USA.
| | - Dimitrios N Sidiropoulos
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Convergence Institute, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
2
|
Zhao L, Jin S, Wang S, Zhang Z, Wang X, Chen Z, Wang X, Huang S, Zhang D, Wu H. Tertiary lymphoid structures in diseases: immune mechanisms and therapeutic advances. Signal Transduct Target Ther 2024; 9:225. [PMID: 39198425 PMCID: PMC11358547 DOI: 10.1038/s41392-024-01947-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/02/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024] Open
Abstract
Tertiary lymphoid structures (TLSs) are defined as lymphoid aggregates formed in non-hematopoietic organs under pathological conditions. Similar to secondary lymphoid organs (SLOs), the formation of TLSs relies on the interaction between lymphoid tissue inducer (LTi) cells and lymphoid tissue organizer (LTo) cells, involving multiple cytokines. Heterogeneity is a distinguishing feature of TLSs, which may lead to differences in their functions. Growing evidence suggests that TLSs are associated with various diseases, such as cancers, autoimmune diseases, transplant rejection, chronic inflammation, infection, and even ageing. However, the detailed mechanisms behind these clinical associations are not yet fully understood. The mechanisms by which TLS maturation and localization affect immune function are also unclear. Therefore, it is necessary to enhance the understanding of TLS development and function at the cellular and molecular level, which may allow us to utilize them to improve the immune microenvironment. In this review, we delve into the composition, formation mechanism, associations with diseases, and potential therapeutic applications of TLSs. Furthermore, we discuss the therapeutic implications of TLSs, such as their role as markers of therapeutic response and prognosis. Finally, we summarize various methods for detecting and targeting TLSs. Overall, we provide a comprehensive understanding of TLSs and aim to develop more effective therapeutic strategies.
Collapse
Affiliation(s)
- Lianyu Zhao
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Song Jin
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Shengyao Wang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Zhe Zhang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Xuan Wang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Zhanwei Chen
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Xiaohui Wang
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Shengyun Huang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| | - Dongsheng Zhang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| | - Haiwei Wu
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| |
Collapse
|
3
|
Zhao Z, Gao Y, Pei X, Wang W, Zhang H. Causal role of immune cells in Hashimoto's thyroiditis: Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1352616. [PMID: 38803479 PMCID: PMC11128540 DOI: 10.3389/fendo.2024.1352616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Objectives Hashimoto's thyroiditis (HT) is a common autoimmune disease whose etiology involves a complex interplay between genetics and environment. Previous studies have demonstrated an association between immune cells and HT. However, the casual relationship was not clear. We aimed to explore the causal associations between signatures of immune cells and HT. Methods In this study, bidirectional two-sample Mendelian randomization (MR) analysis was conducted to investigate the potential causal relationship between 731 immune cell signatures and HT by using genome-wide association study (GWAS) data. Heterogeneity and horizontal pleiotropy were detected through extensive sensitivity analyses. Results The increased levels of six immune phenotypes were observed to be causally associated with increased risk of HT P < 0.01, which were CD3 on CM CD8br, CD3 on CD39+ secreting Treg, HLA DR on CD33dim HLA DR+ CD11b-, CD3 on CD4 Treg, CD62L- plasmacytoid DC %DC, and CD3 on CD45RA+ CD4+. In addition, the levels of FSC-A on HLA DR+ T cell and CD62L on monocyte were associated with disease risk of HT P < 0.01. In addition, HT also had causal effects on CD3 on CM CD8br, CCR2 on monocyte, CD25 on CD39+ resting Treg, and CCR2 on CD62L+ myeloid DC P < 0.05. Conclusions In this study, we demonstrated the genetic connection between immune cell traits and HT, thereby providing guidance and direction for future treatment and clinical research.
Collapse
Affiliation(s)
| | | | | | | | - Huawei Zhang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
4
|
Ruddle NH. Posttransplant Tertiary Lymphoid Organs. Transplantation 2024; 108:1090-1099. [PMID: 37917987 PMCID: PMC11042531 DOI: 10.1097/tp.0000000000004812] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/20/2023] [Accepted: 07/07/2023] [Indexed: 11/04/2023]
Abstract
Tertiary lymphoid organs (TLOs), also known as tertiary or ectopic lymphoid structures or tissues, are accumulations of lymphoid cells in sites other than canonical lymphoid organs, that arise through lymphoid neogenesis during chronic inflammation in autoimmunity, microbial infection, cancer, aging, and transplantation, the focus of this review. Lymph nodes and TLOs are compared regarding their cellular composition, organization, vascular components, and migratory signal regulation. These characteristics of posttransplant TLOs (PT-TLOs) are described with individual examples in a wide range of organs including heart, kidney, trachea, lung, artery, skin, leg, hand, and face, in many species including human, mouse, rat, and monkey. The requirements for induction and maintenance of TLOs include sustained exposure to autoantigens, alloantigens, tumor antigens, ischemic reperfusion, nephrotoxic agents, and aging. Several staging schemes have been put forth regarding their function in organ rejection. PT-TLOs most often are associated with organ rejection, but in some cases contribute to tolerance. The role of PT-TLOs in cancer is considered in the case of immunosuppression. Furthermore, TLOs can be associated with development of lymphomas. Challenges for PT-TLO research are considered regarding staging, imaging, and opportunities for their therapeutic manipulation to inhibit rejection and encourage tolerance.
Collapse
Affiliation(s)
- Nancy H. Ruddle
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT
| |
Collapse
|
5
|
Han PZ, Ye WD, Yu PC, Tan LC, Shi X, Chen XF, He C, Hu JQ, Wei WJ, Lu ZW, Qu N, Wang Y, Ji QH, Ji DM, Wang YL. A distinct tumor microenvironment makes anaplastic thyroid cancer more lethal but immunotherapy sensitive than papillary thyroid cancer. JCI Insight 2024; 9:e173712. [PMID: 38478516 PMCID: PMC11141884 DOI: 10.1172/jci.insight.173712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 03/06/2024] [Indexed: 04/23/2024] Open
Abstract
Both anaplastic thyroid cancer (ATC) and papillary thyroid cancer (PTC) originate from thyroid follicular epithelial cells, but ATC has a significantly worse prognosis and shows resistance to conventional therapies. However, clinical trials found that immunotherapy works better in ATC than late-stage PTC. Here, we used single-cell RNA sequencing (scRNA-Seq) to generate a single-cell atlas of thyroid cancer. Differences in ATC and PTC tumor microenvironment components (including malignant cells, stromal cells, and immune cells) leading to the polarized prognoses were identified. Intriguingly, we found that CXCL13+ T lymphocytes were enriched in ATC samples and might promote the development of early tertiary lymphoid structure (TLS). Last, murine experiments and scRNA-Seq analysis of a treated patient's tumor demonstrated that famitinib plus anti-PD-1 antibody could advance TLS in thyroid cancer. We displayed the cellular landscape of ATC and PTC, finding that CXCL13+ T cells and early TLS might make ATC more sensitive to immunotherapy.
Collapse
Affiliation(s)
- Pei-Zhen Han
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei-Dong Ye
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng-Cheng Yu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li-Cheng Tan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Xiao Shi
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xu-Feng Chen
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cong He
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jia-Qian Hu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Jun Wei
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhong-Wu Lu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ning Qu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing-Hai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dong-Mei Ji
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yu-Long Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Ribatti D. Tertiary lymphoid structures, a historical reappraisal. Tissue Cell 2024; 86:102288. [PMID: 38101028 DOI: 10.1016/j.tice.2023.102288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Tertiary lymphoid structures (TLSs) are accumulations of lymphoid cells within non-lymphoid organs that share the cellular compartments, spatial organization, vasculature, chemokines, and function with secondary lymphoid organs, especially lymph nodes. TLSs are organized into a separate T cell and B cell compartments which contain germinal centers with follicular dendritic cells. In most cases, TLSs contain Peripheral Node addressin (PNAD) expressing high endothelial venules (HEVs). TLSs have been described in various mouse models of inflammation and are associated with a wide range of autoimmune diseases. Other than these, TLSs have been described in chronic allograft rejection and cancer.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
7
|
Singh A, Sharma A. Lymphoid tissue inducer cells in cancer: a potential therapeutic target. Mol Cell Biochem 2023; 478:2789-2794. [PMID: 36922480 DOI: 10.1007/s11010-023-04699-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/02/2023] [Indexed: 03/17/2023]
Abstract
Tumor cells are dynamic in nature; these cells first acquire immune surveillance and then escape from the immune system. Hence, progressed cancer cells distribute and metastasize to other organs via blood vessels as well as from the lymphatic system. Prognosis and treatment of metastatic cancer patients remain a major challenge nowadays. Till now, lots of target -based and immune checkpoint blocker therapies are used to treat disease patients. But these therapies fail to control the dissemination and metastasis of cancer. Before designing a treatment regimen for metastatic patients, understanding the mechanism of tumor cells spreading within lymph vessels remain undetermined. Construction of lymphoid structures since embryonic to adult stage are depend upon LTi. Foundation of lymph node, payer patches and TLO is initiated and regulated through these cells in any part of the body. During tumor growth, newly developed lymph node contained MDSCs and Treg cells which inhibit the immune response and promote tumor invasion and metastasis. LTi reconstituted lymph node can be used for both early and high risk detection of cancers. High and low risk of tumor growth and invasion depend upon the location and composition of immune cells within lymph nodes. However, LTi are not reported as predictive marker in cancer till date. Recent reports in cancer indicate that LTi cells are engaged in the spreading of tumor cells into a lymphatic vessel. Through this review we are trying to brief the development and role of the LTi in immune system during homeostasis and cancer.
