1
|
Sun G, Liu Y. Tertiary lymphoid structures in ovarian cancer. Front Immunol 2024; 15:1465516. [PMID: 39569184 PMCID: PMC11576424 DOI: 10.3389/fimmu.2024.1465516] [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: 07/16/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024] Open
Abstract
Ovarian cancer (OC) is a significant cause of cancer-related mortality in women worldwide. Despite advances in treatment modalities, including surgery and chemotherapy, the overall prognosis for OC patients remains poor, particularly for patients with advanced or recurrent disease. Immunotherapy, particularly immune checkpoint blockade (ICB), has revolutionized cancer treatment in various malignancies but has shown limited efficacy in treating OC, which is primarily attributed to the immunologically. Tertiary lymphoid structures (TLSs), which are ectopic aggregates of immune cells, have emerged as potential mediators of antitumor immunity. This review explores the composition, formation, and induction of tumor associated TLS (TA-TLS) in OC, along with their role and therapeutic implications in disease development and treatment. By elucidating the roles TA-TLSs and their cellular compositions played in OC microenvironment, novel therapeutic targets may be identified to overcome immune suppression and enhance immunotherapy efficacy in ovarian cancer.
Collapse
Affiliation(s)
- Guojuan Sun
- The Ward Section of Home Overseas Doctors, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Liu
- Department of Gynaecology and Obstetrics, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
2
|
Zhang T, Liu W, Yang YG. B cell development and antibody responses in human immune system mice: current status and future perspective. SCIENCE CHINA. LIFE SCIENCES 2024; 67:645-652. [PMID: 38270770 DOI: 10.1007/s11427-023-2462-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/28/2023] [Indexed: 01/26/2024]
Abstract
Humanized immune system (HIS) mice have been developed and used as a small surrogate model to study human immune function under normal or disease conditions. Although variations are found between models, most HIS mice show robust human T cell responses. However, there has been unsuccessful in constructing HIS mice that produce high-affinity human antibodies, primarily due to defects in terminal B cell differentiation, antibody affinity maturation, and development of primary follicles and germinal centers. In this review, we elaborate on the current knowledge about and previous attempts to improve human B cell development in HIS mice, and propose a potential strategy for constructing HIS mice with improved humoral immunity by transplantation of human follicular dendritic cells (FDCs) to facilitate the development of secondary follicles.
Collapse
Affiliation(s)
- Tao Zhang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, 130061, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, 130061, China
| | - Wentao Liu
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, 130061, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, 130061, China.
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, 130061, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, 130061, China.
- International Center of Future Science, Jilin University, Changchun, 130061, China.
| |
Collapse
|
3
|
Wang X, Li X, Zhao J, Li Y, Shin SR, Ligresti G, Ng AHM, Bromberg JS, Church G, Lemos DR, Abdi R. Rapid Generation of hPSC-Derived High Endothelial Venule Organoids with In Vivo Ectopic Lymphoid Tissue Capabilities. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308760. [PMID: 38306610 PMCID: PMC11009051 DOI: 10.1002/adma.202308760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/24/2024] [Indexed: 02/04/2024]
Abstract
Bioengineering strategies for the fabrication of implantable lymphoid structures mimicking lymph nodes (LNs) and tertiary lymphoid structures (TLS) could amplify the adaptive immune response for therapeutic applications such as cancer immunotherapy. No method to date has resulted in the consistent formation of high endothelial venules (HEVs), which is the specialized vasculature responsible for naïve T cell recruitment and education in both LNs and TLS. Here orthogonal induced differentiation of human pluripotent stem cells carrying a regulatable ETV2 allele is used to rapidly and efficiently induce endothelial differentiation. Assembly of embryoid bodies combining primitive inducible endothelial cells and primary human LN fibroblastic reticular cells results in the formation of HEV-like structures that can aggregate into 3D organoids (HEVOs). Upon transplantation into immunodeficient mice, HEVOs successfully engraft and form lymphatic structures that recruit both antigen-presenting cells and adoptively-transferred lymphocytes, therefore displaying basic TLS capabilities. The results further show that functionally, HEVOs can organize an immune response and promote anti-tumor activity by adoptively-transferred T lymphocytes. Collectively, the experimental approaches represent an innovative and scalable proof-of-concept strategy for the fabrication of bioengineered TLS that can be deployed in vivo to enhance adaptive immune responses.
Collapse
Affiliation(s)
- Xichi Wang
- Renal Division, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Xiaofei Li
- Renal Division, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Jing Zhao
- Renal Division, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Yi Li
- Renal Division, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Su Ryon Shin
- Renal Division, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Giovanni Ligresti
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Alex H M Ng
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, 02138, USA
| | - Jonathan S Bromberg
- Department of Surgery and Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - George Church
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, 02138, USA
| | - Dario R Lemos
- Renal Division, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Reza Abdi
- Renal Division, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| |
Collapse
|
4
|
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
|
5
|
Sylvestre M, Barbier N, Sibut V, Nayar S, Monvoisin C, Leonard S, Saint-Vanne J, Martin A, Guirriec M, Latour M, Jouan F, Baulande S, Bohec M, Verdière L, Mechta-Grigoriou F, Mourcin F, Bertheuil N, Barone F, Tarte K, Roulois D. KDM6B drives epigenetic reprogramming associated with lymphoid stromal cell early commitment and immune properties. SCIENCE ADVANCES 2023; 9:eadh2708. [PMID: 38019914 PMCID: PMC10686565 DOI: 10.1126/sciadv.adh2708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
Mature lymphoid stromal cells (LSCs) are key organizers of immune responses within secondary lymphoid organs. Similarly, inflammation-driven tertiary lymphoid structures depend on immunofibroblasts producing lymphoid cytokines and chemokines. Recent studies have explored the origin and heterogeneity of LSC/immunofibroblasts, yet the molecular and epigenetic mechanisms involved in their commitment are still unknown. This study explored the transcriptomic and epigenetic reprogramming underlying LSC/immunofibroblast commitment. We identified the induction of lysine demethylase 6B (KDM6B) as the primary epigenetic driver of early immunofibroblast differentiation. In addition, we observed an enrichment for KDM6B gene signature in murine inflammatory fibroblasts and pathogenic stroma of patients with autoimmune diseases. Last, KDM6B was required for the acquisition of LSC/immunofibroblast functional properties, including the up-regulation of CCL2 and the resulting recruitment of monocytes. Overall, our results reveal epigenetic mechanisms that participate in the early commitment and immune properties of immunofibroblasts and support the use of epigenetic modifiers as fibroblast-targeting strategies in chronic inflammation.
