1
|
Kim HM, Bruno TC. An Introduction to Tertiary Lymphoid Structures in Cancer. Methods Mol Biol 2025; 2864:1-19. [PMID: 39527214 DOI: 10.1007/978-1-0716-4184-2_1] [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
Immunotherapy has revolutionized therapeutics for cancer patients, which signifies the importance of effective antitumor immunity in combatting cancer. However, the benefit of immunotherapies is limited to specific patient populations and tumor types, suggesting the overt need for new immunotherapeutic targets. Tertiary lymphoid structures (TLS) are ectopic lymph node-like structures that develop at the sites of chronic inflammation such as cancer. TLS are correlated with favorable clinical outcomes across multiple solid tumors and are associated with increased tumor-infiltrating lymphocytes (TILs), particularly effector memory CD8+ T cells. Despite strong clinical data in humans, there are still major knowledge gaps on the function of TLS in cancer. Herein, we highlight the known biology and clinical impact of TLS, which offer further evidence to harness TLS for improved immunotherapeutics.
Collapse
Affiliation(s)
- Hye Mi Kim
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Tumor Microenvironment Center (TMC), UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tullia C Bruno
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Tumor Microenvironment Center (TMC), UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Cancer Immunology and Immunotherapy Program (CIIP), UPMC Hillman Cancer Center, Pittsburgh, PA, 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
|
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
|
4
|
Liao J, Yu X, Huang Z, He Q, Yang J, Zhang Y, Chen J, Song W, Luo J, Tao Q. Chemokines and lymphocyte homing in Sjögren's syndrome. Front Immunol 2024; 15:1345381. [PMID: 38736890 PMCID: PMC11082322 DOI: 10.3389/fimmu.2024.1345381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
Sjögren's syndrome (SS) is a chronic systemic autoimmune disease that typically presents with lymphocyte, dendritic cell, and macrophage infiltration of exocrine gland ducts and the formation of ectopic germinal centers. The interactions of lymphocyte homing receptors and addressins and chemokines and their receptors, such as α4β7/MAdCAM-1, LFA-1/ICAM-1, CXCL13/CXCR5, CCL25/CCR9, CX3CL1/CX3CR1, play important roles in the migration of inflammatory cells to the focal glands and the promotion of ectopic germinal center formation in SS. A variety of molecules have been shown to be involved in lymphocyte homing, including tumor necrosis factor-α, interferon (IFN)-α, IFN-β, and B cell activating factor. This process mainly involves the Janus kinase-signal transducer and activator of transcription signaling pathway, lymphotoxin-β receptor pathway, and nuclear factor-κB signaling pathway. These findings have led to the development of antibodies to cell adhesion molecules, antagonists of chemokines and their receptors, compounds interfering with chemokine receptor signaling, and gene therapies targeting chemokines and their receptors, providing new targets for the treatment of SS in humans. The aim of this study was to explore the relationship between lymphocyte homing and the pathogenesis of SS, and to provide a review of recent studies addressing lymphocyte homing in targeted therapy for SS.
Collapse
Affiliation(s)
- Jiahe Liao
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Traditional Chinese Medicine Department of Rheumatism, China-Japan Friendship Hospital, Beijing, China
| | - Xinbo Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Traditional Chinese Medicine Department of Rheumatism, China-Japan Friendship Hospital, Beijing, China
| | - Ziwei Huang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Traditional Chinese Medicine Department of Rheumatism, China-Japan Friendship Hospital, Beijing, China
| | - Qian He
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Traditional Chinese Medicine Department of Rheumatism, China-Japan Friendship Hospital, Beijing, China
| | - Jianying Yang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Traditional Chinese Medicine Department of Rheumatism, China-Japan Friendship Hospital, Beijing, China
| | - Yan Zhang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Traditional Chinese Medicine Department of Rheumatism, China-Japan Friendship Hospital, Beijing, China
| | - Jiaqi Chen
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Traditional Chinese Medicine Department of Rheumatism, China-Japan Friendship Hospital, Beijing, China
| | - Weijiang Song
- Traditional Chinese Medicine Department, Peking University Third Hospital, Beijing, China
| | - Jing Luo
- Traditional Chinese Medicine Department of Rheumatism, China-Japan Friendship Hospital, Beijing, China
- Beijing Key Laboratory of Immune Inflammatory Disease, China-Japan Friendship Hospital, Beijing, China
| | - Qingwen Tao
- Traditional Chinese Medicine Department of Rheumatism, China-Japan Friendship Hospital, Beijing, China
- Beijing Key Laboratory of Immune Inflammatory Disease, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
5
|
Zhou X, Xu D, Li M, Zeng X. New investigational drugs to treat Sjogren's syndrome: lessons learnt from immunology. Expert Opin Investig Drugs 2024; 33:105-114. [PMID: 38293750 DOI: 10.1080/13543784.2024.2312216] [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: 10/28/2023] [Accepted: 01/26/2024] [Indexed: 02/01/2024]
Abstract
INTRODUCTION Sjögren's syndrome is a heterogeneous autoimmune condition that impairs quality of life because of dryness, fatigue, pain, and systemic involvements. Current treatment largely depends on empirical evidence, with no effective therapy approved. Clinical trials on targeted drugs often fail to report efficacy due to common factors. AREAS COVERED This review summarizes the pathogenesis and what caused the failure of new investigational drugs in clinical trials, highlighting solutions for more effective investigations, with greater consistency between research outcomes, clinical use, and patient needs. EXPERT OPINION Unlinked pathobiology with symptoms resulted in misidentified targets and disappointing trials. Useful stratification tools are necessary for the heterogeneous SS patients. Composite endpoints or improvements in ESSDAI scores are needed, considering the high placebo response, and the unbalance between symptom burden and disease activity. Compared to classic biologics, targeted cell therapy will be a more promising field of investigation in the coming years.
Collapse
Affiliation(s)
- Xingyu Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Dong Xu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| |
Collapse
|
6
|
M. S. Barron A, Fabre T, De S. Distinct fibroblast functions associated with fibrotic and immune-mediated inflammatory diseases and their implications for therapeutic development. F1000Res 2024; 13:54. [PMID: 38681509 PMCID: PMC11053351 DOI: 10.12688/f1000research.143472.1] [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] [Accepted: 12/28/2023] [Indexed: 05/01/2024] Open
Abstract
Fibroblasts are ubiquitous cells that can adopt many functional states. As tissue-resident sentinels, they respond to acute damage signals and shape the earliest events in fibrotic and immune-mediated inflammatory diseases. Upon sensing an insult, fibroblasts produce chemokines and growth factors to organize and support the response. Depending on the size and composition of the resulting infiltrate, these activated fibroblasts may also begin to contract or relax thus changing local stiffness within the tissue. These early events likely contribute to the divergent clinical manifestations of fibrotic and immune-mediated inflammatory diseases. Further, distinct changes to the cellular composition and signaling dialogue in these diseases drive progressive fibroblasts specialization. In fibrotic diseases, fibroblasts support the survival, activation and differentiation of myeloid cells, granulocytes and innate lymphocytes, and produce most of the pathogenic extracellular matrix proteins. Whereas, in immune-mediated inflammatory diseases, sequential accumulation of dendritic cells, T cells and B cells programs fibroblasts to support local, destructive adaptive immune responses. Fibroblast specialization has clear implications for the development of effective induction and maintenance therapies for patients with these clinically distinct diseases.
Collapse
Affiliation(s)
- Alexander M. S. Barron
- Inflammation & Immunology Research Unit, Pfizer, Inc., Cambridge, Massachusetts, 02139, USA
| | - Thomas Fabre
- Inflammation & Immunology Research Unit, Pfizer, Inc., Cambridge, Massachusetts, 02139, USA
| | - Saurav De
- Inflammation & Immunology Research Unit, Pfizer, Inc., Cambridge, Massachusetts, 02139, USA
| |
Collapse
|
7
|
Dong Y, Wang T, Wu H. Tertiary lymphoid structures in autoimmune diseases. Front Immunol 2024; 14:1322035. [PMID: 38259436 PMCID: PMC10800951 DOI: 10.3389/fimmu.2023.1322035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Tertiary lymphoid structures (TLSs) are organized lymphoid-like aggregations in non-lymphoid tissues. Tissues with chronic and persistent inflammation infiltration may drive and form ectopic germinal center-like structures, which are very common in autoimmune diseases, chronic infections, and tumor microenvironments. However, the mechanisms governing the formation of TLSs are still being explored. At present, it is not clear whether the formation of TLSs is associated with local uncontrolled immune inflammatory responses. While TLSs suggest a good prognosis in tumors, the opposite is true in autoimmune diseases. This review article will discuss the current views on initiating and maintaining TLSs and the potential therapeutic target in autoimmune diseases.
Collapse
Affiliation(s)
- Yuanji Dong
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ting Wang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huaxiang Wu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Li CR. Role of lymphotoxin alpha as a new molecular biomarker in revolutionizing tear diagnostic testing for dry eye disease. Int J Ophthalmol 2023; 16:1883-1889. [PMID: 38028523 PMCID: PMC10626360 DOI: 10.18240/ijo.2023.11.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/24/2023] [Indexed: 12/01/2023] Open
Abstract
Dry eye disease (DED), primarily classified as multifactorial ocular surface disorder, afflicts tens of millions of individuals worldwide, adversely impacting their quality of life. Extensive research has been conducted on tear film analysis over the past decades, offering a range of tests to evaluate its volume, health, and integrity. Yet, early diagnosis and effective treatment for DED continue to pose significant challenges in clinical settings. Nevertheless, by recognizing key phenomena in DED such as ocular surface inflammation, hyperosmolarity, and tear film instability, this article provides a comprehensive overview of both traditional and recently developed methods for diagnosing and monitoring DED. The information serves as a valuable resource not only for clinical diagnosis but also for further research into DED.