Collapse
Affiliation(s)
- Ashu Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Alpana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
8
|
Ruddle NH. Regulation, Maintenance, and Remodeling of High Endothelial Venules in Homeostasis, Inflammation, and Cancer. CURRENT OPINION IN PHYSIOLOGY 2023; 36:100705. [PMID: 38523879 PMCID: PMC10956444 DOI: 10.1016/j.cophys.2023.100705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
High endothelial venules (HEVs), high walled cuboidal blood vessels, through their expression of adhesion molecules and chemokines, allow the entrance of lymphoid cells into primary, secondary, and tertiary lymphoid structures (aka tertiary lymphoid organs). HEV heterogeneity exists between various lymphoid organs in their expression of peripheral node addressin (PNAd) and mucosal vascular addressin adhesion molecule 1(MAdCAM-1). Transcriptomic analyses reveal extensive heterogeneity, plasticity, and regulation of HEV gene expression in ontogeny, acute inflammation, and chronic inflammation within and between lymphoid organs. Rules regulating HEV development are flexible in inflammation. HEVs in tumor tertiary lymphoid structures are diagnostic of favorable clinical outcome and response to Immunotherapy, including immune check point blockade. Immunotherapy induces HEVs and provides an entrance for naïve, central memory, and effector cells and a niche for stem like precursor cells. Understanding HEV regulation will permit their exploitation as routes for drug delivery to autoimmune lesions, rejecting organs, and tumors.
Collapse
Affiliation(s)
- Nancy H Ruddle
- Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06520-8034
| |
Collapse
|
9
|
Fang M, Zou J, Xu F, Wang X, Hua S, Zhou Q, Yang YG, Hu Z. Modeling human anti-pig xenoimmune responses in a pig artery tissue grafted humanized mouse model. Xenotransplantation 2023; 30:e12824. [PMID: 37695083 DOI: 10.1111/xen.12824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/28/2023] [Accepted: 08/29/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Blood vessels that contain endothelial cells (ECs) on the surface are in direct contact with host blood and are the first target of xenograft rejection. Currently, our understanding of human anti-pig vessel immune responses is primarily based on in vitro assays using pig ECs. Therefore, it is necessary to develop an animal model that permits in vivo study of human immunological rejection of pig vessels. METHODS Pig artery tissues (PAT) were transplanted into human immune system (HIS) mice or immunodeficient NSG mice (as controls). Intragraft human immune cell infiltration and antibody deposition were quantified using histology and immunohistochemistry. Donor antigen-specific immune responses were quantified using a mixed lymphocyte reaction and a complement-dependent killing assay. RESULTS Pig CD31+ ECs were detected and increased 2-fold from weeks 3 to 5 in PAT xenografts from immunodeficient NSG mice. However, compared with NSG mice, PAT xenografts in HIS mice had significantly lower numbers of porcine CD31+ ECs and showed a marked reduction from week 3 to week 5. PAT xenograft rejection in HIS mice is associated with intensive infiltration of human immune cells, deposition of human IgM and IgG antibodies, and the formation of a tertiary lymphoid structure. Robust donor pig antigen-specific human T cells and antibody responses were detected in PAT-transplanted HIS mice. CONCLUSION We have developed a humanized mouse model to evaluate human anti-pig xenoimmune responses by PAT transplantation in vivo. This model is expected to facilitate the refinement of pig gene-editing strategies (the expression on EC surface) and the testing of local immunosuppressive strategies for clinical pig organ xenotransplantation.
Collapse
Affiliation(s)
- Minghui Fang
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, and National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
| | - Jun Zou
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, and National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
| | - Fei Xu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, and National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
| | - Xue Wang
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, and National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
| | - Shucheng Hua
- Department of Respiration, The First Hospital of Jilin University, Changchun, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, and National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Zheng Hu
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, and National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
Magliozzi R, Howell OW, Calabrese M, Reynolds R. Meningeal inflammation as a driver of cortical grey matter pathology and clinical progression in multiple sclerosis. Nat Rev Neurol 2023:10.1038/s41582-023-00838-7. [PMID: 37400550 DOI: 10.1038/s41582-023-00838-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2023] [Indexed: 07/05/2023]
Abstract
Growing evidence from cerebrospinal fluid samples and post-mortem brain tissue from individuals with multiple sclerosis (MS) and rodent models indicates that the meninges have a key role in the inflammatory and neurodegenerative mechanisms underlying progressive MS pathology. The subarachnoid space and associated perivascular spaces between the membranes of the meninges are the access points for entry of lymphocytes, monocytes and macrophages into the brain parenchyma, and the main route for diffusion of inflammatory and cytotoxic molecules from the cerebrospinal fluid into the brain tissue. In addition, the meningeal spaces act as an exit route for CNS-derived antigens, immune cells and metabolites. A number of studies have demonstrated an association between chronic meningeal inflammation and a more severe clinical course of MS, suggesting that the build-up of immune cell aggregates in the meninges represents a rational target for therapeutic intervention. Therefore, understanding the precise cell and molecular mechanisms, timing and anatomical features involved in the compartmentalization of inflammation within the meningeal spaces in MS is vital. Here, we present a detailed review and discussion of the cellular, molecular and radiological evidence for a role of meningeal inflammation in MS, alongside the clinical and therapeutic implications.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy.
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| | - Owain W Howell
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
- Institute of Life Sciences, Swansea University, Swansea, UK
| | - Massimiliano Calabrese
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Richard Reynolds
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
11
|
Sato Y, Silina K, van den Broek M, Hirahara K, Yanagita M. The roles of tertiary lymphoid structures in chronic diseases. Nat Rev Nephrol 2023:10.1038/s41581-023-00706-z. [PMID: 37046081 PMCID: PMC10092939 DOI: 10.1038/s41581-023-00706-z] [Citation(s) in RCA: 124] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 04/14/2023]
Abstract
Tertiary lymphoid structures (TLSs) are ectopic lymphoid tissues that drive antigen-specific immune responses at sites of chronic inflammation. Unlike secondary lymphoid organs such as lymph nodes, TLSs lack capsules and have their own unique characteristics and functions. The presumed influence of TLSs on the disease course has led to widespread interest in obtaining a better understanding of their biology and function. Studies using single-cell analyses have suggested heterogeneity in TLS composition and phenotype, and consequently, functional correlates with disease progression are sometimes conflicting. The presence of TLSs correlates with a favourable disease course in cancer and infection. Conversely, in autoimmune diseases and chronic age-related inflammatory diseases including chronic kidney disease, the presence of TLSs is associated with a more severe disease course. However, the detailed mechanisms that underlie these clinical associations are not fully understood. To what extent the mechanisms of TLS development and maturation are shared across organs and diseases is also still obscure. Improved understanding of TLS development and function at the cellular and molecular levels may enable the exploitation of these structures to improve therapies for chronic diseases, including chronic kidney disease.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Karina Silina
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | | | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.
| |
Collapse
|
12
|
Laumont CM, Nelson BH. B cells in the tumor microenvironment: Multi-faceted organizers, regulators, and effectors of anti-tumor immunity. Cancer Cell 2023; 41:466-489. [PMID: 36917951 DOI: 10.1016/j.ccell.2023.02.017] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/11/2023] [Accepted: 02/12/2023] [Indexed: 03/14/2023]
Abstract
Our understanding of tumor-infiltrating lymphocytes (TILs) is rapidly expanding beyond T cell-centric perspectives to include B cells and plasma cells, collectively referred to as TIL-Bs. In many cancers, TIL-Bs carry strong prognostic significance and are emerging as key predictors of response to immune checkpoint inhibitors. TIL-Bs can perform multiple functions, including antigen presentation and antibody production, which allow them to focus immune responses on cognate antigen to support both T cell responses and innate mechanisms involving complement, macrophages, and natural killer cells. In the stroma of the most immunologically "hot" tumors, TIL-Bs are prominent components of tertiary lymphoid structures, which resemble lymph nodes structurally and functionally. Additionally, TIL-Bs participate in a variety of other lympho-myeloid aggregates and engage in dynamic interactions with the tumor stroma. Here, we summarize our current understanding of TIL-Bs in human cancer, highlighting the compelling therapeutic opportunities offered by their unique tumor recognition and effector mechanisms.
Collapse
Affiliation(s)
- Céline M Laumont
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Brad H Nelson
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8P 3E6, Canada.
| |
Collapse
|
13
|
Sato Y, Tamura M, Yanagita M. Tertiary lymphoid tissues: a regional hub for kidney inflammation. Nephrol Dial Transplant 2023; 38:26-33. [PMID: 34245300 DOI: 10.1093/ndt/gfab212] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Indexed: 01/26/2023] Open
Abstract
Tertiary lymphoid tissues (TLTs) are inducible ectopic lymphoid tissues that develop at sites of chronic inflammation in nonlymphoid organs. As with lymph nodes, TLTs initiate adaptive immune responses and coordinate local tissue immunity. Although virtually ignored for decades, TLTs have recently received a great deal of attention for their ability to influence disease severity, prognosis and response to therapy in various diseases, including cancer, autoimmune disorders and infections. TLTs are also induced in kidneys of patients with chronic kidney diseases such as immunoglobulin A nephropathy and lupus nephritis. Nevertheless, TLTs in the kidney have not been extensively investigated and their mechanism of development, functions and clinical relevance remain unknown, mainly because of the absence of adequate murine kidney TLT models and limited availability of human kidney samples containing TLTs. We recently found that aged kidneys, but not young kidneys, exhibit multiple TLTs after injury. Interestingly, although they are a minor component of TLTs, resident fibroblasts in the kidneys diversify into several distinct phenotypes that play crucial roles in TLT formation. Furthermore, the potential of TLTs as a novel kidney injury/inflammation marker as well as a novel therapeutic target for kidney diseases is also suggested. In this review article we describe the current understanding of TLTs with a focus on age-dependent TLTs in the kidney and discuss their potential as a novel therapeutic target and kidney inflammation marker.