Collapse
Affiliation(s)
- Marvin Sylvestre
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Nicolas Barbier
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Vonick Sibut
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Saba Nayar
- Centre for Translational inflammation Research, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, UK
| | - Céline Monvoisin
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Simon Leonard
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, F-35043 Nantes, France
| | - Julien Saint-Vanne
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
- SITI, Pôle Biologie, CHU Rennes, F-35033 Rennes, France
| | - Ansie Martin
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Marion Guirriec
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Maëlle Latour
- SITI, Pôle Biologie, CHU Rennes, F-35033 Rennes, France
| | - Florence Jouan
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Sylvain Baulande
- Institut Curie Genomics of Excellence (ICGex) Platform, Institut Curie Research Center, PSL Research University, F-75005 Paris, France
| | - Mylène Bohec
- Institut Curie Genomics of Excellence (ICGex) Platform, Institut Curie Research Center, PSL Research University, F-75005 Paris, France
| | - Léa Verdière
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Fatima Mechta-Grigoriou
- Stress and Cancer Laboratory, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, INSERM, U830, PSL Research University, 26, rue d’Ulm, F-75005 Paris, France
| | - Frédéric Mourcin
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Nicolas Bertheuil
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
- Department of Plastic Surgery, CHU Rennes, F-35033 Rennes, France
| | | | - Karin Tarte
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
- SITI, Pôle Biologie, CHU Rennes, F-35033 Rennes, France
| | - David Roulois
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| |
Collapse
|
6
|
Lancaster JN. Aging of lymphoid stromal architecture impacts immune responses. Semin Immunol 2023; 70:101817. [PMID: 37572552 PMCID: PMC10929705 DOI: 10.1016/j.smim.2023.101817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
The secondary lymphoid organs (SLOs) undergo structural changes with age, which correlates with diminishing immune responses against infectious disease. A growing body of research suggests that the aged tissue microenvironment can contribute to decreased immune function, independent of intrinsic changes to hematopoietic cells with age. Stromal cells impart structural integrity, facilitate fluid transport, and provide chemokine and cytokine signals that are essential for immune homeostasis. Mechanisms that drive SLO development have been described, but their roles in SLO maintenance with advanced age are unknown. Disorganization of the fibroblasts of the T cell and B cell zones may reduce the maintenance of naïve lymphocytes and delay immune activation. Reduced lymphatic transport efficiency with age can also delay the onset of the adaptive immune response. This review focuses on recent studies that describe age-associated changes to the stroma of the lymph nodes and spleen. We also review recent investigations into stromal cell biology, which include high-dimensional analysis of the stromal cell transcriptome and viscoelastic testing of lymph node mechanical properties, as they constitute an important framework for understanding aging of the lymphoid tissues.
Collapse
Affiliation(s)
- Jessica N Lancaster
- Department of Immunology, Mayo Clinic, 13400 E. Shea Blvd., Scottsdale, AZ, USA; Department of Cancer Biology, Mayo Clinic, 13400 E. Shea Blvd., Scottsdale, AZ, USA.
| |
Collapse
|
7
|
Vallecillo-García P, Orgeur M, Comai G, Poehle-Kronawitter S, Fischer C, Gloger M, Dumas CE, Giesecke-Thiel C, Sauer S, Tajbakhsh S, Höpken UE, Stricker S. A local subset of mesenchymal cells expressing the transcription factor Osr1 orchestrates lymph node initiation. Immunity 2023; 56:1204-1219.e8. [PMID: 37160119 DOI: 10.1016/j.immuni.2023.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 12/05/2022] [Accepted: 04/13/2023] [Indexed: 05/11/2023]
Abstract
During development, lymph node (LN) initiation is coordinated by lymphoid tissue organizer (LTo) cells that attract lymphoid tissue inducer (LTi) cells at strategic positions within the embryo. The identity and function of LTo cells during the initial attraction of LTi cells remain poorly understood. Using lineage tracing, we demonstrated that a subset of Osr1-expressing cells was mesenchymal LTo progenitors. By investigating the heterogeneity of Osr1+ cells, we uncovered distinct mesenchymal LTo signatures at diverse anatomical locations, identifying a common progenitor of mesenchymal LTos and LN-associated adipose tissue. Osr1 was essential for LN initiation, driving the commitment of mesenchymal LTo cells independent of neural retinoic acid, and for LN-associated lymphatic vasculature assembly. The combined action of chemokines CXCL13 and CCL21 was required for LN initiation. Our results redefine the role and identity of mesenchymal organizer cells and unify current views by proposing a model of cooperative cell function in LN initiation.