Collapse
Affiliation(s)
- Chao-Ran Li
- Tianjin Medical University Eye Hospital, Tianjin 310011, China
| |
Collapse
|
9
|
Mohammadnezhad L, Shekarkar Azgomi M, La Manna MP, Guggino G, Botta C, Dieli F, Caccamo N. B-Cell Receptor Signaling Is Thought to Be a Bridge between Primary Sjogren Syndrome and Diffuse Large B-Cell Lymphoma. Int J Mol Sci 2023; 24:ijms24098385. [PMID: 37176092 PMCID: PMC10179133 DOI: 10.3390/ijms24098385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Primary Sjogren syndrome (pSS) is the second most common autoimmune disorder worldwide, which, in the worst scenario, progresses to Non-Hodgkin Lymphoma (NHL). Despite extensive studies, there is still a lack of knowledge about developing pSS for NHL. This study focused on cells' signaling in pSS progression to the NHL type of diffuse large B-cell lymphoma (DLBCL). Using bulk RNA and single cell analysis, we found five novel pathologic-independent clusters in DLBCL based on cells' signaling. B-cell receptor (BCR) signaling was identified as the only enriched signal in DLBCL and pSS peripheral naive B-cells or salivary gland-infiltrated cells. The evaluation of the genes in association with BCR has revealed that targeting CD79A, CD79B, and LAMTOR4 as the shared genes can provide novel biomarkers for pSS progression into lymphoma.
Collapse
Affiliation(s)
- Leila Mohammadnezhad
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AOUP Paolo Giaccone, 90127 Palermo, Italy
- Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, 90127 Palermo, Italy
| | - Mojtaba Shekarkar Azgomi
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AOUP Paolo Giaccone, 90127 Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, 90127 Palermo, Italy
| | - Marco Pio La Manna
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AOUP Paolo Giaccone, 90127 Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, 90127 Palermo, Italy
| | - Giuliana Guggino
- Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, 90127 Palermo, Italy
| | - Cirino Botta
- Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, 90127 Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AOUP Paolo Giaccone, 90127 Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, 90127 Palermo, Italy
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AOUP Paolo Giaccone, 90127 Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
10
|
Galletti JG, Scholand KK, Trujillo-Vargas CM, Yu Z, Mauduit O, Delcroix V, Makarenkova HP, de Paiva CS. Ectopic lymphoid structures in the aged lacrimal glands. Clin Immunol 2023; 248:109251. [PMID: 36740002 PMCID: PMC10323865 DOI: 10.1016/j.clim.2023.109251] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
Aging is a complex biological process in which many organs are pathologically affected. We previously reported that aged C57BL/6J had increased lacrimal gland (LG) lymphoid infiltrates that suggest ectopic lymphoid structures. However, these ectopic lymphoid structures have not been fully investigated. Using C57BL/6J mice of different ages, we analyzed the transcriptome of aged murine LGs and characterized the B and T cell populations. Age-related changes in the LG include increased differentially expressed genes associated with B and T cell activation, germinal center formation, and infiltration by marginal zone-like B cells. We also identified an age-related increase in B1+ cells and CD19+B220+ cells. B220+CD19+ cells were GL7+ (germinal center-like) and marginal zone-like and progressively increased with age. There was an upregulation of transcripts related to T follicular helper cells, and the number of these cells also increased as mice aged. Compared to a mouse model of Sjögren syndrome, aged LGs have similar transcriptome responses but also unique ones. And lastly, the ectopic lymphoid structures in aged LGs are not exclusive to a specific mouse background as aged diverse outbred mice also have immune infiltration. Altogether, this study identifies a profound change in the immune landscape of aged LGs where B cells become predominant. Further studies are necessary to investigate the specific function of these B cells during the aged LGs.
Collapse
Affiliation(s)
- Jeremias G Galletti
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, USA; Institute of Experimental Medicine (CONICET), National Academy of Medicine of Buenos Aires, Buenos Aires, Argentina
| | - Kaitlin K Scholand
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, USA; Biochemistry and Cell Biology Graduate Program, Department of BioSciences, Rice University, Houston, TX, USA.
| | - Claudia M Trujillo-Vargas
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, USA; Grupo de Inmunodeficiencias Primarias, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellín, Colombia.
| | - Zhiyuan Yu
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, USA.
| | - Olivier Mauduit
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Vanessa Delcroix
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Helen P Makarenkova
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Cintia S de Paiva
- Ocular Surface Center, Department of Ophthalmology, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
11
|
Ghobadinezhad F, Ebrahimi N, Mozaffari F, Moradi N, Beiranvand S, Pournazari M, Rezaei-Tazangi F, Khorram R, Afshinpour M, Robino RA, Aref AR, Ferreira LMR. The emerging role of regulatory cell-based therapy in autoimmune disease. Front Immunol 2022; 13:1075813. [PMID: 36591309 PMCID: PMC9795194 DOI: 10.3389/fimmu.2022.1075813] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Autoimmune disease, caused by unwanted immune responses to self-antigens, affects millions of people each year and poses a great social and economic burden to individuals and communities. In the course of autoimmune disorders, including rheumatoid arthritis, systemic lupus erythematosus, type 1 diabetes mellitus, and multiple sclerosis, disturbances in the balance between the immune response against harmful agents and tolerance towards self-antigens lead to an immune response against self-tissues. In recent years, various regulatory immune cells have been identified. Disruptions in the quality, quantity, and function of these cells have been implicated in autoimmune disease development. Therefore, targeting or engineering these cells is a promising therapeutic for different autoimmune diseases. Regulatory T cells, regulatory B cells, regulatory dendritic cells, myeloid suppressor cells, and some subsets of innate lymphoid cells are arising as important players among this class of cells. Here, we review the roles of each suppressive cell type in the immune system during homeostasis and in the development of autoimmunity. Moreover, we discuss the current and future therapeutic potential of each one of these cell types for autoimmune diseases.
Collapse
Affiliation(s)
- Farbod Ghobadinezhad
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran,Universal Scientific Education and Research Network (USERN) Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nasim Ebrahimi
- Division of Genetics, Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Fatemeh Mozaffari
- Department of Nutrition, School of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Neda Moradi
- Division of Biotechnology, Department of Cell and Molecular Biology and Microbiology, Nourdanesh Institute of Higher Education, University of Meymeh, Isfahan, Iran
| | - Sheida Beiranvand
- Department of Biology, Faculty of Basic Sciences, Islamic Azad University, Shahrekord, Iran
| | - Mehran Pournazari
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Roya Khorram
- Bone and Joint Diseases Research Center, Department of Orthopedic Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maral Afshinpour
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
| | - Rob A. Robino
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States,Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States,Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Amir Reza Aref
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States,Xsphera Biosciences, Boston, MA, United States,*Correspondence: Leonardo M. R. Ferreira, ; Amir Reza Aref,
| | - Leonardo M. R. Ferreira
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States,Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States,Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States,*Correspondence: Leonardo M. R. Ferreira, ; Amir Reza Aref,
| |
Collapse
|
12
|
Bagavant H, Trzeciak M, Biswas I, Papinska JA, Cizio K, Deshmukh US. Antibody deposition on vascular endothelial cells contributes to localized inflammation in salivary glands. J Oral Pathol Med 2022; 51:674-677. [PMID: 35766433 PMCID: PMC9388553 DOI: 10.1111/jop.13330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/18/2022] [Accepted: 06/25/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Oral and ocular dryness due to reduced saliva and tear production, exocrine gland inflammation, and autoantibodies to multiple cellular proteins are the cardinal features of Sjögren's Disease. Among the autoantibody specificities, anti-Ro52 is linked with higher disease severity. We have previously reported that mice immunized with recombinant Ro52 developed IgG deposits in salivary and lacrimal glands and showed reduced saliva and tear production. Furthermore, passive transfer of sera from Ro52-immunized mice rapidly induced glandular dysfunction without immune cell infiltration in recipient mice. METHODS To identify mechanisms driving antibody-mediated salivary gland dysfunction, hyperimmune rabbit antiserum to mouse Ro52 was passively transferred into NZM2758 female mice, pretreated with alum adjuvant. Alum-pretreated mice given hyperimmune rabbit antiserum to maltose-binding protein served as controls. Antibody deposition and its distribution in the salivary glands were studied by immunofluorescence staining for rabbit IgG, nerve fibers, and endothelial cells. The nCounter inflammation panel was used to determine differentially expressed genes in the salivary gland. RESULTS Rabbit IgG deposits were detected in salivary glands of anti-Ro52 immune sera recipients. The rabbit IgG was present on the endothelial cells in small blood vessels, and it did not co-localize with nerve fibers. Ingenuity pathway analysis of the gene expression dataset predicted the canonical vascular endothelial growth factor (VEGF) pathway as the most activated and Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) as the most inhibited pathway in the salivary glands of anti-Ro52 sera recipients. CONCLUSION Our study suggests that autoantibody deposition on salivary gland endothelial cells might play a critical role in the pathogenesis of Sjögren's Disease.