Collapse
Affiliation(s)
- Yuki Sato
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masaru Tamura
- Technology and Development Team for Mouse Phenotype Analysis, Japan Mouse Clinic, RIKEN BioResource Research Center (BRC), Tsukuba, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| |
Collapse
|
14
|
Nie Y, Fan H, Li J, Lei X, Zhang T, Wang Y, Mao Z, Tao K, Song W. Tertiary lymphoid structures: Associated multiple immune cells and analysis their formation in hepatocellular carcinoma. FASEB J 2022; 36:e22586. [PMID: 36190431 DOI: 10.1096/fj.202200269rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 09/04/2022] [Accepted: 09/21/2022] [Indexed: 11/11/2022]
Abstract
The prognostic value of immune cells in tertiary lymphoid structures (TLSs) remains unclear in hepatocellular carcinoma (HCC). Here, 59 of 145 patients had TLSs in training set, 48 of 120 patients had TLSs in testing set. Immunohistochemistry (IHC) were used to label CD3+ T cells, CD20+ B cells, CD8+ T cells, CD208+ dendritic cells, and CD21+ follicular dendritic cells in TLSs. High CD20+, CD208+, and CD8+ cell densities were favorable prognostic factors for overall survival (OS). High CD3+, CD20+, CD208+, and CD8+ cell densities were significantly associated with reduced early recurrence. TLSs were divided into three grades (A, B, and C) based on immune cell density. Patients with grade C or B had significantly improved OS. Patients with grade C had the lowest recurrence rate, followed by those with grade B, while patients with grade A had the highest recurrence rate. The stromal, immune, and ESTIMATE scores derived from the ESTIMATE package were significantly higher and tumor purity was significantly lower in patients with TLSs. Patients with TLSs had significantly higher relative numbers of memory B cells, plasma cells, CD8+ T cells, NK cells, and dendritic cells and lower relative numbers of Treg cells, macrophages, and M2 macrophages according to the CIBERSORT assessment. Bioinformatics analysis and experiments confirmed that KLRK1 and GZMA expression are associated TLSs formation and can predict TLSs existence. Grade B and grade C were favorable prognostic factors for OS and recurrence and could represent immune-active tumors.
Collapse
Affiliation(s)
- Ye Nie
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hanlu Fan
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jianhui Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xinjun Lei
- Department of General Surgery, The Centre Hospital Weinan Shaanxi, Weinan, China
| | - Tianchen Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Yanfang Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.,Xi'an Medical University, Xi'an, China
| | - Zhenzhen Mao
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.,Xi'an Medical University, Xi'an, China
| | - Kaishan Tao
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wenjie Song
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
15
|
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: 42] [Impact Index Per Article: 14.0] [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
|
16
|
Hoch T, Schulz D, Eling N, Gómez JM, Levesque MP, Bodenmiller B. Multiplexed imaging mass cytometry of the chemokine milieus in melanoma characterizes features of the response to immunotherapy. Sci Immunol 2022; 7:eabk1692. [PMID: 35363540 DOI: 10.1126/sciimmunol.abk1692] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Intratumoral immune cells are crucial for tumor control and antitumor responses during immunotherapy. Immune cell trafficking into tumors is mediated by binding of specific immune cell receptors to chemokines, a class of secreted chemotactic cytokines. To broadly characterize chemokine expression and function in melanoma, we used multiplexed mass cytometry-based imaging of protein markers and RNA transcripts to analyze the chemokine landscape and immune infiltration in metastatic melanoma samples. Tumors that lacked immune infiltration were devoid of most of the profiled chemokines and exhibited low levels of antigen presentation and markers of inflammation. Infiltrated tumors were characterized by expression of multiple chemokines. CXCL9 and CXCL10 were often localized in patches associated with dysfunctional T cells expressing the B lymphocyte chemoattractant CXCL13. In tumors with B cells but no B cell follicles, T cells were the sole source of CXCL13, suggesting that T cells play a role in B cell recruitment and potentially in B cell follicle formation. B cell patches and follicles were also enriched with TCF7+ naïve-like T cells, a cell type that is predictive of response to immune checkpoint blockade. Our data highlight the strength of targeted RNA and protein codetection to analyze tumor immune microenvironments based on chemokine expression and suggest that the formation of tertiary lymphoid structures may be accompanied by naïve and naïve-like T cell recruitment, which may contribute to antitumor activity.
Collapse
Affiliation(s)
- Tobias Hoch
- University of Zürich, Department of Quantitative Biomedicine, Zürich 8057, Switzerland.,ETH-Zürich, Institute for Molecular Health Sciences, Zürich 8093, Switzerland.,Particles-Biology Interactions Laboratory, Empa, Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Daniel Schulz
- University of Zürich, Department of Quantitative Biomedicine, Zürich 8057, Switzerland.,ETH-Zürich, Institute for Molecular Health Sciences, Zürich 8093, Switzerland
| | - Nils Eling
- University of Zürich, Department of Quantitative Biomedicine, Zürich 8057, Switzerland.,ETH-Zürich, Institute for Molecular Health Sciences, Zürich 8093, Switzerland
| | - Julia Martínez Gómez
- University Hospital Zürich, Department of Dermatology, Schlieren 8952, Switzerland
| | - Mitchell P Levesque
- University Hospital Zürich, Department of Dermatology, Schlieren 8952, Switzerland
| | - Bernd Bodenmiller
- University of Zürich, Department of Quantitative Biomedicine, Zürich 8057, Switzerland.,ETH-Zürich, Institute for Molecular Health Sciences, Zürich 8093, Switzerland
| |
Collapse
|
17
|
Trujillo-Vargas CM, Mauk KE, Hernandez H, de Souza RG, Yu Z, Galletti JG, Dietrich J, Paulsen F, de Paiva CS. Immune phenotype of the CD4 + T cells in the aged lymphoid organs and lacrimal glands. GeroScience 2022; 44:2105-2128. [PMID: 35279788 DOI: 10.1007/s11357-022-00529-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/11/2022] [Indexed: 02/07/2023] Open
Abstract
Aging is associated with a massive infiltration of T lymphocytes in the lacrimal gland. Here, we aimed to characterize the immune phenotype of aged CD4+ T cells in this tissue as compared with lymphoid organs. To perform this, we sorted regulatory T cells (Tregs, CD4+CD25+GITR+) and non-Tregs (CD4+CD25negGITRneg) in lymphoid organs from female C57BL/6J mice and subjected these cells to an immunology NanoString® panel. These results were confirmed by flow cytometry, live imaging, and tissue immunostaining in the lacrimal gland. Importantly, effector T helper 1 (Th1) genes were highly upregulated on aged Tregs, including the master regulator Tbx21. Among the non-Tregs, we also found a significant increase in the levels of EOMESmed/high, TbetnegIFN-γ+, and CD62L+CD44negCD4+ T cells with aging, which are associated with cell exhaustion, immunopathology, and the generation of tertiary lymphoid tissue. At the functional level, aged Tregs from lymphoid organs are less able to decrease proliferation and IFN-γ production of T responders at any age. More importantly, human lacrimal glands (age range 55-81 years) also showed the presence of CD4+Foxp3+ cells. Further studies are needed to propose potential molecular targets to avoid immune-mediated lacrimal gland dysfunction with aging.
Collapse
Affiliation(s)
- Claudia M Trujillo-Vargas
- Grupo de Inmunodeficiencias Primarias, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellín, Colombia.,Department of Ophthalmology, Ocular Surface Center, Cullen Eye Institute, Baylor College of Medicine, 6565 Fannin Street, Houston, TX, NC 505G, USA
| | - Kelsey E Mauk
- Graduate Program in Immunology & Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Humberto Hernandez
- Department of Ophthalmology, Ocular Surface Center, Cullen Eye Institute, Baylor College of Medicine, 6565 Fannin Street, Houston, TX, NC 505G, USA
| | - Rodrigo G de Souza
- Department of Ophthalmology, Ocular Surface Center, Cullen Eye Institute, Baylor College of Medicine, 6565 Fannin Street, Houston, TX, NC 505G, USA
| | - Zhiyuan Yu
- Department of Ophthalmology, Ocular Surface Center, Cullen Eye Institute, Baylor College of Medicine, 6565 Fannin Street, Houston, TX, NC 505G, USA
| | - Jeremias G Galletti
- Institute of Experimental Medicine, CONICET-National Academy of Medicine of Buenos Aires, Buenos Aires, Argentina
| | - Jana Dietrich
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Cintia S de Paiva
- Department of Ophthalmology, Ocular Surface Center, Cullen Eye Institute, Baylor College of Medicine, 6565 Fannin Street, Houston, TX, NC 505G, USA.