Collapse
Affiliation(s)
| | - Mickael Orgeur
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, 75015 Paris, France
| | - Glenda Comai
- Institut Pasteur, Stem Cells & Development Unit, CNRS UMR 3738, Paris, France
| | | | - Cornelius Fischer
- Core Facility Genomics, Berlin Institute of Health at Charité, 10178 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115, Berlin, Germany
| | - Marleen Gloger
- Max Delbrück Center for Molecular Medicine, Department of Translational Tumor Immunology, 13125 Berlin, Germany; Uppsala University, Immunology Genetics and Pathology, 75237 Uppsala, Sweden
| | - Camille E Dumas
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | | | - Sascha Sauer
- Core Facility Genomics, Berlin Institute of Health at Charité, 10178 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115, Berlin, Germany
| | - Shahragim Tajbakhsh
- Institut Pasteur, Stem Cells & Development Unit, CNRS UMR 3738, Paris, France
| | - Uta E Höpken
- Max Delbrück Center for Molecular Medicine, Department of Microenvironmental Regulation in Autoimmunity and Cancer, 13125 Berlin, Germany
| | - Sigmar Stricker
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany.
| |
Collapse
|
8
|
Howley K, Berthelette A, Ceglia S, Kang J, Reboldi A. Embryonic type 3 innate lymphoid cells sense maternal dietary cholesterol to control local Peyer's patch development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.19.533339. [PMID: 36993524 PMCID: PMC10055282 DOI: 10.1101/2023.03.19.533339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Lymphoid tissue inducer (LTi) cells develop during intrauterine life and rely on developmental programs to initiate the organogenesis of secondary lymphoid organs (SLOs). This evolutionary conserved process endows the fetus with the ability to orchestrate the immune response after birth and to react to the triggers present in the environment. While it is established that LTi function can be shaped by maternal-derived cues and is critical to prepare the neonate with a functional scaffold to mount immune response, the cellular mechanisms that control anatomically distinct SLO organogenesis remain unclear. We discovered that LTi cells forming Peyer's patches, gut-specific SLOs, require the coordinated action of two migratory G protein coupled receptors (GPCR) GPR183 and CCR6. These two GPCRs are uniformly expressed on LTi cells across SLOs, but their deficiency specifically impacts Peyer's patch formation, even when restricted to fetal window. The unique CCR6 ligand is CCL20, while the ligand for GPR183 is the cholesterol metabolite 7α,25-Dihydroxycholesterol (7α,25-HC), whose production is controlled by the enzyme cholesterol 25-hydroxylase (CH25H). We identified a fetal stromal cell subset that expresses CH25H and attracts LTi cells in the nascent Peyer's patch anlagen. GPR183 ligand concentration can be modulated by the cholesterol content in the maternal diet and impacts LTi cell maturation in vitro and in vivo, highlighting a link between maternal nutrients and intestinal SLO organogenesis. Our findings revealed that in the fetal intestine, cholesterol metabolite sensing by GPR183 in LTi cells for Peyer's patch formation is dominant in the duodenum, the site of cholesterol absorption in the adult. This anatomic requirement suggests that embryonic, long-lived non-hematopoietic cells might exploit adult metabolic functions to ensure highly specialized SLO development in utero.
Collapse
Affiliation(s)
- Kelsey Howley
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Alyssa Berthelette
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Simona Ceglia
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Joonsoo Kang
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA
| |
Collapse
|
9
|
CHEN J, CHEN J, WANG L. Tertiary lymphoid structures as unique constructions associated with the organization, education, and function of tumor-infiltrating immunocytes. J Zhejiang Univ Sci B 2022; 23:812-822. [PMID: 36226536 PMCID: PMC9561406 DOI: 10.1631/jzus.b2200174] [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: 11/05/2022]
Abstract
Tertiary lymphoid structures (TLSs) are formations at sites with persistent inflammatory stimulation, including tumors. These ectopic lymphoid organs mainly consist of chemo-attracting B cells, T cells, and supporting dendritic cells (DCs). Mature TLSs exhibit functional organization for the optimal development and collaboration of adaptive immune response, delivering an augmented effect on the tumor microenvironment (TME). The description of the positive correlation between TLSs and tumor prognosis is reliable only under a certain condition involving the localization and maturation of TLSs. Emerging evidence suggests that underlying mechanisms of the anti-tumor effect of TLSs pave the way for novel immunotherapies. Several approaches have been developed to take advantage of intratumoral TLSs, either by combining it with therapeutic agents or by inducing the neogenesis of TLSs.
Collapse
Affiliation(s)
- Jing CHEN
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310009, China,Institute of Immunology and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310003, China
| | - Jian CHEN
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310009, China,Jian CHEN,
| | - Lie WANG
- Institute of Immunology and Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310003, China,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou311121, China,Cancer Center, Zhejiang University, Hangzhou310058, China,Lie WANG,
| |
Collapse
|
10
|
Kwok T, Medovich SC, Silva-Junior IA, Brown EM, Haug JC, Barrios MR, Morris KA, Lancaster JN. Age-Associated Changes to Lymph Node Fibroblastic Reticular Cells. FRONTIERS IN AGING 2022; 3:838943. [PMID: 35821826 PMCID: PMC9261404 DOI: 10.3389/fragi.2022.838943] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/07/2022] [Indexed: 12/26/2022]
Abstract
The decreased proportion of antigen-inexperienced, naïve T cells is a hallmark of aging in both humans and mice, and contributes to reduced immune responses, particularly against novel and re-emerging pathogens. Naïve T cells depend on survival signals received during their circulation among the lymph nodes by direct contacts with stroma, in particular fibroblastic reticular cells. Macroscopic changes to the architecture of the lymph nodes have been described, but it is unclear how lymph node stroma are altered with age, and whether these changes contribute to reduced naïve T cell maintenance. Here, using 2-photon microscopy, we determined that the aged lymph node displayed increased fibrosis and correspondingly, that naïve T-cell motility was impaired in the aged lymph node, especially in proximity to fibrotic deposition. Functionally, adoptively transferred young naïve T-cells exhibited reduced homeostatic turnover in aged hosts, supporting the role of T cell-extrinsic mechanisms that regulate their survival. Further, we determined that early development of resident fibroblastic reticular cells was impaired, which may correlate to the declining levels of naïve T-cell homeostatic factors observed in aged lymph nodes. Thus, our study addresses the controversy as to whether aging impacts the composition lymph node stroma and supports a model in which impaired differentiation of lymph node fibroblasts and increased fibrosis inhibits the interactions necessary for naïve T cell homeostasis.