Collapse
Affiliation(s)
- Harini Bagavant
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Marta Trzeciak
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Indranil Biswas
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Joanna A Papinska
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Katarzyna Cizio
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Umesh S Deshmukh
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| |
Collapse
|
13
|
Kim DS, Na HS, Cho KH, Lee KH, Choi J, Kwok SK, Bae YS, Cho ML, Park SH. Sphingosylphosphorylcholine ameliorates experimental sjögren's syndrome by regulating salivary gland inflammation and hypofunction, and regulatory B cells. Immunol Lett 2022; 248:62-69. [PMID: 35732207 DOI: 10.1016/j.imlet.2022.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 06/14/2022] [Accepted: 06/18/2022] [Indexed: 11/24/2022]
Abstract
Sjögren syndrome (SS) is an autoimmune disease in which immune cells infiltrate the exocrine gland. Since SS is caused by a disorder of the immune system, treatments should regulate the immune response. Sphingosylphosphorylcholine (SPC) is a sphingolipid that mediates cellular signaling. In immune cells, SPC has several immunomodulatory functions. Accordingly, this study verifies the immunomodulatory ability and therapeutic effect of SPC in SS. To understand the function of SPC in SS, we treated SPC in female NOD/ShiJcl (NOD) mice. The mice were monitored for 10 weeks, and inflammation in the salivary glands was checked. After SPC treatment, we detected the expression of regulatory B (Breg) cells in mouse splenocytes and the level of salivary secretion-related genes in human submandibular gland (HSG) cells. Salivary flow rate was maintained in the SPC-treated group compared to the vehicle-treated group, and inflammation in the salivary gland tissues was relieved by SPC. SPC treatment in mouse cells and HSG cells enhanced Breg cells and salivary secretion markers, respectively. This study revealed that SPC can be considered as a new therapeutic agent against SS.
Collapse
Affiliation(s)
- Da Som Kim
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Hyun Sik Na
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Keun-Hyung Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Kun Hee Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - JeongWon Choi
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Ki Kwok
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Yoe-Sik Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea.
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| | - Sung-Hwan Park
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 222, Banpo-Daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| |
Collapse
|
14
|
Huang MW, Stock AD, Putterman C. CXCL13 Neutralization Attenuates Neuropsychiatric Manifestations in Lupus-Prone Mice. Front Immunol 2021; 12:763065. [PMID: 34868008 PMCID: PMC8633419 DOI: 10.3389/fimmu.2021.763065] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/04/2021] [Indexed: 01/02/2023] Open
Abstract
Neuropsychiatric lupus (NPSLE), the nervous system presentation of systemic lupus erythematosus (SLE), remains challenging to treat due to its unclear pathogenesis and lack of available targeted therapies. A potential contributor to disease progression is brain tertiary lymphoid structures (TLS); these ectopic lymphoid follicles that can develop tissue-targeted antibodies have recently been described in the MRL/lpr lupus mouse strain, a classic model for studying NPSLE. The brains of MRL/lpr mice show a significant increase of CXCL13, an important chemokine in lymphoid follicle formation and retention that may also play a role in the disease progression of NPSLE. The aim of the present study was to inhibit CXCL13 and examine the effect of this intervention on lymphoid formation and the development of neurobehavioral manifestations in lupus mice. Female MRL/lpr mice were injected with an anti-CXCL13 antibody, an IgG1 isotype-matched antibody, or PBS either three times a week for 12 weeks intraperitoneally (IP) starting at 6-8 weeks of age, or continuously intracerebroventricularly (ICV) with an osmotic pump over a two-week period starting at 15 weeks of age. Cognitive dysfunction and depression-like behavior were assessed at the end of treatment. When treatment was delivered IP, anti-CXCL13 treated mice showed significant improvement in cognitive function when compared to control treated mice. Depression-like behavior was attenuated as well. Furthermore, mice that received anti-CXCL13 by the ICV route showed similar beneficial effects. However, the extent of lymphocyte infiltration into the brain and the general composition of the aggregates were not substantively changed by anti-CXCL13 irrespective of the mode of administration. Nevertheless, analysis of brain gene expression in anti-CXCL13 treated mice showed significant differences in key immunological and neuro-inflammatory pathways that most likely explained the improvement in the behavioral phenotype. Our results indicate that CXCL13 affects the behavioral manifestations in the MRL/lpr strain and is important to the pathogenesis of murine NPSLE, suggesting it as a potential therapeutic target.
Collapse
Affiliation(s)
- Michelle W Huang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ariel D Stock
- Department of Neurological Surgery, Montefiore Medical Center, Bronx, NY, United States
| | - Chaim Putterman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.,Division of Rheumatology, Albert Einstein College of Medicine, Bronx, NY, United States.,Azrieli Faculty of Medicine, Bar-Ilan University, Zefat, Israel.,Galilee Medical Center Research Institute, Nahariya, Israel
| |
Collapse
|
15
|
Targeted Therapy for Primary Sjögren's Syndrome: Where are We Now? BioDrugs 2021; 35:593-610. [PMID: 34731460 DOI: 10.1007/s40259-021-00505-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2021] [Indexed: 10/19/2022]
Abstract
Primary Sjögren's syndrome (pSS) is an autoimmune exocrinopathy characterized by dryness symptoms. This review briefly describes recent advances in the targeted therapies for pSS. Biologics evaluated for pSS treatment mainly include B cell-depleting agents, inhibitors of B cell activation, and agents that target co-signaling molecules or proinflammatory cytokines. Small molecule inhibitors that target signaling pathways have also been evaluated. However, current evidence for the efficacy of targeted therapies in pSS is still sparse. Although ianalumab (an anti-B cell-activating factor [BAFF]-receptor antibody) and iscalimab (an anti-CD40 antibody) are promising biologics for pSS, their efficacy still needs to be evaluated in larger clinical trials. For other biologics, clinical trials have found no differences versus placebo in the change from baseline in European League Against Rheumatism Sjögren's Syndrome Disease Activity Index (ESSDAI) score and fatigue score. Possible causes of the disappointing outcomes mainly include the inefficacy of those evaluated biologics in treating pSS, the high heterogeneous nature of pSS, irreversible exocrine glandular failure at advanced disease stages, inappropriate recruitment strategy in clinical trials, and outcome measures. Early diagnosis and glandular function-centered outcome measures may help to improve the current situation in the systemic therapy of pSS.
Collapse
|
16
|
Wolff ASB, Braun S, Husebye ES, Oftedal BE. B Cells and Autoantibodies in AIRE Deficiency. Biomedicines 2021; 9:1274. [PMID: 34572460 PMCID: PMC8466229 DOI: 10.3390/biomedicines9091274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/11/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022] Open
Abstract
Autoimmune polyendocrine syndrome type 1 (APS-1) is a rare but severe monogenetic autoimmune endocrine disease caused by failure of the Autoimmune Regulator (AIRE). AIRE regulates the negative selection of T cells in the thymus, and the main pathogenic mechanisms are believed to be T cell-mediated, but little is known about the role of B cells. Here, we give an overview of the role of B cells in thymic and peripheral tolerance in APS-1 patients and different AIRE-deficient mouse models. We also look closely into which autoantibodies have been described for this disorder, and their implications. Based on what is known about B cell therapy in other autoimmune disorders, we outline the potential of B cell therapies in APS-1 and highlight the unresolved research questions to be answered.
Collapse
Affiliation(s)
- Anette S. B. Wolff
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (A.S.B.W.); (S.B.); (E.S.H.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- KG Jebsen Center for Autoimmune Disorders, University of Bergen, 5021 Bergen, Norway
| | - Sarah Braun
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (A.S.B.W.); (S.B.); (E.S.H.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls University, 69120 Heidelberg, Germany
| | - Eystein S. Husebye
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (A.S.B.W.); (S.B.); (E.S.H.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- KG Jebsen Center for Autoimmune Disorders, University of Bergen, 5021 Bergen, Norway
| | - Bergithe E. Oftedal
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (A.S.B.W.); (S.B.); (E.S.H.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- KG Jebsen Center for Autoimmune Disorders, University of Bergen, 5021 Bergen, Norway
| |
Collapse
|
17
|
Parlak Ak T. Bronchus-Associated Lymphoid Tissue (BALT) Histology and Its Role in Various Pathologies. Vet Med Sci 2021. [DOI: 10.5772/intechopen.99366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The lower respiratory tract is in direct communication with the external environment for gas exchange to occur. Therefore, it is constantly exposed to allergens, antigens, bacteria, viruses, and a wide variety of airborne foreign bodies. Bronchus-associated lymphoid tissue (BALT), which develops in response to these exposures and is one of the most prominent representatives of mucosa-associated lymphoid tissue (MALT), is important for generating rapid and specific bronchopulmonary adaptive immune responses. Therefore, this chapter focuses on the lymphoid architecture of BALT, which was first discovered in the bronchial wall of rabbits, its inducible form called inducible BALT (iBALT), its immunological response mechanisms, and its roles in certain pathologies including infectious and autoimmune diseases as well as in allergic and malignant conditions. In conclusion, it is hypothesized that BALT plays an important role in maintaining health and in the development of lower respiratory tract diseases; thanks to the pulmonary immune system in which it functions as a functional lymphoid tissue.
Collapse
|
18
|
Singh A, Behl T, Sehgal A, Singh S, Sharma N, Naved T, Bhatia S, Al-Harrasi A, Chakrabarti P, Aleya L, Vargas-De-La-Cruz C, Bungau S. Mechanistic insights into the role of B cells in rheumatoid arthritis. Int Immunopharmacol 2021; 99:108078. [PMID: 34426116 DOI: 10.1016/j.intimp.2021.108078] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 11/25/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease epitomized by severe inflammation that induces tendon, cartilage, and bone damage over time. Although different types of cells undertake pathogenic functions in RA, the B cell's significant involvement has increasingly been known following the development of rheumatoid factor and it has been re-emphasized in recent years. Therefore, the rheumatoid factors and anti-cyclic citrullinated peptide antibodies are well-known indications of infection and clinical manifestations, and that they can precede the development of illness by several years. The emergence of rituximab a B cell reducing chimeric antidote in 1997 and 1998 transformed B-cell-targeted therapy for inflammatory disorder from a research hypothesis to a functional fact. Ever since, several autoantibody-related conditions were addressed, including the more intriguing indications of effectiveness seen in rheumatoid arthritis patients. Numerous types of B-cell-targeted compounds are currently being researched. From the beginning, one of the primary goals of B-cell therapy was to reinstate some kind of immune tolerance. While B cells have long been recognized as essential autoantibody producers, certain antibody-independent functions and usefulness as a key targeted therapy were not recognized until recently. The knowledge of B cells' diverse physical and pathogenic roles in autoimmune diseases is growing. As a result, the number of successful agents targeting the B cell complex is becoming more ubiquitous. Therefore, in this article, we explore fresh perspectives upon the roles of B cells in arthritis treatment, as well as new evidence regarding the effectiveness of B lymphocytes reduction and the therapeutic outcome of biological markers.