| |
Collapse
|
18
|
Chaurio RA, Anadon CM, Costich TL, Payne KK, Biswas S, Harro CM, Moran C, Ortiz AC, Cortina C, Rigolizzo KE, Sprenger KB, Mine JA, Innamarato PP, Mandal G, Powers JJ, Martin A, Wang Z, Mehta S, Perez BA, Li R, Robinson J, Kroeger JL, Curiel TJ, Yu X, Rodriguez PC, Conejo-Garcia JR. TGF-β-mediated silencing of genomic organizer SATB1 promotes Tfh cell differentiation and formation of intra-tumoral tertiary lymphoid structures. Immunity 2022; 55:115-128.e9. [PMID: 35021053 PMCID: PMC8852221 DOI: 10.1016/j.immuni.2021.12.007] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 09/17/2021] [Accepted: 12/08/2021] [Indexed: 01/13/2023]
Abstract
The immune checkpoint receptor PD-1 on T follicular helper (Tfh) cells promotes Tfh:B cell interactions and appropriate positioning within tissues. Here, we examined the impact of regulation of PD-1 expression by the genomic organizer SATB1 on Tfh cell differentiation. Vaccination of CD4CreSatb1f/f mice enriched for antigen-specific Tfh cells, and TGF-β-mediated repression of SATB1 enhanced Tfh differentiation of human T cells. Mechanistically, high Icos expression in Satb1-/- CD4+ T cells promoted Tfh cell differentiation by preventing T follicular regulatory cell skewing and resulted in increased isotype-switched B cell responses in vivo. Ovarian tumors in CD4CreSatb1f/f mice accumulated tumor antigen-specific, LIGHT+CXCL13+IL-21+ Tfh cells and tertiary lymphoid structures (TLS). TLS formation decreased tumor growth in a CD4+ T cell and CXCL13-dependent manner. The transfer of Tfh cells, but not naive CD4+ T cells, induced TLS at tumor beds and decreased tumor growth. Thus, TGF-β-mediated silencing of Satb1 licenses Tfh cell differentiation, providing insight into the genesis of TLS within tumors.
Collapse
Affiliation(s)
- Ricardo A Chaurio
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Carmen M Anadon
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Tara Lee Costich
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kyle K Payne
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Subir Biswas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Carly M Harro
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Carlos Moran
- Department of Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Antonio C Ortiz
- Department of Analytic Microscopy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Carla Cortina
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kristen E Rigolizzo
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Kimberly B Sprenger
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Jessica A Mine
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Pasquale P Innamarato
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Gunjan Mandal
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - John J Powers
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Alexandra Martin
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Zhitao Wang
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Sumit Mehta
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Bradford A. Perez
- Department of Radiation Therapy, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Roger Li
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - John Robinson
- Department of Flow Cytometry Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Jodi L Kroeger
- Department of Flow Cytometry Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Tyler J Curiel
- Mays Cancer Center, University of Texas Health, San Antonio, TX 78229
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Paulo C. Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.,Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.,Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.,CORRESPONDENCE: Jose R Conejo-Garcia, MD, PhD (LEAD CONTACT), H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, , Phone: (813) 745-8282, Fax: (813) 745-5580
| |
Collapse
|
19
|
Lin J, Chen Y, Zhu H, Cheng K, Wang H, Yu X, Tang M, Chen J. Lymphatic Reconstruction in Kidney Allograft Aggravates Chronic Rejection by Promoting Alloantigen Presentation. Front Immunol 2021; 12:796260. [PMID: 34956231 PMCID: PMC8695730 DOI: 10.3389/fimmu.2021.796260] [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: 10/16/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic rejection of the renal allograft remains a major cause of graft loss. Here, we demonstrated that the remodeling of lymphatic vessels (LVs) after their broken during transplantation contributes to the antigen presenting and lymph nodes activating. Our studies observed a rebuilt of interrupted lymph draining one week after mouse kidney transplantation, involving preexisting lymphatic endothelial cells (LECs) from both the donor and recipient. These expanding LVs also release C-C chemokine ligand 21 (CCL21) and recruit CCR7+ cells, mainly dendritic cells (DCs), toward lymph nodes and spleen, evoking the adaptive response. This rejection could be relieved by LYVE-1 specific LVs knockout or CCR7 migration inhibition in mouse model. Moreover, in retrospective analysis, posttransplant patients exhibiting higher area density of LVs presented with lower eGFR, severe serum creatinine and proteinuria, and greater interstitial fibrosis. These results reveal a rebuilt pathway for alloantigen trafficking and lymphocytes activation, providing strategies to alleviate chronic transplantation rejection.
Collapse
Affiliation(s)
- Jinwen Lin
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Huijuan Zhu
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Kai Cheng
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Huiping Wang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xianping Yu
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Mengmeng Tang
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China,*Correspondence: Jianghua Chen,
| |
Collapse
|
20
|
Mustapha R, Ng K, Monypenny J, Ng T. Insights Into Unveiling a Potential Role of Tertiary Lymphoid Structures in Metastasis. Front Mol Biosci 2021; 8:661516. [PMID: 34568423 PMCID: PMC8455920 DOI: 10.3389/fmolb.2021.661516] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Tertiary lymphoid structures (TLSs) develop in non-lymphatic tissue in chronic inflammation and cancer. TLS can mature to lymph node (LN) like structures with germinal centers and associated vasculature. TLS neogenesis in cancer is highly varied and tissue dependent. The role of TLS in adaptive antitumor immunity is of great interest. However, data also show that TLS can play a role in cancer metastasis. The importance of lymphatics in cancer distant metastasis is clear yet the precise detail of how various immunosurveillance mechanisms interplay within TLS and/or draining LN is still under investigation. As part of the tumor lymphatics, TLS vasculature can provide alternative routes for the establishment of the pre-metastatic niche and cancer dissemination. The nature of the cytokine and chemokine signature at the heart of TLS induction can be key in determining the success of antitumor immunity or in promoting cancer invasiveness. Understanding the biochemical and biomechanical factors underlying TLS formation and the resulting impact on the primary tumor will be key in deciphering cancer metastasis and in the development of the next generation of cancer immunotherapeutics.
Collapse
Affiliation(s)
- Rami Mustapha
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Medical School Campus, London, United Kingdom
- Cancer Research UK King’s Health Partners Centre, London, United Kingdom
| | - Kenrick Ng
- UCL Cancer Institute, University College London, London, United Kingdom
- Department of Medical Oncology, University College Hospitals NHS Foundation Trust, London, United Kingdom
| | - James Monypenny
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Medical School Campus, London, United Kingdom
| | - Tony Ng
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Medical School Campus, London, United Kingdom
- Cancer Research UK King’s Health Partners Centre, London, United Kingdom
- UCL Cancer Institute, University College London, London, United Kingdom
- Cancer Research UK City of London Centre, London, United Kingdom
| |
Collapse
|
21
|
Asam S, Nayar S, Gardner D, Barone F. Stromal cells in tertiary lymphoid structures: Architects of autoimmunity. Immunol Rev 2021; 302:184-195. [PMID: 34060101 DOI: 10.1111/imr.12987] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
The molecular mediators present within the inflammatory microenvironment are able, in certain conditions, to favor the initiation of tertiary lymphoid structure (TLS) development. TLS is organized lymphocyte clusters able to support antigen-specific immune response in non-immune organs. Importantly, chronic inflammation does not always result in TLS formation; instead, TLS has been observed to develop specifically in permissive organs, suggesting the presence of tissue-specific cues that are able to imprint the immune responses and form TLS hubs. Fibroblasts are tissue-resident cells that define the anatomy and function of a specific tissue. Fibroblast plasticity and specialization in inflammatory conditions have recently been unraveled in both immune and non-immune organs revealing a critical role for these structural cells in human physiology. Here, we describe the role of fibroblasts in the context of TLS formation and its functional maintenance in the tissue, highlighting their potential role as therapeutic disease targets in TLS-associated diseases.
Collapse
Affiliation(s)
- Saba Asam
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Saba Nayar
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, UK
| | - David Gardner
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Francesca Barone
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
22
|
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
|
23
|
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: 106] [Impact Index Per Article: 26.5] [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
|
24
|
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: 105] [Impact Index Per Article: 26.3] [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
|
25
|
Lim JH, Han MH, Kim YJ, Jeon Y, Jung HY, Choi JY, Cho JH, Kim CD, Kim YL, Lee H, Kim DK, Moon KC, Park SH. Novel histopathologic predictors for renal outcomes in crescentic glomerulonephritis. PLoS One 2020; 15:e0236051. [PMID: 32716952 PMCID: PMC7384637 DOI: 10.1371/journal.pone.0236051] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/27/2020] [Indexed: 01/11/2023] Open
Abstract
Introduction Crescentic glomerulonephritis (CrGN) is a histologic feature of severe glomerular injury, clinically characterized by a rapid decline of renal function when not treated in a timely fashion. Factors associated with CrGN prognosis have not been thoroughly investigated. This study investigated the prognostic predictors of renal outcomes associated with CrGN, such as the histopathologic classification of anti-neutrophil cytoplasmic antibody (ANCA)-associated glomerulonephritis, arteriosclerosis, and tertiary lymphoid organ (TLO) formation. Methods A total of 114 patients diagnosed with CrGN between 2010 and 2018 at two university-based hospitals has been retrospectively analyzed. Relationships between potential predictors and renal outcomes were analyzed using Cox proportional hazards model and linear regression analysis. Results The mean age was 61.0 ± 15.3 years, and 49.1% were male. Among them, 92 (80.7%) and 11 (9.6%) patients were positive for ANCA and for anti-glomerular basement membrane antibody, respectively. During the median follow-up of 458.0 days, 55 patients (48.2%) had advanced to end-stage renal disease (ESRD). Cox proportional hazards analysis revealed that patients under the mixed and sclerotic classes had worse renal survival compared to those in the focal class (mixed: hazard ratio [HR], 3.74; 95% confidence interval [CI], 1.18 to 11.82; P = 0.025; sclerotic: HR, 4.84; 95% CI, 1.44 to 16.32; P = 0.011). Severe arteriosclerosis was also associated with poor renal survival (HR, 2.44; 95% CI, 1.04 to 5.77; P = 0.042). TLOs were observed in 41 patients (36.0%). Moreover, TLO formation was also a prognostic factor for ESRD (HR, 1.82; 95% CI, 1.03 to 3.21; P = 0.040). In the multivariate linear regression analysis, age and sclerotic class were independent predictors for the change in estimated glomerular filtration rate during 1 year after biopsy. Conclusions Specific histopathologic findings, histopathologic classification, severity of arteriosclerosis, and TLO formation provide helpful information in predicting renal outcomes associated with CrGN.