Collapse
Affiliation(s)
- Tina Kwok
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | | | | | - Elise M Brown
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | - Joel C Haug
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | | | - Karina A Morris
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | | |
Collapse
|
11
|
James Bates RE, Browne E, Schalks R, Jacobs H, Tan L, Parekh P, Magliozzi R, Calabrese M, Mazarakis ND, Reynolds R. Lymphotoxin-alpha expression in the meninges causes lymphoid tissue formation and neurodegeneration. Brain 2022; 145:4287-4307. [PMID: 35776111 DOI: 10.1093/brain/awac232] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 05/24/2022] [Accepted: 06/17/2022] [Indexed: 11/14/2022] Open
Abstract
Organised meningeal immune cell infiltrates are suggested to play an important role in cortical grey matter pathology in the multiple sclerosis brain, but the mechanisms involved are as yet unresolved. Lymphotoxin-alpha plays a key role in lymphoid organ development and cellular cytotoxicity in the immune system and its expression is increased in the cerebrospinal fluid of naïve and progressive multiple sclerosis patients and post-mortem meningeal tissue. Here we show that persistently increased levels of lymphotoxin alpha in the cerebral meninges can give rise to lymphoid-like structures and underlying multiple sclerosis-like cortical pathology. Stereotaxic injections of recombinant lymphotoxin-alpha into the rat meninges led to acute meningeal inflammation and subpial demyelination that resolved after 28 days, with demyelination being dependent on prior sub-clinical immunisation with myelin oligodendrocyte glycoprotein. Injection of a lymphotoxin-alpha lentiviral vector into the cortical meningeal space, to produce chronic localised over-expression of the cytokine, induced extensive lymphoid-like immune cell aggregates, maintained over 3 months, including T-cell rich zones containing podoplanin+ fibroblastic reticular stromal cells and B-cell rich zones with a network of follicular dendritic cells, together with expression of lymphoid chemokines and their receptors. Extensive microglial and astroglial activation, subpial demyelination and marked neuronal loss occurred in the underlying cortical parenchyma. Whereas subpial demyelination was partially dependent on prior myelin oligodendrocyte glycoprotein immunisation, the neuronal loss was present irrespective of immunisation. Conditioned medium from LTα treated microglia was able to induce a reactive phenotype in astrocytes. Our results show that chronic lymphotoxin-alpha overexpression alone is sufficient to induce formation of meningeal lymphoid-like structures and subsequent neurodegeneration, similar to that seen in the progressive multiple sclerosis brain.
Collapse
Affiliation(s)
- Rachel E James Bates
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Eleanor Browne
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Renee Schalks
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Heather Jacobs
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Li Tan
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Puja Parekh
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Roberta Magliozzi
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK.,Neurology Section, Department of Neurological and Movement Sciences, University of Verona, Verona 37134, Italy
| | - Massimiliano Calabrese
- Neurology Section, Department of Neurological and Movement Sciences, University of Verona, Verona 37134, Italy
| | - Nicholas D Mazarakis
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK
| | - Richard Reynolds
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, Hammersmith, Hospital Campus, UK.,Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
12
|
Jacob JM, Di Carlo SE, Stzepourginski I, Lepelletier A, Ndiaye PD, Varet H, Legendre R, Kornobis E, Benabid A, Nigro G, Peduto L. PDGFRα-induced stromal maturation is required to restrain postnatal intestinal epithelial stemness and promote defense mechanisms. Cell Stem Cell 2022; 29:856-868.e5. [PMID: 35523143 DOI: 10.1016/j.stem.2022.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 01/18/2022] [Accepted: 04/07/2022] [Indexed: 11/03/2022]
Abstract
After birth, the intestine undergoes major changes to shift from an immature proliferative state to a functional intestinal barrier. By combining inducible lineage tracing and transcriptomics in mouse models, we identify a prodifferentiation PDGFRαHigh intestinal stromal lineage originating from postnatal LTβR+ perivascular stromal progenitors. The genetic blockage of this lineage increased the intestinal stem cell pool while decreasing epithelial and immune maturation at weaning age, leading to reduced postnatal growth and dysregulated repair responses. Ablating PDGFRα in the LTBR stromal lineage demonstrates that PDGFRα has a major impact on the lineage fate and function, inducing a transcriptomic switch from prostemness genes, such as Rspo3 and Grem1, to prodifferentiation factors, including BMPs, retinoic acid, and laminins, and on spatial organization within the crypt-villus and repair responses. Our results show that the PDGFRα-induced transcriptomic switch in intestinal stromal cells is required in the first weeks after birth to coordinate postnatal intestinal maturation and function.
Collapse
Affiliation(s)
- Jean-Marie Jacob
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Selene E Di Carlo
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Igor Stzepourginski
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Anthony Lepelletier
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Papa Diogop Ndiaye
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Hugo Varet
- Transcriptome and Epigenome Platform-Biomics Pole, Institut Pasteur, Université Paris Cité, Paris, France; Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Rachel Legendre
- Transcriptome and Epigenome Platform-Biomics Pole, Institut Pasteur, Université Paris Cité, Paris, France; Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Etienne Kornobis
- Transcriptome and Epigenome Platform-Biomics Pole, Institut Pasteur, Université Paris Cité, Paris, France; Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Adam Benabid
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Giulia Nigro
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Lucie Peduto
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France.
| |
Collapse
|
13
|
Lenti E, Genovese L, Bianchessi S, Maurizio A, Sain SB, di Lillo A, Mattavelli G, Harel I, Bernassola F, Hehlgans T, Pfeffer K, Crosti M, Abrignani S, Evans SM, Sitia G, Guimarães-Camboa N, Russo V, van de Pavert SA, Garcia-Manteiga JM, Brendolan A. Fate mapping and scRNA sequencing reveal origin and diversity of lymph node stromal precursors. Immunity 2022; 55:606-622.e6. [PMID: 35358427 DOI: 10.1016/j.immuni.2022.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/30/2021] [Accepted: 03/03/2022] [Indexed: 11/25/2022]
Abstract
Lymph node (LN) stromal cells play a crucial role in LN development and in supporting adaptive immune responses. However, their origin, differentiation pathways, and transcriptional programs are still elusive. Here, we used lineage-tracing approaches and single-cell transcriptome analyses to determine origin, transcriptional profile, and composition of LN stromal and endothelial progenitors. Our results showed that all major stromal cell subsets and a large proportion of blood endothelial cells originate from embryonic Hoxb6+ progenitors of the lateral plate mesoderm (LPM), whereas lymphatic endothelial cells arise from Pax3+ progenitors of the paraxial mesoderm (PXM). Single-cell RNA sequencing revealed the existence of different Cd34+ and Cxcl13+ stromal cell subsets and showed that embryonic LNs contain proliferating progenitors possibly representing the amplifying populations for terminally differentiated cells. Taken together, our work identifies the earliest embryonic sources of LN stromal and endothelial cells and demonstrates that stromal diversity begins already during LN development.