Collapse
Affiliation(s)
- Anuja Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tanveer Naved
- Amity Institute of Pharmacy, Amity University, Noida, India
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman; School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | | | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, France
| | - Celia Vargas-De-La-Cruz
- Faculty of Pharmacy and Biochemistry, Academic Department of Pharmacology, Bromatology and Toxicology, Centro Latinoamericano de Ensenanza e Investigacion en Bacteriologia Alimentaria, Universidad Nacinol Mayor de San Marcos, Lima, Peru; E-Health Research Center, Universidad de Ciencias y Humanidades, Lima, Peru
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
19
|
Retamozo S, Sisó-Almirall A, Flores-Chávez A, Ramos-Casals M, Brito-Zerón P. An update of targeted therapeutic options for primary Sjögren syndrome: current status and future development. Expert Opin Pharmacother 2021; 22:2359-2371. [PMID: 34323636 DOI: 10.1080/14656566.2021.1951224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: Primary Sjögren syndrome (pSS) is a systemic autoimmune disease that may affect 3 in 1,000 people within the general population. The therapeutic scenario is complex, and no therapy has proved to be able to modify the natural course of the disease, nor to prevent the most severe systemic complications.Areas covered: Recently, the EULAR 2020 Recommendations for pSS have underlined the low level of evidence supporting efficacious therapeutic approaches, lacking a definition of specific treatment targets and being far from the 'disease modification' concept that is frequently used in other diseases. Herein, the authors review the status of current targeted therapies and provide the reader with their expert opinion.Expert opinion: The progress in discovering novel treatments for pSS seem to be focused on searching new biological therapies as highly-selective drugs that can be effective without the adverse effects related to the wide, nonspecific immunosuppression induced by the drugs currently used. Most likely, the more disruptive therapeutic approach in pSS that could be seen in a few years is the use of combination strategies targeting different etiopathogenic pathways.
Collapse
Affiliation(s)
- Soledad Retamozo
- Sjogren Syndrome Research Group (AGAUR), Department of Autoinmune Diseases, ICMiD, Hospital Clinic, Barcelona, Spain.,Rheumatology Department, Hospital Universitari Parc Taulí, Sabadell, Barcelona, Spain
| | - Antoni Sisó-Almirall
- Primary Care Centre Les Corts, Consorci d'Atenció Primària De Salut Barcelona Esquerra (CAPSBE), Barcelona, Spain.,Primary Healthcare Transversal Research Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Manuel Ramos-Casals
- Sjogren Syndrome Research Group (AGAUR), Department of Autoinmune Diseases, ICMiD, Hospital Clinic, Barcelona, Spain.,Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Barcelona, Spain
| | - Pilar Brito-Zerón
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Barcelona, Spain.,Autoimmune Diseases Unit, Department of Medicine, Hospital CIMA-Sanitas, Barcelona, Spain
| |
Collapse
|
20
|
Rodriguez AB, Peske JD, Woods AN, Leick KM, Mauldin IS, Meneveau MO, Young SJ, Lindsay RS, Melssen MM, Cyranowski S, Parriott G, Conaway MR, Fu YX, Slingluff CL, Engelhard VH. Immune mechanisms orchestrate tertiary lymphoid structures in tumors via cancer-associated fibroblasts. Cell Rep 2021; 36:109422. [PMID: 34289373 PMCID: PMC8362934 DOI: 10.1016/j.celrep.2021.109422] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 02/26/2021] [Accepted: 06/28/2021] [Indexed: 12/21/2022] Open
Abstract
Tumor-associated tertiary lymphoid structures (TA-TLS) are associated with enhanced patient survival and responsiveness to cancer therapies, but the mechanisms underlying their development are unknown. We show here that TA-TLS development in murine melanoma is orchestrated by cancer-associated fibroblasts (CAF) with characteristics of lymphoid tissue organizer cells that are induced by tumor necrosis factor receptor signaling. CAF organization into reticular networks is mediated by CD8 T cells, while CAF accumulation and TA-TLS expansion depend on CXCL13-mediated recruitment of B cells expressing lymphotoxin-α1β2. Some of these elements are also overrepresented in human TA-TLS. Additionally, we demonstrate that immunotherapy induces more and larger TA-TLS that are more often organized with discrete T and B cell zones, and that TA-TLS presence, number, and size are correlated with reduced tumor size and overall response to checkpoint immunotherapy. This work provides a platform for manipulating TA-TLS development as a cancer immunotherapy strategy.
Collapse
Affiliation(s)
- Anthony B Rodriguez
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - J David Peske
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Amber N Woods
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Katie M Leick
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Ileana S Mauldin
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Max O Meneveau
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Samuel J Young
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Robin S Lindsay
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Marit M Melssen
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Salwador Cyranowski
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Geoffrey Parriott
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Mark R Conaway
- Division of Translational Research & Applied Statistics, Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Craig L Slingluff
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Victor H Engelhard
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| |
Collapse
|
21
|
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: 99] [Impact Index Per Article: 24.8] [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
|
22
|
Blanchard L, Girard JP. High endothelial venules (HEVs) in immunity, inflammation and cancer. Angiogenesis 2021; 24:719-753. [PMID: 33956259 PMCID: PMC8487881 DOI: 10.1007/s10456-021-09792-8] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022]
Abstract
High endothelial venules (HEVs) are specialized blood vessels mediating lymphocyte trafficking to lymph nodes (LNs) and other secondary lymphoid organs. By supporting high levels of lymphocyte extravasation from the blood, HEVs play an essential role in lymphocyte recirculation and immune surveillance for foreign invaders (bacterial and viral infections) and alterations in the body’s own cells (neoantigens in cancer). The HEV network expands during inflammation in immune-stimulated LNs and is profoundly remodeled in metastatic and tumor-draining LNs. HEV-like blood vessels expressing high levels of the HEV-specific sulfated MECA-79 antigens are induced in non-lymphoid tissues at sites of chronic inflammation in many human inflammatory and allergic diseases, including rheumatoid arthritis, Crohn’s disease, allergic rhinitis and asthma. Such vessels are believed to contribute to the amplification and maintenance of chronic inflammation. MECA-79+ tumor-associated HEVs (TA-HEVs) are frequently found in human tumors in CD3+ T cell-rich areas or CD20+ B-cell rich tertiary lymphoid structures (TLSs). TA-HEVs have been proposed to play important roles in lymphocyte entry into tumors, a process essential for successful antitumor immunity and lymphocyte-mediated cancer immunotherapy with immune checkpoint inhibitors, vaccines or adoptive T cell therapy. In this review, we highlight the phenotype and function of HEVs in homeostatic, inflamed and tumor-draining lymph nodes, and those of HEV-like blood vessels in chronic inflammatory diseases. Furthermore, we discuss the role and regulation of TA-HEVs in human cancer and mouse tumor models.
Collapse
Affiliation(s)
- Lucas Blanchard
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
23
|
Galletti JG, de Paiva CS. The ocular surface immune system through the eyes of aging. Ocul Surf 2021; 20:139-162. [PMID: 33621658 PMCID: PMC8113112 DOI: 10.1016/j.jtos.2021.02.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/04/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023]
Abstract
Since the last century, advances in healthcare, housing, and education have led to an increase in life expectancy. Longevity is accompanied by a higher prevalence of age-related diseases, such as cancer, autoimmunity, diabetes, and infection, and part of this increase in disease incidence relates to the significant changes that aging brings about in the immune system. The eye is not spared by aging either, presenting with age-related disorders of its own, and interestingly, many of these diseases have immune pathophysiology. Being delicate organs that must be exposed to the environment in order to capture light, the eyes are endowed with a mucosal environment that protects them, the so-called ocular surface. As in other mucosal sites, immune responses at the ocular surface need to be swift and potent to eliminate threats but are at the same time tightly controlled to prevent excessive inflammation and bystander damage. This review will detail how aging affects the mucosal immune response of the ocular surface as a whole and how this process relates to the higher incidence of ocular surface disease in the elderly.
Collapse
Affiliation(s)
- Jeremias G Galletti
- Innate Immunity Laboratory, Institute of Experimental Medicine (IMEX), CONICET-National Academy of Medicine, Buenos Aires, Argentina.
| | - Cintia S de Paiva
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
24
|
He SH, Xuan YY, Liang X, Guo KJ, Wang Y, Fu XN, Li TF. Investigative biological therapies for primary Sjögren's syndrome. Arch Med Sci 2020; 20:506-516. [PMID: 38757038 PMCID: PMC11094820 DOI: 10.5114/aoms.2020.97288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/19/2019] [Indexed: 05/18/2024] Open
Abstract
Primary Sjögren's syndrome (pSS) is a chronic, systemic autoimmune disease characterized by dryness of the eyes and mouth. The histological feature is mononuclear cell infiltration in exocrine glands, primarily salivary and lachrymal glands. As the disease progresses, some other tissues and organs may be involved and extraglandular manifestations ensue. The major current treatments are palliative and empirical, and in most cases the outcomes are not satisfactory. Emerging data indicate a critical role of lymphocytes in its development and progression. While pioneering work targeting B cells has demonstrated some encouraging results, more trials are warranted to validate the safety and efficacy. In addition, modulation of T cell function with abatacept ameliorates the severity of pSS. Furthermore, clinical trials to inhibit important cytokines involved in its formation have been carried out. In this article, we summarize and compare current biological therapies in order to find new and effective treatments for pSS.