Collapse
Affiliation(s)
- Jeong-Hoon Lim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Man-Hoon Han
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Yong-Jin Kim
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Yena Jeon
- Department of Statistics, Kyungpook National University, Daegu, South Korea
| | - Hee-Yeon Jung
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Ji-Young Choi
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Jang-Hee Cho
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Chan-Duck Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Yong-Lim Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyung Chul Moon
- Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
| | - Sun-Hee Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, South Korea
- * E-mail:
| |
Collapse
|
26
|
Marinkovic D, Marinkovic T. Putative role of marginal zone B cells in pathophysiological processes. Scand J Immunol 2020; 92:e12920. [PMID: 32594535 DOI: 10.1111/sji.12920] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/21/2020] [Indexed: 12/16/2022]
Abstract
The maintenance of inner integrity of an organism is founded on the proper performance of two immunity branches, innate and adaptive immune responses. Recently, it became apparent that subset of splenic B cells named marginal zone B cells (MZB cells) exhibits unique developmental and functional features that bridge these two immunity branches. Strategically positioned at the site where blood and lymph are filtered, MZB cells represent a population of sentinels that rapidly proliferate and differentiate into IgM plasmablast cells when encountered with blood-borne, thymus-independent (TI) Ags. Moreover, MZB cells have intrinsic capability to induce potent CD4+ helper T cell response and cytokine production upon stimulation with soluble antigens. Due to their ability to overcome a time gap prior the establishment of the full adaptive response towards pathogens, MZB cells connect and direct innate and adaptive immunity. An additional interesting characteristic of MZB cells is capacity to function as regulatory cells in autoimmune processes. MZB cells may also contribute to the control of autoimmunity via the induction of tolerance by apoptotic cells. Importantly, in the clear association with inflammation and autoimmunity, MZB cells may transform into MALT lymphoma, representing a concurrence point for the infection, immunity and malignancy. This paper presents an insight into the complex biology of marginal zone B cells and their role in intertwining and directing innate and adaptive immune processes at the physiological and pathological level.
Collapse
Affiliation(s)
- Dragan Marinkovic
- Faculty of Special Education and Rehabilitation, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
27
|
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: 6] [Impact Index Per Article: 1.2] [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
|
28
|
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
|
29
|
Conejo-Garcia JR, Biswas S, Chaurio R. Humoral immune responses: Unsung heroes of the war on cancer. Semin Immunol 2020; 49:101419. [PMID: 33183950 PMCID: PMC7738315 DOI: 10.1016/j.smim.2020.101419] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/23/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
Solid cancers progress from primordial lesions through complex interactions between tumor-promoting and anti-tumor immune cell types, ultimately leading to the orchestration of humoral and T cell adaptive immune responses, albeit in an immunosuppressive environment. B cells infiltrating most established tumors have been associated with a dual role: Some studies have associated antibodies produced by tumor-associated B cells with the promotion of regulatory activities on myeloid cells, and also with direct immunosuppression through the production of IL-10, IL-35 or TGF-β. In contrast, recent studies in multiple human malignancies identify B cell responses with delayed malignant progression and coordinated T cell protective responses. This includes the elusive role of Tertiary Lymphoid Structures identified in many human tumors, where the function of B cells remains unknown. Here, we discuss emerging data on the dual role of B cell responses in the pathophysiology of human cancer, providing a perspective on future directions and possible novel interventions to restore the coordinated action of both branches of the adaptive immune response, with the goal of maximizing immunotherapeutic effectiveness.
Collapse
Affiliation(s)
- Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | - Subir Biswas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Ricardo Chaurio
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
30
|
Silva-Cayetano A, Linterman MA. Stromal cell control of conventional and ectopic germinal centre reactions. Curr Opin Immunol 2020; 64:26-33. [PMID: 32325390 DOI: 10.1016/j.coi.2020.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/21/2020] [Accepted: 03/10/2020] [Indexed: 12/21/2022]
Abstract
The germinal centre (GC) is a specialized cellular structure that forms in response to antigenic stimulation. It generates long-term humoral immunity through the production of memory B cells and long-lived antibody-secreting plasma cells. Conventional GCs form within secondary lymphoid organs, where networks of specialised stromal cells that form during embryogenesis act as the stage upon which the various GC immune cell players are brought together, nurtured and co-ordinated to generate a productive response. In non-lymphoid organs, ectopic GCs can form in response to persistent antigenic and inflammatory stimuli. Unlike secondary lymphoid tissues, non-lymphoid organs do not have a developmentally programmed stromal cell network capable of supporting the germinal centre reaction; therefore, the local tissue stroma must be remodelled by inflammatory stimuli in order to host a GC reaction. These ectopic GCs produce memory B cells and plasma cells that form a critical component of the humoral immune response.
Collapse
|
31
|
Sautès-Fridman C, Verneau J, Sun CM, Moreira M, Chen TWW, Meylan M, Petitprez F, Fridman WH. Tertiary Lymphoid Structures and B cells: Clinical impact and therapeutic modulation in cancer. Semin Immunol 2020; 48:101406. [DOI: 10.1016/j.smim.2020.101406] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022]
|
32
|
Abstract
Tertiary lymphoid organs (TLOs), also known as inducible lymphoid organs, tertiary lymphoid structures, tertiary lymphoid tissues, or ectopic lymphoid organs are accumulations of cells in chronic inflammation that have been observed in most tissues in autoimmunity, infection, and cancer in mouse and man. They share many properties with secondary lymphoid organs (SLOs), particularly lymph nodes, with regard to cellular composition, function, and regulation. TLOs include T and B cells, dendritic cells, follicular dendritic cells, and many other stromal cells, and high endothelial venules (HEVs) and lymphatic vessels. They serve as sites of antigen presentation and tolerance induction; they are harmful in autoimmunity and can be both harmful and beneficial in cancer. SLO induction in ontogeny is mediated by interactions of several cell types, including CD4+ CD3- lymphoid tissue inducer (LTi) RORγt+ cells that express LTαβ and interact with mesenchymal lymphoid tissue organizer (LTo) FAP+ cells in the presence of lymphatic and blood vessels. A variety of inducer cells initiate TLOs, including bona fide LTi cells, T cells, B cells, and NK cells. The mesenchymal organizer cells are less well characterized but can include FAP+ cells. Current challenges include identification of methods to inhibit TLOs in autoimmunity without affecting SLOs, and enhancement of TLOs for defense against tumors.
Collapse
Affiliation(s)
- Nancy H Ruddle
- Epidemiology of Microbial Diseases, Yale School of Public Health, 60 College St., New Haven, CT, 06510, USA.
| |
Collapse
|
33
|
Luo S, Zhu R, Yu T, Fan H, Hu Y, Mohanta SK, Hu D. Chronic Inflammation: A Common Promoter in Tertiary Lymphoid Organ Neogenesis. Front Immunol 2019; 10:2938. [PMID: 31921189 PMCID: PMC6930186 DOI: 10.3389/fimmu.2019.02938] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 11/29/2019] [Indexed: 12/15/2022] Open
Abstract
Tertiary lymphoid organs (TLOs) frequently develop locally in adults in response to non-resolving inflammation. Chronic inflammation leads to the differentiation of stromal fibroblast cells toward lymphoid tissue organizer-like cells, which interact with lymphotoxin α1β2+ immune cells. The interaction initiates lymphoid neogenesis by recruiting immune cells to the site of inflammation and ultimately leads to the formation of TLOs. Mature TLOs harbor a segregated T-cell zone, B-cell follicles with an activated germinal center, follicular dendritic cells, and high endothelial venules, which architecturally resemble those in secondary lymphoid organs. Since CXCL13 and LTα1β2 play key roles in TLO neogenesis, they might constitute potential biomarkers of TLO activity. The well-developed TLOs actively regulate local immune responses and influence disease progression, and they are thereby regarded as the powerhouses of local immunity. In this review, we recapitulated the determinants for TLOs development, with great emphasis on the fundamental role of chronic inflammation and tissue-resident stromal cells for TLO neogenesis, hence offering guidance for therapeutic interventions in TLO-associated diseases.
Collapse
Affiliation(s)
- Shanshan Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Yu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sarajo Kumar Mohanta
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Desheng Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
34
|
Couillault C, Germain C, Dubois B, Kaplon H. Identification of Tertiary Lymphoid Structure-Associated Follicular Helper T Cells in Human Tumors and Tissues. Methods Mol Biol 2019; 1845:205-222. [PMID: 30141015 DOI: 10.1007/978-1-4939-8709-2_12] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Follicular helper T (Tfh) cells are major components of the humoral immune response due to their pivotal role in germinal center formation and antibody affinity maturation following B-cell isotype switching. This CD4+ T-cell subtype is mainly found in the B-cell zone of secondary lymphoid organs as well as in tertiary lymphoid structures (TLS), which are highly organized structures composed of T and B cells, occasionally found at the invasive margin in the tumor microenvironment.We describe here how to perform immunofluorescence staining of tumor tissue sections and multicolor flow cytometry on tumor cell suspensions to identify and visualize these TLS-associated Tfh cells within the tumor microenvironment of various human cancers. These assays take advantage of combinations of markers and molecules involved in Tfh differentiation and function.