Collapse
Affiliation(s)
- Elisa Lenti
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Genovese
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Bianchessi
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Aurora Maurizio
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Simona Baghai Sain
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessia di Lillo
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Greta Mattavelli
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Itamar Harel
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 9190401, Israel
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Thomas Hehlgans
- Leibniz Institute of Immunotherapy (LIT), Chair for Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Klaus Pfeffer
- Institute of Medical, Microbiology and Hospital Hygiene, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
| | - Mariacristina Crosti
- INGM, Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy
| | - Sergio Abrignani
- INGM, Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy; Department of Clinical Science and Community Health (DISCCO), University of Milan, Milan, Italy
| | - Sylvia M Evans
- Skaggs School of Pharmacy, University of California at San Diego, La Jolla, CA 92093, USA
| | - Giovanni Sitia
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nuno Guimarães-Camboa
- Institute of Cardiovascular Regeneration, Goethe-University, Frankfurt 60590, Germany; German Center for Cardiovascular Research, Berlin (partner site Frankfurt Rhine-Main), Germany
| | - Vincenzo Russo
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serge A van de Pavert
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, INSERM, CNRS, Marseille, France
| | | | - Andrea Brendolan
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
14
|
Makris S, de Winde CM, Horsnell HL, Cantoral-Rebordinos JA, Finlay RE, Acton SE. Immune function and dysfunction are determined by lymphoid tissue efficacy. Dis Model Mech 2022; 15:dmm049256. [PMID: 35072206 PMCID: PMC8807573 DOI: 10.1242/dmm.049256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Lymphoid tissue returns to a steady state once each immune response is resolved, and although this occurs multiple times throughout life, its structural integrity and functionality remain unaffected. Stromal cells orchestrate cellular interactions within lymphoid tissue, and any changes to the microenvironment can have detrimental outcomes and drive disease. A breakdown in lymphoid tissue homeostasis can lead to a loss of tissue structure and function that can cause aberrant immune responses. This Review highlights recent advances in our understanding of lymphoid tissue function and remodelling in adaptive immunity and in disease states. We discuss the functional role of lymphoid tissue in disease progression and explore the changes to lymphoid tissue structure and function driven by infection, chronic inflammatory conditions and cancer. Understanding the role of lymphoid tissues in immune responses to a wide range of pathologies allows us to take a fuller systemic view of disease progression.
Collapse
Affiliation(s)
- Spyridon Makris
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Charlotte M. de Winde
- Department for Molecular Cell Biology and Immunology, Amsterdam UMC, location VUmc, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands
| | - Harry L. Horsnell
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Jesús A. Cantoral-Rebordinos
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Rachel E. Finlay
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester M13 9PL, UK
| | - Sophie E. Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
15
|
Matrix biophysical cues direct mesenchymal stromal cell functions in immunity. Acta Biomater 2021; 133:126-138. [PMID: 34365041 DOI: 10.1016/j.actbio.2021.07.075] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 07/14/2021] [Accepted: 07/30/2021] [Indexed: 12/25/2022]
Abstract
Hydrogels have been used to design synthetic matrices that capture salient features of matrix microenvironments to study and control cellular functions. Recent advances in understanding of both extracellular matrix biology and biomaterial design have shown that biophysical cues are powerful mediators of cell biology, especially that of mesenchymal stromal cells (MSCs). MSCs have been tested in many clinical trials because of their ability to modulate immune cells in different pathological conditions. While roles of biophysical cues in MSC biology have been studied in the context of multilineage differentiation, their significance in regulating immunomodulatory functions of MSCs is just beginning to be elucidated. This review first describes design principles behind how biophysical cues in native microenvironments influence the ability of MSCs to regulate immune cell production and functions. We will then discuss how biophysical cues can be leveraged to optimize cell isolation, priming, and delivery, which can help improve the success of MSC therapy for immunomodulation. Finally, a perspective is presented on how implementing biophysical cues in MSC potency assay can be important in predicting clinical outcomes. STATEMENT OF SIGNIFICANCE: Stromal cells of mesenchymal origin are known to direct immune cell functions in vivo by secreting paracrine mediators. This property has been leveraged in developing mesenchymal stromal cell (MSC)-based therapeutics by adoptive transfer to treat immunological rejection and tissue injuries, which have been tested in over one thousand clinical trials to date, but with mixed success. Advances in biomaterial design have enabled precise control of biophysical cues based on how stromal cells interact with the extracellular matrix in microenvironments in situ. Investigators have begun to use this approach to understand how different matrix biophysical parameters, such as fiber orientation, porosity, dimensionality, and viscoelasticity impact stromal cell-mediated immunomodulation. The insights gained from this effort can potentially be used to precisely define the microenvironmental cues for isolation, priming, and delivery of MSCs, which can be tailored based on different disease indications for optimal therapeutic outcomes.