Collapse
Affiliation(s)
- Shi-Hao He
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi-Yi Xuan
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xu Liang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kuang-Jin Guo
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-Ning Fu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tian-Fang Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
25
|
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
|
26
|
Tanaka S, Gauthier JM, Fuchs A, Li W, Tong AY, Harrison MS, Higashikubo R, Terada Y, Hachem RR, Ruiz-Perez D, Ritter JH, Cella M, Colonna M, Turnbull IR, Krupnick AS, Gelman AE, Kreisel D. IL-22 is required for the induction of bronchus-associated lymphoid tissue in tolerant lung allografts. Am J Transplant 2020; 20:1251-1261. [PMID: 31721409 PMCID: PMC7183893 DOI: 10.1111/ajt.15701] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 10/28/2019] [Accepted: 11/11/2019] [Indexed: 01/25/2023]
Abstract
Long-term survival after lung transplantation remains profoundly limited by graft rejection. Recent work has shown that bronchus-associated lymphoid tissue (BALT), characterized by the development of peripheral nodal addressin (PNAd)-expressing high endothelial venules and enriched in B and Foxp3+ T cells, is important for the maintenance of allograft tolerance. Mechanisms underlying BALT induction in tolerant pulmonary allografts, however, remain poorly understood. Here, we show that the development of PNAd-expressing high endothelial venules within intragraft lymphoid follicles and the recruitment of B cells, but not Foxp3+ cells depends on IL-22. We identify graft-infiltrating gamma-delta (γδ) T cells and Type 3 innate lymphoid cells (ILC3s) as important producers of IL-22. Reconstitution of IL-22 at late time points through retransplantation into wildtype hosts mediates B cell recruitment into lymphoid follicles within the allograft, resulting in a significant increase in their size, but does not induce PNAd expression. Our work has identified cellular and molecular requirements for the induction of BALT in pulmonary allografts during tolerance induction and may provide a platform for the development of new therapies for lung transplant patients.
Collapse
Affiliation(s)
- Satona Tanaka
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - Jason M. Gauthier
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - Anja Fuchs
- Section of Acute and Critical Care Surgery, Department of Surgery, Washington University, Saint Louis, Missouri
| | - Wenjun Li
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - Alice Y. Tong
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - M. Shea Harrison
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - Ryuji Higashikubo
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - Yuriko Terada
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
| | - Ramsey R. Hachem
- Department of Medicine, Washington University, Saint Louis, Missouri
| | - Daniel Ruiz-Perez
- Department of Pathology & Immunology, Washington University, Saint Louis, Missouri
| | - Jon H. Ritter
- Division of Experimental Surgery, La Paz University Hospital, Madrid, Spain
| | - Marina Cella
- Division of Experimental Surgery, La Paz University Hospital, Madrid, Spain
| | - Marco Colonna
- Division of Experimental Surgery, La Paz University Hospital, Madrid, Spain
| | - Isaiah R. Turnbull
- Section of Acute and Critical Care Surgery, Department of Surgery, Washington University, Saint Louis, Missouri
| | - Alexander S. Krupnick
- Division of Thoracic Surgery, Department of Surgery, University of Virginia, Charlottesville, Virginia
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia
| | - Andrew E. Gelman
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
- Division of Experimental Surgery, La Paz University Hospital, Madrid, Spain
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Washington University, Saint Louis, Missouri
- Division of Experimental Surgery, La Paz University Hospital, Madrid, Spain
| |
Collapse
|
27
|
Khalafalla MG, Woods LT, Jasmer KJ, Forti KM, Camden JM, Jensen JL, Limesand KH, Galtung HK, Weisman GA. P2 Receptors as Therapeutic Targets in the Salivary Gland: From Physiology to Dysfunction. Front Pharmacol 2020; 11:222. [PMID: 32231563 PMCID: PMC7082426 DOI: 10.3389/fphar.2020.00222] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
Although often overlooked in our daily lives, saliva performs a host of necessary physiological functions, including lubricating and protecting the oral cavity, facilitating taste sensation and digestion and maintaining tooth enamel. Therefore, salivary gland dysfunction and hyposalivation, often resulting from pathogenesis of the autoimmune disease Sjögren's syndrome or from radiotherapy of the head and neck region during cancer treatment, severely reduce the quality of life of afflicted patients and can lead to dental caries, periodontitis, digestive disorders, loss of taste and difficulty speaking. Since their initial discovery in the 1970s, P2 purinergic receptors for extracellular nucleotides, including ATP-gated ion channel P2X and G protein-coupled P2Y receptors, have been shown to mediate physiological processes in numerous tissues, including the salivary glands where P2 receptors represent a link between canonical and non-canonical saliva secretion. Additionally, extracellular nucleotides released during periods of cellular stress and inflammation act as a tissue alarmin to coordinate immunological and tissue repair responses through P2 receptor activation. Accordingly, P2 receptors have gained widespread clinical interest with agonists and antagonists either currently undergoing clinical trials or already approved for human use. Here, we review the contributions of P2 receptors to salivary gland function and describe their role in salivary gland dysfunction. We further consider their potential as therapeutic targets to promote physiological saliva flow, prevent salivary gland inflammation and enhance tissue regeneration.
Collapse
Affiliation(s)
- Mahmoud G. Khalafalla
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Lucas T. Woods
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - Kimberly J. Jasmer
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - Kevin Muñoz Forti
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - Jean M. Camden
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - Janicke L. Jensen
- Institute of Clinical Dentistry, Section of Oral Surgery and Oral Medicine, University of Oslo, Oslo, Norway
| | - Kirsten H. Limesand
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States
| | - Hilde K. Galtung
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Gary A. Weisman
- Department of Biochemistry, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
28
|
Banach-Orłowska M, Wyszyńska R, Pyrzyńska B, Maksymowicz M, Gołąb J, Miączyńska M. Cholesterol restricts lymphotoxin β receptor-triggered NF-κB signaling. Cell Commun Signal 2019; 17:171. [PMID: 31878945 PMCID: PMC6933913 DOI: 10.1186/s12964-019-0460-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/10/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Lymphotoxin β receptor (LTβR) plays important roles in the development of the immune system and immune response. At the cellular level, ligand-bound LTβR activates the pro-inflammatory NF-κB pathway but the detailed mechanisms regulating its signaling remain unknown. Understanding them is of high importance since LTβR and its ligands are promising therapeutic targets. Here, we studied the consequences of perturbed cellular cholesterol content on LTβR-induced NF-κB signaling. METHODS To modulate cholesterol availability and/or level in lung carcinoma A549 and H2228, and endothelial HUVEC cells different treatment regimens with filipin, methyl-β-cyclodextrin and simvastatin were applied. LTβR localization was studied by confocal microscopy. The activity of LTβR-induced NF-κB pathway was assessed by measuring the levels of NF-κB pathway inhibitor IκBα and phosphorylation of RelA transcription factor by Western blotting. The NF-κB transcriptional response, production of chemokines and adhesion molecules were examined by qRT-PCR, ELISA, and Western blotting, respectively. Adherence of different types of primary immune cells to epithelial A549 cells and endothelial HUVECs was measured fluorometrically. Interactions of LTβR with its protein partners were investigated by immunoprecipitation. RESULTS We showed that filipin-mediated sequestration of cholesterol or its depletion from the plasma membrane with methyl-β-cyclodextrin impaired LTβR internalization and potentiated LTβR-dependent activation of the canonical branch of the NF-κB pathway. The latter was manifested by enhanced degradation of IκBα inhibitor, elevated RelA phosphorylation, substantial increase in the expression of NF-κB target genes encoding, among others, cytokines and adhesion molecules known to play important roles in immune response. It was followed by robust secretion of CXCL8 and upregulation of ICAM1, that favored the adhesion of immune cells (NK and T cells, neutrophils) to A549 cells and HUVECs. Mechanistically, we showed that cholesterol depletion stabilized interactions of ligand-stimulated LTβR with modified forms of TRAF2 and NEMO proteins. CONCLUSIONS Our results showed that the reduction of the plasma membrane content of cholesterol or its sequestration strongly potentiated signaling outcome initiated by LTβR. Thus, drugs modulating cholesterol levels could potentially improve efficacy of LTβR-based therapies. Video abstract.
Collapse
Affiliation(s)
- Magdalena Banach-Orłowska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109, Warsaw, Poland.
| | - Renata Wyszyńska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109, Warsaw, Poland
| | - Beata Pyrzyńska
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Małgorzata Maksymowicz
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109, Warsaw, Poland
| | - Jakub Gołąb
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Marta Miączyńska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109, Warsaw, Poland
| |
Collapse
|
29
|
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
|
30
|
Skarstein K, Jensen JL, Galtung H, Jonsson R, Brokstad K, Aqrawi LA. Autoantigen-specific B cells and plasma cells are prominent in areas of fatty infiltration in salivary glands of patients with primary Sjögren’s syndrome. Autoimmunity 2019; 52:242-250. [DOI: 10.1080/08916934.2019.1684475] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Kathrine Skarstein
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | | | - Hilde Galtung
- Department of Oral Biology, University of Oslo, Oslo, Norway
| | - Roland Jonsson
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Rheumatology, Haukeland University Hospital, Bergen, Norway
| | - Karl Brokstad
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lara A. Aqrawi
- Department of Oral Surgery and Oral Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
31
|
Leverenz DL, St. Clair EW. Recent advances in the search for a targeted immunomodulatory therapy for primary Sjögren's syndrome. F1000Res 2019; 8:F1000 Faculty Rev-1532. [PMID: 31508200 PMCID: PMC6719673 DOI: 10.12688/f1000research.19842.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/22/2019] [Indexed: 12/20/2022] Open
Abstract
Primary Sjögren's syndrome is a chronic autoimmune disease characterized by salivary and lacrimal gland dysfunction, leading to substantial morbidity and reduced quality of life. Many patients with primary Sjögren's syndrome also have extraglandular systemic complications, some of which can be organ- or life-threatening. Over the last decade, numerous targeted immunomodulatory therapies for primary Sjögren's syndrome have failed to show a benefit in clinical trials, and as yet no disease-modifying therapy has been approved for this disease. Herein, we provide an updated review of the clinical trial landscape for primary Sjögren's syndrome and the numerous efforts to move the field forward, including the development of new classification criteria and outcome measures, the results of recent clinical trials in this field, the challenges faced in the search for effective therapies, and the expanding pipeline of novel therapies under development.