Collapse
Affiliation(s)
- Coline Couillault
- Laboratory "Targeting of the Tumor and Its Immune Microenvironment", Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, University Claude Bernard Lyon 1, Institut National de la Santé et de la Recherche Médicale (INSERM) U1052, CNRS 5286, Lyon, France
| | - Claire Germain
- Cordeliers Research Center, Laboratory "Cancer, Immune Control and Escape", INSERM, UMRS 1138, Paris, France.,Cordeliers Research Center, Paris Descartes University, Sorbonne Paris Cité, UMRS 1138, Paris, France.,Cordeliers Research Center, Sorbonne University, UMRS 1138, Paris, France.,Laboratory "Immune Intervention and Biotherapies", UPMC UMRS CR7-Inserm U1135-CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI), Paris, France
| | - Bertrand Dubois
- Laboratory "Targeting of the Tumor and Its Immune Microenvironment", Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, University Claude Bernard Lyon 1, Institut National de la Santé et de la Recherche Médicale (INSERM) U1052, CNRS 5286, Lyon, France.
| | - Hélène Kaplon
- Cordeliers Research Center, Laboratory "Cancer, Immune Control and Escape", INSERM, UMRS 1138, Paris, France.,Cordeliers Research Center, Paris Descartes University, Sorbonne Paris Cité, UMRS 1138, Paris, France.,Cordeliers Research Center, Sorbonne University, UMRS 1138, Paris, France
| |
Collapse
|
35
|
Abstract
Although common evolutionary principles drive the growth of cancer cells regardless of the tissue of origin, the microenvironment in which tumours arise substantially differs across various organ sites. Recent studies have established that, in addition to cell-intrinsic effects, tumour growth regulation also depends on local cues driven by tissue environmental factors. In this Review, we discuss how tissue-specific determinants might influence tumour development and argue that unravelling the tissue-specific contribution to tumour immunity should help the development of precise immunotherapeutic strategies for patients with cancer.
Collapse
Affiliation(s)
- Hélène Salmon
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Precision Immunology Institute and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- INSERM U932, Institut Curie, Paris, France.
| | | | - Sacha Gnjatic
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Precision Immunology Institute and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
36
|
Tan HX, Esterbauer R, Vanderven HA, Juno JA, Kent SJ, Wheatley AK. Inducible Bronchus-Associated Lymphoid Tissues (iBALT) Serve as Sites of B Cell Selection and Maturation Following Influenza Infection in Mice. Front Immunol 2019; 10:611. [PMID: 30984186 PMCID: PMC6450362 DOI: 10.3389/fimmu.2019.00611] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/07/2019] [Indexed: 12/14/2022] Open
Abstract
Seasonally recurrent influenza virus infections are a significant cause of global morbidity and mortality. In murine models, primary influenza infection in the respiratory tract elicits potent humoral responses concentrated in the draining mediastinal lymph node and the spleen. In addition to immunity within secondary lymphoid organs (SLO), pulmonary infection is also associated with formation of ectopic inducible bronchus-associated tissues (iBALT) in the lung. These structures display a lymphoid organization, but their function and protective benefits remain unclear. Here we examined the phenotype, transcriptional profile and antigen specificity of B cell populations forming iBALT in influenza infected mice. We show that the cellular composition of iBALT was comparable to SLO, containing populations of follicular dendritic cells (FDC), T-follicular helper (Tfh) cells, and germinal center (GC)-like B cells with classical dark- and light-zone polarization. Transcriptional profiles of GC B cells in iBALT and SLO were conserved regardless of anatomical localization. The architecture of iBALT was pleiomorphic and less structurally defined than SLO. Nevertheless, we show that GC-like structures within iBALT serve as a distinct niche that independently support the maturation and selection of B cells primarily targeted against the influenza virus nucleoprotein. Our findings suggest that iBALT, which are positioned at the frontline of the lung mucosa, drive long-lived, and unique GC reactions that contribute to the diversity of the humoral response targeting influenza.
Collapse
Affiliation(s)
- Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Robyn Esterbauer
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Hillary A Vanderven
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.,Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.,Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia.,ARC Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, VIC, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
37
|
Abstract
Pulmonary respiration inevitably exposes the mucosal surface of the lung to potentially noxious stimuli, including pathogens, allergens, and particulates, each of which can trigger pulmonary damage and inflammation. As inflammation resolves, B and T lymphocytes often aggregate around large bronchi to form inducible Bronchus-Associated Lymphoid Tissue (iBALT). iBALT formation can be initiated by a diverse array of molecular pathways that converge on the activation and differentiation of chemokine-expressing stromal cells that serve as the scaffolding for iBALT and facilitate the recruitment, retention, and organization of leukocytes. Like conventional lymphoid organs, iBALT recruits naïve lymphocytes from the blood, exposes them to local antigens, in this case from the airways, and supports their activation and differentiation into effector cells. The activity of iBALT is demonstrably beneficial for the clearance of respiratory pathogens; however, it is less clear whether it dampens or exacerbates inflammatory responses to non-infectious agents. Here, we review the evidence regarding the role of iBALT in pulmonary immunity and propose that the final outcome depends on the context of the disease.
Collapse
|
38
|
Barron AMS, Mantero JC, Ho JD, Nazari B, Horback KL, Bhawan J, Lafyatis R, Lam C, Browning JL. Perivascular Adventitial Fibroblast Specialization Accompanies T Cell Retention in the Inflamed Human Dermis. THE JOURNAL OF IMMUNOLOGY 2018; 202:56-68. [PMID: 30510068 DOI: 10.4049/jimmunol.1801209] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Perivascular accumulation of lymphocytes can be a prominent histopathologic feature of various human inflammatory skin diseases. Select examples include systemic sclerosis, spongiotic dermatitis, and cutaneous lupus. Although a large body of work has described various aspects of the endothelial and vascular smooth muscle layers in these diseases, the outer adventitial compartment is poorly explored. The goal of the current study was to characterize perivascular adventitial fibroblast states in inflammatory human skin diseases and relate these states to perivascular lymphocyte accumulation. In normal skin, adventitial fibroblasts are distinguished by CD90 expression, and dense perivascular lymphocytic infiltrates are uncommon. In systemic sclerosis, this compartment expands, but lymphocyte infiltrates remain sparse. In contrast, perivascular adventitial fibroblast expression of VCAM1 is upregulated in spongiotic dermatitis and lupus and is associated with a dense perivascular T cell infiltrate. VCAM1 expression marks transitioned fibroblasts that show some resemblance to the reticular stromal cells in secondary lymphoid organs. Expanded adventitial compartments with perivascular infiltrates similar to the human settings were not seen in the inflamed murine dermis. This species difference may hinder the dissection of aspects of perivascular adventitial pathology. The altered perivascular adventitial compartment and its associated reticular network form a niche for lymphocytes and appear to be fundamental in the development of an inflammatory pattern.
Collapse
Affiliation(s)
- Alexander M S Barron
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| | - Julio C Mantero
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| | - Jonathan D Ho
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Banafsheh Nazari
- Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118
| | - Katharine L Horback
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| | - Jag Bhawan
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Robert Lafyatis
- Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118.,Division of Rheumatology and Clinical Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213
| | - Christina Lam
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Jeffrey L Browning
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118; .,Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118
| |
Collapse
|
39
|
Abstract
Autoantibodies (Ab) against the thyroid-stimulating hormone receptor (TSHR) are frequently found in autoimmune thyroid disease (AITD). Autoantibodies to the TSHR (anti-TSHR-Ab) may mimic or block the action of TSH or be functionally neutral. Measurement of anti-TSHR-Ab can be done either via competitive-binding immunoassays or with functional cell-based bioassays. Antibody-binding assays do not assess anti-TSHR-Ab functionality, but rather measure the concentration of total anti-TSHR binding activity. In contrast, functional cell-based bioassays indicate whether anti-TSHR-Ab have stimulatory or blocking activity. Historically bioassays for anti-TSHR-Ab were research tools and were used to study the pathophysiology of Graves' disease and Hashimoto's thyroiditis. In the past, bioassays for anti-TSHR-Abs were laborious and time-consuming and varied widely in performance from laboratory to laboratory. Recent advances in the development of cell-based assays, including the application of molecular engineering, have led to significant improvements that have enabled bioassays to be employed routinely in clinical laboratories. The prevalence and functional significance of TSHR blocking autoantibodies (TBAb) in autoimmune hypothyroidism has been less well investigated compared to TSHR stimulating Ab. There is an increasing body of data, however, that demonstrate the clinical utility and relevance of TBAb, and thus the importance of TBAb bioassays, in the diagnosis and management of patients with AITD. In the present review, we summarize the different methods used to measure TBAb, and discuss their prevalence and clinical relevance.