Collapse
|
16
|
Martin-Almedina S, Mortimer PS, Ostergaard P. Development and physiological functions of the lymphatic system: insights from human genetic studies of primary lymphedema. Physiol Rev 2021; 101:1809-1871. [PMID: 33507128 DOI: 10.1152/physrev.00006.2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Primary lymphedema is a long-term (chronic) condition characterized by tissue lymph retention and swelling that can affect any part of the body, although it usually develops in the arms or legs. Due to the relevant contribution of the lymphatic system to human physiology, while this review mainly focuses on the clinical and physiological aspects related to the regulation of fluid homeostasis and edema, clinicians need to know that the impact of lymphatic dysfunction with a genetic origin can be wide ranging. Lymphatic dysfunction can affect immune function so leading to infection; it can influence cancer development and spread, and it can determine fat transport so impacting on nutrition and obesity. Genetic studies and the development of imaging techniques for the assessment of lymphatic function have enabled the recognition of primary lymphedema as a heterogenic condition in terms of genetic causes and disease mechanisms. In this review, the known biological functions of several genes crucial to the development and function of the lymphatic system are used as a basis for understanding normal lymphatic biology. The disease conditions originating from mutations in these genes are discussed together with a detailed clinical description of the phenotype and the up-to-date knowledge in terms of disease mechanisms acquired from in vitro and in vivo research models.
Collapse
Affiliation(s)
- Silvia Martin-Almedina
- Molecular and Clinical Sciences Institute, St. George's University of London, London, United Kingdom
| | - Peter S Mortimer
- Molecular and Clinical Sciences Institute, St. George's University of London, London, United Kingdom
- Dermatology and Lymphovascular Medicine, St. George's Universities NHS Foundation Trust, London, United Kingdom
| | - Pia Ostergaard
- Molecular and Clinical Sciences Institute, St. George's University of London, London, United Kingdom
| |
Collapse
|
17
|
Asam S, Nayar S, Gardner D, Barone F. Stromal cells in tertiary lymphoid structures: Architects of autoimmunity. Immunol Rev 2021; 302:184-195. [PMID: 34060101 DOI: 10.1111/imr.12987] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
The molecular mediators present within the inflammatory microenvironment are able, in certain conditions, to favor the initiation of tertiary lymphoid structure (TLS) development. TLS is organized lymphocyte clusters able to support antigen-specific immune response in non-immune organs. Importantly, chronic inflammation does not always result in TLS formation; instead, TLS has been observed to develop specifically in permissive organs, suggesting the presence of tissue-specific cues that are able to imprint the immune responses and form TLS hubs. Fibroblasts are tissue-resident cells that define the anatomy and function of a specific tissue. Fibroblast plasticity and specialization in inflammatory conditions have recently been unraveled in both immune and non-immune organs revealing a critical role for these structural cells in human physiology. Here, we describe the role of fibroblasts in the context of TLS formation and its functional maintenance in the tissue, highlighting their potential role as therapeutic disease targets in TLS-associated diseases.
Collapse
Affiliation(s)
- Saba Asam
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Saba Nayar
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, UK
| | - David Gardner
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Francesca Barone
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
18
|
Lütge M, Pikor NB, Ludewig B. Differentiation and activation of fibroblastic reticular cells. Immunol Rev 2021; 302:32-46. [PMID: 34046914 PMCID: PMC8361914 DOI: 10.1111/imr.12981] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/17/2021] [Accepted: 04/30/2021] [Indexed: 12/29/2022]
Abstract
Secondary lymphoid organs (SLO) are underpinned by fibroblastic reticular cells (FRC) that form dedicated microenvironmental niches to secure induction and regulation of innate and adaptive immunity. Distinct FRC subsets are strategically positioned in SLOs to provide niche factors and govern efficient immune cell interaction. In recent years, the use of specialized mouse models in combination with single-cell transcriptomics has facilitated the elaboration of the molecular FRC landscape at an unprecedented resolution. While single-cell RNA-sequencing has advanced the resolution of FRC subset characterization and function, the high dimensionality of the generated data necessitates careful analysis and validation. Here, we reviewed novel findings from high-resolution transcriptomic analyses that refine our understanding of FRC differentiation and activation processes in the context of infection and inflammation. We further discuss concepts, strategies, and limitations for the analysis of single-cell transcriptome data from FRCs and the wide-ranging implications for our understanding of stromal cell biology.
Collapse
Affiliation(s)
- Mechthild Lütge
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Natalia B Pikor
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
19
|
Nosenko MA, Moysenovich AM, Arkhipova AY, Atretkhany KSN, Nedospasov SA, Drutskaya MS, Moisenovich MM. Fibroblasts upregulate expression of adhesion molecules and promote lymphocyte retention in 3D fibroin/gelatin scaffolds. Bioact Mater 2021; 6:3449-3460. [PMID: 33817420 PMCID: PMC7988350 DOI: 10.1016/j.bioactmat.2021.03.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/05/2021] [Accepted: 03/04/2021] [Indexed: 01/16/2023] Open
Abstract
Bioengineered scaffolds are crucial components in artificial tissue construction. In general, these scaffolds provide inert three-dimensional (3D) surfaces supporting cell growth. However, some scaffolds can affect the phenotype of cultured cells, especially, adherent stromal cells, such as fibroblasts. Here we report on unique properties of 3D fibroin/gelatin materials, which may rapidly induce expression of adhesion molecules, such as ICAM-1 and VCAM-1, in cultured primary murine embryonic fibroblasts (MEFs). In contrast, two-dimensional (2D) fibroin/gelatin films did not show significant effects on gene expression profiles in fibroblasts as compared to 3D culture conditions. Interestingly, TNF expression was induced in MEFs cultured in 3D fibroin/gelatin scaffolds, while genetic or pharmacological TNF ablation resulted in diminished ICAM-1 and VCAM-1 expression by these cells. Using selective MAPK inhibitors, we uncovered critical contribution of JNK to 3D-induced upregulation of these adhesion molecules. Moreover, we observed ICAM-1/VCAM-1-dependent adhesion of lymphocytes to fibroblasts cultured in 3D fibroin/gelatin scaffolds, but not on 2D fibroin/gelatin films, suggesting functional reprogramming in stromal cells, when exposed to 3D environment. Finally, we observed significant infiltration of lymphocytes into 3D fibroin/gelatin, but not into collagen scaffolds in vivo upon subcapsular kidney implantation in mice. Together our data highlight the important features of fibroin/gelatin scaffolds, when they are produced as 3D sponges rather than 2D films, which should be considered when using these materials for tissue engineering. 3D, but not 2D fibroin-based scaffolds promote expression of adhesion molecules in murine fibroblasts. Overexpression of adhesion molecules in 3D fibroin/gelatin-cultured fibroblasts is TNF- and JNK-dependent. Culturing of fibroblasts in 3D fibroin/gelatin scaffolds promotes adhesion of T-lymphocytes. Implantation of 3D fibroin/gelatin scaffolds in vivo induces infiltration and clustering of T- and B-lymphocytes.