Collapse
Affiliation(s)
- David L. Leverenz
- Department of Medicine, Division of Rheumatology and Immunology, School of Medicine, Duke University, 40 Duke Medicine Circle, Durham, NC, 27110, USA
| | - E. William St. Clair
- Department of Medicine, Division of Rheumatology and Immunology, School of Medicine, Duke University, 40 Duke Medicine Circle, Durham, NC, 27110, USA
| |
Collapse
|
32
|
Mielle J, Tison A, Cornec D, Le Pottier L, Daien C, Pers JO. B cells in Sjögren's syndrome: from pathophysiology to therapeutic target. Rheumatology (Oxford) 2019; 60:2545-2560. [PMID: 30770916 DOI: 10.1093/rheumatology/key332] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/04/2018] [Indexed: 12/12/2022] Open
Abstract
Biological abnormalities associated with B lymphocytes are a hallmark of patients with primary Sjögren's syndrome. Those patients present abnormal distribution of B lymphocytes in peripheral blood and B cells in exocrine glands. B cells produce auto-antibodies, cytokines and present antigens but can also suppressive functions. In this review, we will summarize current knowledge on B cells in primary Sjögren's syndrome patients, demonstrate their critical role in the immunopathology of the disease and describe the past and current trials targeting B cells.
Collapse
Affiliation(s)
- Julie Mielle
- Departement of Rheumatology, UMR5535, Inflammation and Cancer, University of Montpellier and Teaching hospital of Montpellier, Montpellier, France
| | - Alice Tison
- UMR1227, Lymphocytes B et Autoimmunité, Université de Brest, Inserm, France.,Service de Rhumatologie, CHU de Brest, Brest, France
| | - Divi Cornec
- UMR1227, Lymphocytes B et Autoimmunité, Université de Brest, Inserm, France.,Service de Rhumatologie, CHU de Brest, Brest, France
| | | | - Claire Daien
- Departement of Rheumatology, UMR5535, Inflammation and Cancer, University of Montpellier and Teaching hospital of Montpellier, Montpellier, France
| | | |
Collapse
|
33
|
Affiliation(s)
- Toshio Odani
- Adeno-Associated Virus Biology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - John A. Chiorini
- Adeno-Associated Virus Biology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
34
|
MyD88 signaling causes autoimmune sialadenitis through formation of high endothelial venules and upregulation of LTβ receptor-mediated signaling. Sci Rep 2018; 8:14272. [PMID: 30250175 PMCID: PMC6155371 DOI: 10.1038/s41598-018-32690-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 09/13/2018] [Indexed: 01/17/2023] Open
Abstract
Autoimmune sialadenitis (AS), chronic inflammation of the salivary glands (SGs) with focal lymphocyte infiltration, appears in autoimmune diseases such as Sjӧgren’s syndrome. The pathological role of MyD88-dependent innate immune signaling in autoimmune diseases including AS has been studied using mouse models, such as NOD mice. Although AS development in NOD mice was reported to be suppressed by Myd88 deficiency, its specific role remains unclear. Here, we determined the potent suppressive effects of Myd88 deficiency on AS development in lupus-prone B6/lpr mice, which have lymphoproliferation abnormalities, and also in NOD mice, which have no lymphoproliferation abnormalities. This indicates that MyD88 signaling triggers AS through both lymphoproliferation-dependent and -independent mechanisms. To address the MyD88-dependent lymphoproliferation-independent AS manifestation, SGs from C57BL/6 mice were analyzed. Remarkable upregulation of Glycam1 and high endothelial venule (HEV)-associated changes were unexpectedly found in Myd88+/+ mice, compared with Myd88−/− mice. MyD88-dependent HEV-associated changes were also observed in NOD mice. Additionally, Lta, Ltb, and Ltbr in SGs of NOD mice were lowered by Myd88 deficiency. Interestingly, LTβR-induced HEV-associated gene expression in cultured cells was impaired by Myd88 deficiency. Our findings highlight novel roles for MyD88 in AS development, which imply the existence of MyD88-dependent HEV formation in ectopic lymphoid neogenesis.
Collapse
|
35
|
St Clair EW, Baer AN, Wei C, Noaiseh G, Parke A, Coca A, Utset TO, Genovese MC, Wallace DJ, McNamara J, Boyle K, Keyes-Elstein L, Browning JL, Franchimont N, Smith K, Guthridge JM, Sanz I, James JA. Clinical Efficacy and Safety of Baminercept, a Lymphotoxin β Receptor Fusion Protein, in Primary Sjögren's Syndrome: Results From a Phase II Randomized, Double-Blind, Placebo-Controlled Trial. Arthritis Rheumatol 2018; 70:1470-1480. [PMID: 29604186 PMCID: PMC6115299 DOI: 10.1002/art.40513] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 03/22/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To evaluate the clinical efficacy and safety of baminercept, a lymphotoxin β receptor IgG fusion protein (LTβR-Ig), for the treatment of primary Sjögren's syndrome (SS), and to explore the possible mechanisms of action of this treatment. METHODS In this multicenter trial, 52 patients with primary SS were randomized in a 2:1 ratio to receive subcutaneous injections of 100 mg of baminercept every week for 24 weeks or matching placebo. The primary end point was the change between screening and week 24 in the stimulated whole salivary flow (SWSF) rate. Secondary end points included the European League Against Rheumatism Sjögren's Syndrome Disease Activity Index (ESSDAI), as well as measurements of select chemokines and cytokines and enumeration of peripheral blood B and T cell subsets. RESULTS The change from baseline to week 24 in the SWSF rate was not significantly different between the baminercept and placebo treatment groups (baseline-adjusted mean change -0.01 versus 0.07 ml/minute; P = 0.332). The change in the ESSDAI during treatment was also not significantly different between the treatment groups (baseline-adjusted mean change -1.23 versus -0.15; P = 0.104). Although the incidence of adverse events was similar between the treatment groups, baminercept therapy was associated with a higher incidence of liver toxicity, including 2 serious adverse events. Baminercept also produced a significant decrease in plasma levels of CXCL13 and significant changes in the number of circulating B and T cells, consistent with its known inhibitory effects on LTβR signaling. CONCLUSION In this trial, treatment with baminercept failed to significantly improve glandular and extraglandular disease in patients with primary SS, despite evidence from mechanistic studies showing that it blocks LTβR signaling.
Collapse
Affiliation(s)
| | - Alan N Baer
- Johns Hopkins University, Baltimore, Maryland
| | - Chungwen Wei
- Ignacio Sanz, Emory University, Atlanta, Georgia
| | | | - Anne Parke
- Saint Francis Medical Group, Hartford, Connecticut
| | | | | | | | | | - James McNamara
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | | | | | | | | | | | - Ignacio Sanz
- Ignacio Sanz, Emory University, Atlanta, Georgia
| | | |
Collapse
|
36
|
Woods LT, Camden JM, Khalafalla MG, Petris MJ, Erb L, Ambrus JL, Weisman GA. P2Y 2 R deletion ameliorates sialadenitis in IL-14α-transgenic mice. Oral Dis 2018; 24:761-771. [PMID: 29297959 DOI: 10.1111/odi.12823] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 12/04/2017] [Accepted: 12/22/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Interleukin-14α-transgenic (IL-14αTG) mice develop an autoimmune exocrinopathy with characteristics similar to Sjögren's syndrome, including sialadenitis and hyposalivation. The P2Y2 receptor (P2Y2 R) for extracellular ATP and UTP is upregulated during salivary gland inflammation (i.e., sialadenitis) where it regulates numerous inflammatory responses. This study investigated the role of P2Y2 Rs in autoimmune sialadenitis in the IL-14αTG mouse model of Sjögren's syndrome. MATERIALS AND METHODS IL-14αTG mice were bred with P2Y2 R-/- mice to generate IL-14αTG × P2Y2 R-/- mice. P2Y2 R expression, lymphocytic focus scores, B- and T-cell accumulation, and lymphotoxin-α expression were evaluated in the submandibular glands (SMG) along with carbachol-stimulated saliva secretion in IL-14αTG, IL-14αTG × P2Y2 R-/- , and C57BL/6 control mice at 9 and 12 months of age. RESULTS Genetic ablation of P2Y2 Rs in IL-14αTG mice significantly reduced B and T lymphocyte infiltration of SMGs. However, reduced sialadenitis did not restore saliva secretion in IL-14αTG × P2Y2 R-/- mice. Decreased sialadenitis in IL-14αTG × P2Y2 R-/- mice correlated with decreased lymphotoxin-α levels, a critical proinflammatory cytokine associated with autoimmune pathology in IL-14αTG mice. CONCLUSIONS The results of this study suggest that P2Y2 Rs contribute to the development of salivary gland inflammation in IL-14αTG mice and may also contribute to autoimmune sialadenitis in humans.