Collapse
Affiliation(s)
- Tanja Diana
- Molecular Thyroid Research Laboratory, Department of Medicine I, Johannes Gutenberg University (JGU) Medical Center, Mainz, Germany
| | - Paul D. Olivo
- Department of Molecular Microbiology, Washington University Medical School, St. Louis, Missouri, USA
| | - George J. Kahaly
- Molecular Thyroid Research Laboratory, Department of Medicine I, Johannes Gutenberg University (JGU) Medical Center, Mainz, Germany
- Correspondence Prof. George J. Kahaly JGU Medical CenterLangenbeckstraße 155131 MainzGermany+49-6131-17-2290+49-6131-17-3460
| |
Collapse
|
40
|
Gallotta M, Assi H, Degagné É, Kannan SK, Coffman RL, Guiducci C. Inhaled TLR9 Agonist Renders Lung Tumors Permissive to PD-1 Blockade by Promoting Optimal CD4 + and CD8 + T-cell Interplay. Cancer Res 2018; 78:4943-4956. [PMID: 29945961 DOI: 10.1158/0008-5472.can-18-0729] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/04/2018] [Accepted: 06/22/2018] [Indexed: 11/16/2022]
Abstract
Currently approved inhibitors of the PD-1/PD-L1 pathway represent a major advance for the treatment of lung cancers, yet they are ineffective in a majority of patients due to lack of preexisting T-cell reactivity. Here, we show that a TLR9 agonist delivered by inhalation is able to prime T-cell responses against poorly immunogenic lung tumors and to complement the effects of PD-1 blockade. Inhaled TLR9 agonist causes profound remodeling in tumor-bearing lungs, leading to the formation of tertiary lymphoid structures adjacent to the tumors, CD8+ T-cell infiltration into the tumors, dendritic cell expansion, and antibody production. Inhalation of TLR9 agonist also increased the pool of functional PD-1lowT-bethigh effector CD8+ T cells in tumor-bearing lungs. Effector CD8+ T cells generated by inhaled TLR9 agonist treatment were licensed by PD-1 blockade to become highly functional CTLs, leading to a durable rejection of both lung tumors and tumor lesions outside the lungs. CD4+ T cells activated in response to inhaled TLR9 play a critical role in this process by controlling the proliferation, preventing exhaustion, and guiding the differentiation of optimally functional CTLs. This study characterizes a strategy to apply localized TLR9 stimulation to a tumor type not accessible for direct injection, a strategy that may expand the therapeutic potential of PD-1 blockade in non-small cell lung cancer.Significance: These findings demonstrate that local delivery of a toll-like receptor 9 agonist can change the immune content of an entire organ and enhance the efficacy of immune checkpoint inhibition.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/17/4943/F1.large.jpg Cancer Res; 78(17); 4943-56. ©2018 AACR.
Collapse
Affiliation(s)
| | - Hikmat Assi
- Dynavax Technologies Corporation, Berkeley, California
| | | | | | | | | |
Collapse
|
41
|
Engelhard VH, Rodriguez AB, Mauldin IS, Woods AN, Peske JD, Slingluff CL. Immune Cell Infiltration and Tertiary Lymphoid Structures as Determinants of Antitumor Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:432-442. [PMID: 29311385 PMCID: PMC5777336 DOI: 10.4049/jimmunol.1701269] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/19/2017] [Indexed: 12/22/2022]
Abstract
Limited representation of intratumoral immune cells is a major barrier to tumor control. However, simply enhancing immune responses in tumor-draining lymph nodes or through adoptive transfer may not overcome the limited ability of tumor vasculature to support effector infiltration. An alternative is to promote a sustained immune response intratumorally. This idea has gained traction with the observation that many tumors are associated with tertiary lymphoid structures (TLS), which organizationally resemble lymph nodes. These peri- and intratumoral structures are usually, but not always, associated with positive prognoses in patients. Preclinical and clinical data support a role for TLS in modulating immunity in the tumor microenvironment. However, there appear to be varied functions of TLS, potentially based on their structure or location in relation to the tumor or the origin or location of the tumor itself. Understanding more about TLS development, composition, and function may offer new therapeutic opportunities to modulate antitumor immunity.
Collapse
Affiliation(s)
- Victor H Engelhard
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908;
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908; and
| | - Anthony B Rodriguez
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908; and
| | - Ileana S Mauldin
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Amber N Woods
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908; and
| | - J David Peske
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908; and
| | - Craig L Slingluff
- Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908
| |
Collapse
|
42
|
Mueller CG, Nayar S, Campos J, Barone F. Molecular and Cellular Requirements for the Assembly of Tertiary Lymphoid Structures. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:55-72. [PMID: 30155622 DOI: 10.1007/978-3-319-78127-3_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
At sites of chronic inflammation, recruited immune cells form structures that resemble secondary lymphoid organs (SLOs). Those are characterized by segregated areas of prevalent T- or B-cell aggregation, differentiation of high endothelial venules (HEVs) and local activation of resident stromal cells. B-cell proliferation and affinity maturation towards locally displayed autoantigens have been demonstrated at those sites, known as tertiary lymphoid structures (TLSs). TLS formation has been associated with local disease persistence and progression as well as increased systemic manifestations. While bearing a similar histological structure to SLO, the signals that regulate TLS and SLO formation can diverge, and a series of pro-inflammatory cytokines has been ascribed as responsible for TLS formation at different anatomical sites. Here we review the structural elements as well as the signals responsible for TLS aggregation, aiming to provide an overview to this complex immunological phenomenon.
Collapse
Affiliation(s)
- C G Mueller
- CNRS UPR 3572, Laboratory of Immunopathology and Therapeutic Chemistry/Laboratory of Excellence MEDALIS, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Strasbourg, France
| | - S Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham, UK
| | - J Campos
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham, UK
| | - F Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham, UK.
| |
Collapse
|
43
|
Tertiary Lymphoid Structures Among the World of Noncanonical Ectopic Lymphoid Organizations. Methods Mol Biol 2018; 1845:1-15. [PMID: 30141004 DOI: 10.1007/978-1-4939-8709-2_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Tertiary lymphoid structures (TLOs), also known as ectopic lymphoid structures, are associated with chronic infections and inflammatory diseases. Despite their association with pathology, these structures are actually a normal, albeit transient, component of the immune system and facilitate local immune responses that are meant to mitigate inflammation and resolve infection. Many of the mechanisms controlling the formation and function of tertiary lymphoid structures have been identified, in part by experimentally triggering their formation using defined stimuli under controlled conditions. Here, we introduce the experimental and pathological conditions in which tertiary lymphoid tissues are formed, describe the mechanisms linked to their formation, and discuss their functions in the context of both infection and inflammation.
Collapse
|
44
|
Mueller CG, Nayar S, Gardner D, Barone F. Cellular and Vascular Components of Tertiary Lymphoid Structures. Methods Mol Biol 2018; 1845:17-30. [PMID: 30141005 DOI: 10.1007/978-1-4939-8709-2_2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Inflammatory immune cells recruited at the site of chronic inflammation form structures that resemble secondary lymphoid organs (SLO). These are characterized by segregated areas of prevalent T- or B-cell aggregation, differentiation of high endothelial venules, and local activation of resident stromal cells, including lymphatic endothelial cells. B-cell proliferation and affinity maturation toward locally displayed autoantigens have been demonstrated at these sites, known as tertiary lymphoid structures (TLS). TLS formation during chronic inflammation has been associated with local disease persistence and progression, as well as increased systemic manifestations. While bearing a similar histological structure to SLO, the signals that regulate TLS and SLO formation can diverge and a series of pro-inflammatory cytokines have been ascribed as responsible for TLS formation at different anatomical sites. Moreover, for a long time the structural compartment that regulates TLS homeostasis, including survival and recirculation of leucocytes has been neglected. In this chapter, we summarize the novel data available on TLS formation, structural organization, and the functional and anatomical links connecting TLS and SLOs.
Collapse
Affiliation(s)
- Christopher George Mueller
- Laboratoire d'Immunologie, Immunopathologie et Chimie Thérapeutique, Institut de Biologie Moléculaire et Cellulaire (IBMC), CNRS UPR 3572, University of Strasbourg, Strasbourg, France
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - David Gardner
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Francesca Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK.
| |
Collapse
|
45
|
Colbeck EJ, Ager A, Gallimore A, Jones GW. Tertiary Lymphoid Structures in Cancer: Drivers of Antitumor Immunity, Immunosuppression, or Bystander Sentinels in Disease? Front Immunol 2017; 8:1830. [PMID: 29312327 PMCID: PMC5742143 DOI: 10.3389/fimmu.2017.01830] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/04/2017] [Indexed: 12/16/2022] Open
Abstract
Secondary lymphoid organs are integral to initiation and execution of adaptive immune responses. These organs provide a setting for interactions between antigen-specific lymphocytes and antigen-presenting cells recruited from local infected or inflamed tissues. Secondary lymphoid organs develop as a part of a genetically preprogrammed process during embryogenesis. However, organogenesis of secondary lymphoid tissues can also be recapitulated in adulthood during de novo lymphoid neogenesis of tertiary lymphoid structures (TLSs). These ectopic lymphoid-like structures form in the inflamed tissues afflicted by various pathological conditions, including cancer, autoimmunity, infection, or allograft rejection. Studies are beginning to shed light on the function of such structures in different disease settings, raising important questions regarding their contribution to progression or resolution of disease. Data show an association between the tumor-associated TLSs and a favorable prognosis in various types of human cancer, attracting the speculation that TLSs support effective local antitumor immune responses. However, definitive evidence for the role for TLSs in fostering immune responses in vivo are lacking, with current data remaining largely correlative by nature. In fact, some more recent studies have even demonstrated an immunosuppressive, tumor-promoting role for cancer-associated TLSs. In this review, we will discuss what is known about the development of cancer-associated TLSs and the current understanding of their potential role in the antitumor immune response.