Collapse
Affiliation(s)
- Maxim A Nosenko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | | | - Anastasia Y Arkhipova
- Biological Faculty, Lomonosov Moscow State University, Moscow, 119991, Russia.,Biological Faculty, Shenzhen MSU-BIT University, Shenzhen, 518172, China
| | - Kamar-Sulu N Atretkhany
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Sergei A Nedospasov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.,Biological Faculty, Lomonosov Moscow State University, Moscow, 119991, Russia.,Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, 354340, Russia
| | - Marina S Drutskaya
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | | |
Collapse
|
20
|
Pikor NB, Cheng HW, Onder L, Ludewig B. Development and Immunological Function of Lymph Node Stromal Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:257-263. [PMID: 33397739 DOI: 10.4049/jimmunol.2000914] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/16/2020] [Indexed: 01/07/2023]
Abstract
Stromal cells have for a long time been viewed as structural cells that support distinct compartments within lymphoid tissues and little more. Instead, an active cross-talk between endothelial and fibroblastic stromal cells drives the maturation of lymphoid niches, a relationship that is recapitulated during lymph node organogenesis, steady-state conditions, and following inflammation. In this review, we go over recent advances in genetic models and high-resolution transcriptomic analyses that have propelled the finer resolution of the stromal cell infrastructure of lymph nodes, revealing that the distinct subsets are strategically positioned to deliver a catered mixture of niche factors to interacting immune cell populations. Moreover, we discuss how changes in the activation state of poised stromal cell-underpinned niches rather than on-demand differentiation of new stromal cell subsets govern the efficient interaction of Ag, APC, and cognate B and T lymphocytes during adaptive immune responses.
Collapse
Affiliation(s)
- Natalia Barbara Pikor
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, CH-9007 St. Gallen, Switzerland; and
| | - Hung-Wei Cheng
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, CH-9007 St. Gallen, Switzerland; and
| | - Lucas Onder
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, CH-9007 St. Gallen, Switzerland; and
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, CH-9007 St. Gallen, Switzerland; and .,Institute of Experimental Immunology, University of Zürich, 8006 Zürich, Switzerland
| |
Collapse
|
21
|
Furukawa M, Ito S, Suzuki S, Fuchimoto D, Onishi A, Niimi K, Usami K, Wu G, Bazer FW, Ogasawara K, Watanabe K, Aso H, Nochi T. Organogenesis of Ileal Peyer's Patches Is Initiated Prenatally and Accelerated Postnatally With Comprehensive Proliferation of B Cells in Pigs. Front Immunol 2020; 11:604674. [PMID: 33424851 PMCID: PMC7793923 DOI: 10.3389/fimmu.2020.604674] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/05/2020] [Indexed: 11/21/2022] Open
Abstract
Morphogenesis and differentiation of organs is required for subsequent functional maturation. The morphological features of Peyer's patches vary among species. In pigs, they develop extensively in the ileum as ileal Peyer's patches (IPPs). However, the role of IPPs in the porcine immune system remains to be elucidated because of a lack of complete understanding of IPP organogenesis. Results of the present study revealed that development of porcine IPPs is initiated prenatally between embryonic days 76 and 91. The process of IPP organogenesis is concomitant with increased transcriptional patterns of CXCL13 and CCL19. IPPs undergo further development postnatally by forming central, marginal, and subepithelial zones. Importantly, a large number of proliferating B cells and apoptotic cells are found in porcine IPPs postnatally, but not prenatally. The expression level of IgM in proliferating B cells depends on the zone in which distinct B cells are separately localized after birth. Specifically, IgM+ cells are predominantly found in the central zone, whereas IgM-/low cells are abundant in the marginal zone. Importantly, the cellular feature of IPPs differs from that of mesenteric lymph nodes (MLNs) where such distinct zones are not formed both prenatally and postnatally. Our findings suggest that IPPs (not MLNs) in postnatal pigs are involved in complementing functions of the primary lymphoid tissue that promotes the differentiation and maturation of B cells.
Collapse
Affiliation(s)
- Mutsumi Furukawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shun Ito
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shunichi Suzuki
- Division of Animal Science, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Daiichiro Fuchimoto
- Division of Animal Science, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Akira Onishi
- Department of Animal Science and Resources, Nihon University College of Bioresource Sciences, Fujisawa, Japan
| | - Kanae Niimi
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Katsuki Usami
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Kouetsu Ogasawara
- Department of Immunobiology, Tohoku University Institute of Development, Aging and Cancer, Sendai, Japan
| | - Kouichi Watanabe
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Hisashi Aso
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Tomonori Nochi
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
22
|
Koliaraki V, Prados A, Armaka M, Kollias G. The mesenchymal context in inflammation, immunity and cancer. Nat Immunol 2020; 21:974-982. [PMID: 32747813 DOI: 10.1038/s41590-020-0741-2] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/16/2020] [Indexed: 12/19/2022]
Abstract
Mesenchymal cells are mesoderm-derived stromal cells that are best known for providing structural support to organs, synthesizing and remodeling the extracellular matrix (ECM) and regulating development, homeostasis and repair of tissues. Recent detailed mechanistic insights into the biology of fibroblastic mesenchymal cells have revealed they are also significantly involved in immune regulation, stem cell maintenance and blood vessel function. It is now becoming evident that these functions, when defective, drive the development of complex diseases, such as various immunopathologies, chronic inflammatory disease, tissue fibrosis and cancer. Here, we provide a concise overview of the contextual contribution of fibroblastic mesenchymal cells in physiology and disease and bring into focus emerging evidence for both their heterogeneity at the single-cell level and their tissue-specific, spatiotemporal functional diversity.