Collapse
Affiliation(s)
- L T Woods
- Department of Biochemistry, University of Missouri, Columbia, MO, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - J M Camden
- Department of Biochemistry, University of Missouri, Columbia, MO, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - M G Khalafalla
- Department of Biochemistry, University of Missouri, Columbia, MO, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - M J Petris
- Department of Biochemistry, University of Missouri, Columbia, MO, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,Department of Nutritional Sciences and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - L Erb
- Department of Biochemistry, University of Missouri, Columbia, MO, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - J L Ambrus
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, SUNY at Buffalo School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - G A Weisman
- Department of Biochemistry, University of Missouri, Columbia, MO, USA.,Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
37
|
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
|
38
|
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
|
39
|
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
|
40
|
Retamozo S, Flores-Chavez A, Consuegra-Fernández M, Lozano F, Ramos-Casals M, Brito-Zerón P. Cytokines as therapeutic targets in primary Sjögren syndrome. Pharmacol Ther 2017; 184:81-97. [PMID: 29092775 DOI: 10.1016/j.pharmthera.2017.10.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Primary Sjögren syndrome (SjS) is a systemic autoimmune disease that may affect 1 in 1000 people (overwhelmingly women) and that can be a serious disease with excess mortality due to severe organ-specific involvements and the development of B cell lymphoma; systemic involvement clearly marks the disease prognosis, and strongly suggests the need for closer follow-up and more robust therapeutic management. Therapy is established according to the organ involved and severity. As a rule, the management of systemic SjS should be organ-specific, with glucocorticoids and immunosuppressive agents limited to potentially-severe involvements; unfortunately, the limited evidence available for these drugs, together with the potential development of serious adverse events, makes solid therapeutic recommendations difficult. The emergence of biological therapies has increased the therapeutic armamentarium available to treat primary SjS. Biologics currently used in SjS patients are used off-label and are overwhelmingly agents targeting B cells, but the most recent studies are moving on into the evaluation of targeting specific cytokines involved in the SjS pathogenesis. The most recent etiopathogenic advances in SjS are shedding some light in the search for new highly-selective biological therapies without the adverse effects of the standard drugs currently used (corticosteroids and immunosuppressant drugs). This review summarizes the potential pharmacotherapeutic options targeting the main cytokine families involved in the etiopathogenesis of primary SjS and analyzes potential insights for developing new therapies.
Collapse
Affiliation(s)
- Soledad Retamozo
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Spain; Hospital Privado Universitario de Córdoba, Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Córdoba, Argentina; Instituto de Investigaciones en Ciencias de la Salud, Universidad Nacional de Córdoba, Consejo Nacional de Investigaciones Científicas y Técnicas (INICSA-UNC-CONICET), Córdoba, Argentina; Department of Autoimmune Diseases, ICMiD, Hospital Clínic Barcelona, Spain
| | - Alejandra Flores-Chavez
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Spain; Biomedical Research Unit 02, Clinical Epidemiology Research Unit, UMAE, Specialties Hospital, Western Medical Center, Mexican Institute for Social Security (IMSS), Guadalajara, Mexico; Postgraduate Program of Medical Science, University Center for Biomedical Research (CUIB), University of Colima, Colima, Mexico; Department of Autoimmune Diseases, ICMiD, Hospital Clínic Barcelona, Spain
| | - Marta Consuegra-Fernández
- Immunoreceptors del Sistema Innat I Adaptatiu, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Francisco Lozano
- Immunoreceptors del Sistema Innat I Adaptatiu, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Servei d'Immunologia, Centre de Diagnòstic Biomèdic, Hospital Clínic de Barcelona, Barcelona, Spain; Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.
| | - Manuel Ramos-Casals
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain; Department of Autoimmune Diseases, ICMiD, Hospital Clínic Barcelona, Spain.
| | - Pilar Brito-Zerón
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, CELLEX-IDIBAPS, Spain; Autoimmune Diseases Unit, Department of Medicine, Hospital CIMA-Sanitas, Barcelona, Spain; Department of Autoimmune Diseases, ICMiD, Hospital Clínic Barcelona, Spain
| |
Collapse
|
41
|
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
|
42
|
Beyond TNF: TNF superfamily cytokines as targets for the treatment of rheumatic diseases. Nat Rev Rheumatol 2017; 13:217-233. [PMID: 28275260 DOI: 10.1038/nrrheum.2017.22] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
TNF blockers are highly efficacious at dampening inflammation and reducing symptoms in rheumatic diseases such as rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis, and also in nonrheumatic syndromes such as inflammatory bowel disease. As TNF belongs to a superfamily of 19 structurally related proteins that have both proinflammatory and anti-inflammatory activity, reagents that disrupt the interaction between proinflammatory TNF family cytokines and their receptors, or agonize the anti-inflammatory receptors, are being considered for the treatment of rheumatic diseases. Biologic agents that block B cell activating factor (BAFF) and receptor activator of nuclear factor-κB ligand (RANKL) have been approved for the treatment of systemic lupus erythematosus and osteoporosis, respectively. In this Review, we focus on additional members of the TNF superfamily that could be relevant for the pathogenesis of rheumatic disease, including those that can strongly promote activity of immune cells or increase activity of tissue cells, as well as those that promote death pathways and might limit inflammation. We examine preclinical mouse and human data linking these molecules to the control of damage in the joints, muscle, bone or other tissues, and discuss their potential as targets for future therapy of rheumatic diseases.
Collapse
|
43
|
Everett S, Vishwanath S, Cavero V, Shen L, Suresh L, Malyavantham K, Lincoff-Cohen N, Ambrus JL. Analysis of novel Sjogren's syndrome autoantibodies in patients with dry eyes. BMC Ophthalmol 2017; 17:20. [PMID: 28270126 PMCID: PMC5341407 DOI: 10.1186/s12886-017-0412-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 02/11/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dry eye is a common problem in Ophthalmology and may occur for many reasons including Sjogren's syndrome (SS). Recent studies have identified autoantibodies, anti-salivary gland protein 1 (SP1), anti-carbonic anhydrase 6 (CA6) and anti-parotid secretory protein (PSP), which occur early in the course of SS. The current studies were designed to evaluate how many patients with idiopathic dry eye and no evidence of systemic diseases from a dry eye practice have these autoantibodies. METHODS Patients from a dry eye clinic and normal controls were assessed by Schirmer's test for tear flow. Sera were assessed for autoantibodies using ELISA assays. Statistics was performed with Prism 7 software and student's unpaired t test. RESULTS In this study 60% of the dry eye patients expressed one of these autoantibodies. Only 30% expressed one of the autoantibodies associated with long-standing SS, which are included in the diagnostic criteria for SS, anti-Ro and anti-La. Patients with disease for less than 2 years and mild dry eyes did not express anti-Ro or anti-La, while 25% expressed anti-SP1. Similar observations, with smaller numbers, were made when patients had not only dry eye but also dry mouth. CONCLUSIONS Antibodies to SP1, CA6 and PSP occur in some patients with idiopathic dry eyes. Further studies will be needed to determine how many of these patients go on to develop systemic manifestations of SS. Testing for these autoantibodies may allow early recognition of patients with SS. This will lead to improved management of the patients and the development of new strategies to maintain normal lacrimal and salivary gland function in patients with SS.
Collapse
Affiliation(s)
- Sandra Everett
- Department of Ophthalmology, SUNY at Buffalo School of Medicine, Buffalo, NY, USA
| | - Sahana Vishwanath
- Department of Medicine, SUNY at Buffalo School of Medicine, Buffalo, NY, USA
| | - Vanessa Cavero
- Department of Medicine, SUNY at Buffalo School of Medicine, Buffalo, NY, USA
| | | | - Lakshmanan Suresh
- Dental School, SUNY at Buffalo School of Medicine, Buffalo, NY, USA.,Immco Diagnostics, Buffalo, NY, USA
| | | | - Norah Lincoff-Cohen
- Department of Ophthalmology, SUNY at Buffalo School of Medicine, Buffalo, NY, USA.,Department of Neurology, SUNY at Buffalo School of Medicine, Buffalo, NY, USA
| | - Julian L Ambrus
- Department of Medicine, SUNY at Buffalo School of Medicine, Buffalo, NY, USA. .,Division of Allergy, Immunology and Rheumatology, SUNY at Buffalo School of Medicine, Room C281, Buffalo General Hospital, 100 High Street, Buffalo, NY, 14203, USA.
| |
Collapse
|
44
|
Affiliation(s)
- Ana Raquel Rodrigues
- Department of Biochemistry, Faculty of Medicine, University of Porto, Porto, Portugal and
| | - Raquel Soares
- Department of Biochemistry, Faculty of Medicine, University of Porto, Porto, Portugal and
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
| |
Collapse
|
45
|
Bombardieri M, Lewis M, Pitzalis C. Ectopic lymphoid neogenesis in rheumatic autoimmune diseases. Nat Rev Rheumatol 2017; 13:141-154. [PMID: 28202919 DOI: 10.1038/nrrheum.2016.217] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ectopic lymphoid neogenesis often occurs in the target tissues of patients with chronic rheumatic autoimmune diseases such as rheumatoid arthritis, Sjögren syndrome and other connective tissue disorders, including systemic lupus erythematosus and myositis. However, the mechanisms of ectopic lymphoid-like structure (ELS) formation and function are not entirely understood. For example, it is unclear whether ELSs indicate distinct disease phenotypes or whether they are evolutionary manifestations of chronic inflammation. Also unclear is why ELSs form in some patients but not in others. Nonetheless, ELSs frequently display functional features of ectopic germinal centres and can actively contribute to the maintenance of autoimmunity through the production of disease-specific autoantibodies; furthermore, they seem to influence disease severity and response to both synthetic and biologic DMARDs. In this Review, we discuss current knowledge and gaps in understanding of ELS formation and function including their prevalence in the above rheumatic autoimmune diseases; the mechanisms underlying their formation, maintenance and function, including positive and negative regulatory pathways; their functional relevance in the perpetuation of autoimmunity; their relationship with disease phenotypes, clinical outcomes and response to treatment; and the potential for specific targeting of ELSs through novel therapeutic modalities.