Collapse
Affiliation(s)
| | - Ann Ager
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Awen Gallimore
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Gareth Wyn Jones
- Division of Infection and Immunity, School of Medicine and Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
46
|
Alsughayyir J, Pettigrew GJ, Motallebzadeh R. Spoiling for a Fight: B Lymphocytes As Initiator and Effector Populations within Tertiary Lymphoid Organs in Autoimmunity and Transplantation. Front Immunol 2017; 8:1639. [PMID: 29218052 PMCID: PMC5703719 DOI: 10.3389/fimmu.2017.01639] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 11/09/2017] [Indexed: 12/13/2022] Open
Abstract
Tertiary lymphoid organs (TLOs) develop at ectopic sites within chronically inflamed tissues, such as in autoimmunity and rejecting organ allografts. TLOs differ structurally from canonical secondary lymphoid organs (SLOs), in that they lack a mantle zone and are not encapsulated, suggesting that they may provide unique immune function. A notable feature of TLOs is the frequent presence of structures typical of germinal centers (GCs). However, little is known about the role of such GCs, and in particular, it is not clear if the B cell response within is autonomous, or whether it synergizes with concurrent responses in SLOs. This review will discuss ectopic lymphoneogenesis and the role of the B cell in TLO formation and subsequent effector output in the context of autoimmunity and transplantation, with particular focus on the contribution of ectopic GCs to affinity maturation in humoral immune responses and to the potential breakdown of self-tolerance and development of humoral autoimmunity.
Collapse
Affiliation(s)
- Jawaher Alsughayyir
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Gavin J Pettigrew
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Reza Motallebzadeh
- Division of Surgery and Interventional Science, University College London, London, United Kingdom.,Institute of Immunity and Transplantation, University College London, London, United Kingdom.,Department of Nephrology, Urology and Transplantation, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
47
|
Tang H, Zhu M, Qiao J, Fu YX. Lymphotoxin signalling in tertiary lymphoid structures and immunotherapy. Cell Mol Immunol 2017; 14:809-818. [PMID: 28413217 PMCID: PMC5649108 DOI: 10.1038/cmi.2017.13] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/24/2017] [Accepted: 01/24/2017] [Indexed: 12/16/2022] Open
Abstract
Tertiary lymphoid structures (TLS) often develop at sites of persistent inflammation, including cancers and autoimmune diseases. In most cases, the presence of TLS correlates with active immune responses. Because of their proximity to pathological loci, TLS are an intriguing target for the manipulation of immune responses. For several years, it has become clear that lymphotoxin (LT) signalling plays critical roles in lymphoid tissue organogenesis and maintenance. In the current review, we will discuss the role of LT signalling in the development of TLS. With a focus on cancers and autoimmune diseases, we will highlight the correlations between TLS and disease progression. We will also discuss the current efforts and potential directions for manipulating TLS for immunotherapies.
Collapse
Affiliation(s)
- Haidong Tang
- Department of Pathology, University of Texas, Southwestern Medical Center, Dallas, TX 75235, USA
| | - Mingzhao Zhu
- IBP-UTSW Joint Immunotherapy Group, Chinese Academy of Science, Key Laboratory for Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jian Qiao
- Department of Pathology, University of Texas, Southwestern Medical Center, Dallas, TX 75235, USA
| | - Yang-Xin Fu
- Department of Pathology, University of Texas, Southwestern Medical Center, Dallas, TX 75235, USA
- IBP-UTSW Joint Immunotherapy Group, Chinese Academy of Science, Key Laboratory for Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
48
|
Delivanis DA, Gustafson MP, Bornschlegl S, Merten MM, Kottschade L, Withers S, Dietz AB, Ryder M. Pembrolizumab-Induced Thyroiditis: Comprehensive Clinical Review and Insights Into Underlying Involved Mechanisms. J Clin Endocrinol Metab 2017; 102:2770-2780. [PMID: 28609832 PMCID: PMC5546861 DOI: 10.1210/jc.2017-00448] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/02/2017] [Indexed: 12/21/2022]
Abstract
CONTEXT Thyroid immune-related adverse events (irAEs) in patients treated with programmed death receptor-1 (PD-1) blockade are increasingly recognized as one of the most common adverse effects. Our aim was to determine the incidence and examine the potential mechanisms of anti-PD-1-induced thyroid irAEs. DESIGN Single-center, retrospective cohort study. PATIENTS AND MEASUREMENTS We studied 93 patients with advanced cancer (ages 24 to 82 years; 60% males) who received at least one infusion of pembrolizumab. Thyroid test results and thyroid imaging modalities were reviewed. Comprehensive 10-color flow cytometry of peripheral blood was performed. RESULTS Thirteen (14%) thyroid irAEs were observed. Thyroiditis occurred in seven patients (54%), from which four recovered. New onset of hypothyroidism overt/subclinical developed in three patients. Levothyroxine dosing required doubling in three patients with a known history of hypothyroidism. Thyroperoxidase antibodies were positive in the minority of the patients [4/13 (31%)] and diffuse increased 18fludeoxyglucose uptake of the thyroid gland was observed in the majority [7/11 (64%)] of patients. We observed more circulating CD56+CD16+ natural killer (NK) cells and an elevated HLA-DR surface expression in the inflammatory intermediate CD14+CD16+ monocytes in anti-PD-1-treated patients. CONCLUSIONS Thyroid dysfunction is common in cancer patients treated with pembrolizumab. Reversible destructive thyroiditis and overt hypothyroidism are the most common clinical presentations. The mechanism of thyroid destruction appears independent of thyroid autoantibodies and may include T cell, NK cell, and/or monocyte-mediated pathways. Because the thyroid is a frequent target of anti-PD-1 therapies, patients with therapeutically refractory thyroid cancer may be ideal candidates for this treatment.
Collapse
Affiliation(s)
- Danae A. Delivanis
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota 55905
| | - Michael P. Gustafson
- Human Cell Therapy Laboratory, Division of Transfusion Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905
| | - Svetlana Bornschlegl
- Department of Laboratory Medicine & Pathology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Michele M. Merten
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota 55905
| | - Lisa Kottschade
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota 55905
| | - Sarah Withers
- Department of Laboratory Medicine & Pathology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Allan B. Dietz
- Human Cell Therapy Laboratory, Division of Transfusion Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905
| | - Mabel Ryder
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota 55905
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
49
|
Gu-Trantien C, Migliori E, Buisseret L, de Wind A, Brohée S, Garaud S, Noël G, Dang Chi VL, Lodewyckx JN, Naveaux C, Duvillier H, Goriely S, Larsimont D, Willard-Gallo K. CXCL13-producing TFH cells link immune suppression and adaptive memory in human breast cancer. JCI Insight 2017; 2:91487. [PMID: 28570278 DOI: 10.1172/jci.insight.91487] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 04/25/2017] [Indexed: 02/06/2023] Open
Abstract
T follicular helper cells (TFH cells) are important regulators of antigen-specific B cell responses. The B cell chemoattractant CXCL13 has recently been linked with TFH cell infiltration and improved survival in human cancer. Although human TFH cells can produce CXCL13, their immune functions are currently unknown. This study presents data from human breast cancer, advocating a role for tumor-infiltrating CXCL13-producing (CXCR5-) TFH cells, here named TFHX13 cells, in promoting local memory B cell differentiation. TFHX13 cells potentially trigger tertiary lymphoid structure formation and thereby generate germinal center B cell responses at the tumor site. Follicular DCs are not potent CXCL13 producers in breast tumor tissues. We used the TFH cell markers PD-1 and ICOS to identify distinct effector and regulatory CD4+ T cell subpopulations in breast tumors. TFHX13 cells are an important component of the PD-1hiICOSint effector subpopulation and coexpanded with PD-1intICOShiFOXP3hi Tregs. IL2 deprivation induces CXCL13 expression in vitro with a synergistic effect from TGFβ1, providing insight into TFHX13 cell differentiation in response to Treg accumulation, similar to conventional TFH cell responses. Our data suggest that human TFHX13 cell differentiation may be a key factor in converting Treg-mediated immune suppression to de novo activation of adaptive antitumor humoral responses in the chronic inflammatory breast cancer microenvironment.
Collapse
Affiliation(s)
| | | | - Laurence Buisseret
- Molecular Immunology Unit.,Breast Cancer Translational Research Laboratory
| | | | | | | | | | | | | | | | - Hugues Duvillier
- Flow Cytometry Core Facility, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Stanislas Goriely
- Welbio and Institute for Medical Immunology, Université Libre de Bruxelles, Brussels, Belgium
| | | | | |
Collapse
|
50
|
Humby FC, Al Balushi F, Lliso G, Cauli A, Pitzalis C. Can Synovial Pathobiology Integrate with Current Clinical and Imaging Prediction Models to Achieve Personalized Health Care in Rheumatoid Arthritis? Front Med (Lausanne) 2017; 4:41. [PMID: 28516086 PMCID: PMC5413506 DOI: 10.3389/fmed.2017.00041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/27/2017] [Indexed: 11/13/2022] Open
Abstract
Although great progress has been made in the past decade toward understanding the pathogenesis of rheumatoid arthritis (RA), clinicians remain some distance from a goal of personalized health care. The capacity to diagnose RA early, predict prognosis, and moreover predict response to biologic therapies has been a research focus for many years. How currently available clinical prediction models can facilitate such goals is reviewed in this article. In addition, the role of current imaging techniques in this regard is also discussed. Finally, the authors review the current literature regarding synovial biomarkers and consider whether integration of synovial pathobiology into clinical prediction algorithms may enhance their predictive value.
Collapse
Affiliation(s)
- Frances Claire Humby
- Department of Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Gloria Lliso
- Department of Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Alberto Cauli
- Dipartimento di Scienze Mediche, Facoltà di Medicina e Chirurgia, Università degli Studi di Cagliari, Cagliari, Italy
| | - Costantino Pitzalis
- Department of Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| |
Collapse
|