Collapse
Affiliation(s)
- Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, John Vane Science Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Myles Lewis
- Centre for Experimental Medicine and Rheumatology, John Vane Science Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, John Vane Science Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
46
|
Innate lymphoid cells in autoimmunity: emerging regulators in rheumatic diseases. Nat Rev Rheumatol 2017; 13:164-173. [PMID: 28148916 DOI: 10.1038/nrrheum.2016.218] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Innate lymphoid cells (ILCs) are important in the regulation of barrier homeostasis. These cells do not express T cell receptors but share many functional similarities with T helper cells and cytotoxic CD8+ T lymphocytes. ILCs are divided into three groups, namely group 1 ILCs, group 2 ILCs and group 3 ILCs, based on the transcription factors they depend on for their development and function, and the cytokines they produce. Emerging data indicate that ILCs not only have protective functions but can also have detrimental effects when dysregulated, leading to chronic inflammation and autoimmune diseases, including asthma, inflammatory bowel disease, graft-versus-host disease, psoriasis, rheumatoid arthritis and atopic dermatitis. Elucidation of the cytokine pathways involved in various autoimmune diseases - and the identification of ILCs as potent producers of these cytokines - points towards a potential role for these cellular players in the pathophysiology of these diseases. In this Review we discuss the current knowledge of the role of ILCs in the pathogenesis of rheumatic and other autoimmune diseases.
Collapse
|
47
|
Haskett S, Ding J, Zhang W, Thai A, Cullen P, Xu S, Petersen B, Kuznetsov G, Jandreski L, Hamann S, Reynolds TL, Allaire N, Zheng TS, Mingueneau M. Identification of Novel CD4+ T Cell Subsets in the Target Tissue of Sjögren's Syndrome and Their Differential Regulation by the Lymphotoxin/LIGHT Signaling Axis. THE JOURNAL OF IMMUNOLOGY 2016; 197:3806-3819. [PMID: 27815440 DOI: 10.4049/jimmunol.1600407] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 09/11/2016] [Indexed: 12/14/2022]
Abstract
Despite being one of the most common rheumatologic diseases, there is still no disease-modifying drug for primary Sjögren's syndrome (pSS). Advancing our knowledge of the target tissue has been limited by the low dimensionality of histology techniques and the small size of human salivary gland biopsies. In this study, we took advantage of a molecularly validated mouse model of pSS to characterize tissue-infiltrating CD4+ T cells and their regulation by the lymphotoxin/LIGHT signaling axis. Novel cell subsets were identified by combining highly dimensional flow and mass cytometry with transcriptomic analyses. Pharmacologic modulation of the LTβR signaling pathway was achieved by treating mice with LTβR-Ig, a therapeutic intervention currently being tested in pSS patients (Baminercept trial NCT01552681). Using these approaches, we identified two novel CD4+ T cell subsets characterized by high levels of PD1: Prdm1+ effector regulatory T cells expressing immunoregulatory factors, such as Il10, Areg, Fgl2, and Itgb8, and Il21+ effector conventional T cells expressing a pathogenic transcriptional signature. Mirroring these observations in mice, large numbers of CD4+PD1+ T cells were detected in salivary glands from Sjögren's patients but not in normal salivary glands or kidney biopsies from lupus nephritis patients. Unexpectedly, LTβR-Ig selectively halted the recruitment of PD1- naive, but not PD1+, effector T cells to the target tissue, leaving the cells with pathogenic potential unaffected. Altogether, this study revealed new cellular players in pSS pathogenesis, their transcriptional signatures, and differential dependency on the lymphotoxin/LIGHT signaling axis that help to interpret the negative results of the Baminercept trial and will guide future therapeutic interventions.
Collapse
Affiliation(s)
| | - Jian Ding
- Immunology Research, Biogen, Cambridge, MA 02142
| | - Wei Zhang
- Immunology Research, Biogen, Cambridge, MA 02142
| | - Alice Thai
- Immunology Research, Biogen, Cambridge, MA 02142
| | | | - Shanqin Xu
- Immunology Research, Biogen, Cambridge, MA 02142
| | | | | | | | | | | | - Norm Allaire
- Immunology Research, Biogen, Cambridge, MA 02142
| | | | | |
Collapse
|
48
|
Bolstad AI, Skarstein K. Epidemiology of Sjögren's Syndrome-from an Oral Perspective. CURRENT ORAL HEALTH REPORTS 2016; 3:328-336. [PMID: 27891302 PMCID: PMC5104792 DOI: 10.1007/s40496-016-0112-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Oral symptoms are among the most distressing manifestations for patients with Sjögren's syndrome (SS). The feeling of dry mouth is unpleasant, and hyposalivation may contribute to difficulty in speaking, chewing and swallowing and reduced quality of life. Reduced salivary flow increases the risk for dental caries and problems with prosthetic replacement. It seems that SS is not as frequently occurring as previously anticipated. Population-based prevalence studies on primary SS in Europe, conducted on large background populations and in accordance with the AECG criteria, reported of a prevalence of 1-9 cases per 10,000 people. This gives a combined prevalence of nearly 39/100,000 (~0.04 %). The cause of Sjögren's syndrome is even now not fully understood, and the treatment of oral symptoms is still mostly palliative. Hopefully, useful information will appear from the new methods that are now available for genome wide association studies, epigenetics, DNA methylation studies, and proteomics. Similarly, this is anticipated for the immunological side of the story. The interferon signature, the interferon γ/interferon α mRNA ratio, and CXCL13 are among the proposed biomarkers of active disease. In this review, we provide an update on oral aspects of Sjögren's syndrome with emphasis on the latest publications on these topics.
Collapse
Affiliation(s)
- Anne Isine Bolstad
- Department of Clinical Dentistry, Faculty of Medicine and Dentistry, University of Bergen, Årstadveien 19, N-5009 Bergen, Norway
| | - Kathrine Skarstein
- Gade Laboratory for Pathology, Department of Clinical Medicine, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, N-5021 Bergen, Norway
| |
Collapse
|
49
|
McNamee EN, Rivera-Nieves J. Ectopic Tertiary Lymphoid Tissue in Inflammatory Bowel Disease: Protective or Provocateur? Front Immunol 2016; 7:308. [PMID: 27579025 PMCID: PMC4985530 DOI: 10.3389/fimmu.2016.00308] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/29/2016] [Indexed: 12/15/2022] Open
Abstract
Organized lymphoid tissues like the thymus first appeared in jawed vertebrates around 500 million years ago and have evolved to equip the host with a network of specialized sites, strategically located to orchestrate strict immune-surveillance and efficient immune responses autonomously. The gut-associated lymphoid tissues maintain a mostly tolerant environment to dampen our responses to daily dietary and microbial products in the intestine. However, when this homeostasis is perturbed by chronic inflammation, the intestine is able to develop florid organized tertiary lymphoid tissues (TLT), which heralds the onset of regional immune dysregulation. While TLT are a pathologic hallmark of Crohn's disease (CD), their role in the overall process remains largely enigmatic. A critical question remains; are intestinal TLT generated by the immune infiltrated intestine to modulate immune responses and rebuild tolerance to the microbiota or are they playing a more sinister role by generating dysregulated responses that perpetuate disease? Herein, we discuss the main theories of intestinal TLT neogenesis and focus on the most recent findings that open new perspectives to their role in inflammatory bowel disease.
Collapse
Affiliation(s)
- Eóin N McNamee
- Mucosal Inflammation Program, Department of Anesthesiology, School of Medicine, University of Colorado - Anschutz Medical Campus , Aurora, CO , USA
| | - Jesús Rivera-Nieves
- Division of Gastroenterology, Inflammatory Bowel Disease Center, San Diego VAMC, University of California San Diego , La Jolla, CA , USA
| |
Collapse
|
50
|
Ding J, Zhang W, Haskett S, Pellerin A, Xu S, Petersen B, Jandreski L, Hamann S, Reynolds TL, Zheng TS, Mingueneau M. BAFF overexpression increases lymphocytic infiltration in Sjögren's target tissue, but only inefficiently promotes ectopic B-cell differentiation. Clin Immunol 2016; 169:69-79. [PMID: 27352977 DOI: 10.1016/j.clim.2016.06.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/19/2016] [Accepted: 06/23/2016] [Indexed: 12/28/2022]
Abstract
B-cell activating factor (BAFF) levels are increased in rheumatoid arthritis, lupus and primary Sjögren's syndrome (pSS). However, BAFF contribution to pathogenesis is not completely understood. In pSS, immune infiltration of the salivary and lacrimal glands leads to xerostomia and xerophtalmia. Glandular B cell hyperactivation, differentiation into germinal center (GC)-like structures and plasma cell accumulation are histopathological hallmarks that were attributed to increased BAFF. Here, we experimentally tested this hypothesis by overexpressing BAFF in a mouse model of pSS. BAFF overexpression enhanced lymphocytic infiltration and MHCII expression on B cells. Increased BAFF also induced B cell differentiation into GC B cells within the autoimmune target tissue. However, even in these conditions, GC B cells only accounted for <1% of glandular B cells, demonstrating that BAFF is not efficiently promoting ectopic GC formation in pSS and warranting further investigation of therapeutics targeting both BAFF and the related TNF-family member APRIL.
Collapse
Affiliation(s)
- Jian Ding
- Immunology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Wei Zhang
- Immunology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Scott Haskett
- Immunology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Alex Pellerin
- Immunology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Shanqin Xu
- Immunology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Britta Petersen
- Immunology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Luke Jandreski
- Immunology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Stefan Hamann
- Immunology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Taylor L Reynolds
- Immunology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Timothy S Zheng
- Immunology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States
| | - Michael Mingueneau
- Immunology Research, Biogen, 115 Broadway, Cambridge, MA 02142, United States.
| |
Collapse
|