Collapse
Affiliation(s)
- Vasiliki Koliaraki
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.
| | - Alejandro Prados
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Marietta Armaka
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - George Kollias
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece. .,Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece. .,Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
23
|
Jackson-Jones LH, Bénézech C. FALC stromal cells define a unique immunological niche for the surveillance of serous cavities. Curr Opin Immunol 2020; 64:42-49. [PMID: 32353646 DOI: 10.1016/j.coi.2020.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 03/04/2020] [Accepted: 03/10/2020] [Indexed: 01/14/2023]
Abstract
The serous cavities contain specialised adipose tissues which house small clusters of immune cells known as fat-associated lymphoid clusters (FALCs). The continuous flow of fluid from the serous cavities through FALCs makes them unique niches for the clearance of fluid phase contaminants and initiation of locally protective immune responses during infection and inflammation. Development, and activation of FALCs both at homeostasis and following inflammation are co-ordinated by the close interaction of mesothelial and fibroblastic stromal cell populations with immune cells. In this review we discuss recent developments in FALC stromal cell biology and highlight key interactions that occur between FALC stroma and immune cells.
Collapse
Affiliation(s)
- Lucy H Jackson-Jones
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster LA1 4YQ, UK
| | - Cécile Bénézech
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
24
|
Lymph node stromal cells: cartographers of the immune system. Nat Immunol 2020; 21:369-380. [PMID: 32205888 DOI: 10.1038/s41590-020-0635-3] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/17/2020] [Indexed: 01/03/2023]
Abstract
Lymph nodes (LNs) are strategically positioned at dedicated sites throughout the body to facilitate rapid and efficient immunity. Central to the structural integrity and framework of LNs, and the recruitment and positioning of leukocytes therein, are mesenchymal and endothelial lymph node stromal cells (LNSCs). Advances in the last decade have expanded our understanding and appreciation of LNSC heterogeneity, and the role they play in coordinating immunity has grown rapidly. In this review, we will highlight the functional contributions of distinct stromal cell populations during LN development in maintaining immune homeostasis and tolerance and in the activation and control of immune responses.
Collapse
|
25
|
Domingues RG, Hepworth MR. Immunoregulatory Sensory Circuits in Group 3 Innate Lymphoid Cell (ILC3) Function and Tissue Homeostasis. Front Immunol 2020; 11:116. [PMID: 32117267 PMCID: PMC7015949 DOI: 10.3389/fimmu.2020.00116] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/16/2020] [Indexed: 12/12/2022] Open
Abstract
Recent years have seen a revolution in our understanding of how cells of the immune system are modulated and regulated not only via complex interactions with other immune cells, but also through a range of potent inputs derived from diverse and varied biological systems. Within complex tissue environments, such as the gastrointestinal tract and lung, these systems act to orchestrate and temporally align immune responses, regulate cellular function, and ensure tissue homeostasis and protective immunity. Group 3 Innate Lymphoid Cells (ILC3s) are key sentinels of barrier tissue homeostasis and critical regulators of host-commensal mutualism—and respond rapidly to damage, inflammation and infection to restore tissue health. Recent findings place ILC3s as strategic integrators of environmental signals. As a consequence, ILC3s are ideally positioned to detect perturbations in cues derived from the environment—such as the diet and microbiota—as well as signals produced by the host nervous, endocrine and circadian systems. Together these cues act in concert to induce ILC3 effector function, and form critical sensory circuits that continually function to reinforce tissue homeostasis. In this review we will take a holistic, organismal view of ILC3 biology and explore the tissue sensory circuits that regulate ILC3 function and align ILC3 responses with changes within the intestinal environment.
Collapse
Affiliation(s)
- Rita G Domingues
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research, Lydia Becker Institute of Immunology and Inflammation, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Matthew R Hepworth
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research, Lydia Becker Institute of Immunology and Inflammation, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
26
|
YAP/TAZ direct commitment and maturation of lymph node fibroblastic reticular cells. Nat Commun 2020; 11:519. [PMID: 31980640 PMCID: PMC6981200 DOI: 10.1038/s41467-020-14293-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 12/31/2019] [Indexed: 02/07/2023] Open
Abstract
Fibroblastic reticular cells (FRCs) are immunologically specialized myofibroblasts of lymphoid organ, and FRC maturation is essential for structural and functional properties of lymph nodes (LNs). Here we show that YAP and TAZ (YAP/TAZ), the final effectors of Hippo signaling, regulate FRC commitment and maturation. Selective depletion of YAP/TAZ in FRCs impairs FRC growth and differentiation and compromises the structural organization of LNs, whereas hyperactivation of YAP/TAZ enhances myofibroblastic characteristics of FRCs and aggravates LN fibrosis. Mechanistically, the interaction between YAP/TAZ and p52 promotes chemokine expression that is required for commitment of FRC lineage prior to lymphotoxin-β receptor (LTβR) engagement, whereas LTβR activation suppresses YAP/TAZ activity for FRC maturation. Our findings thus present YAP/TAZ as critical regulators of commitment and maturation of FRCs, and hold promise for better understanding of FRC-mediated pathophysiologic processes. Fibroblastic reticular cells (FRC) are important for lymph node (LN) structure and function. Here the authors show that the YAP/TAZ complex downstream of Hippo signalling regulates FRC commitment and maturation, with YAP/TAZ deficiency impairing FRC differentiation, while hyperactivation of YAZ/TAZ inducing myofibroblastic FRCs and LN fibrosis.
Collapse
|
27
|
Eckert N, Permanyer M, Yu K, Werth K, Förster R. Chemokines and other mediators in the development and functional organization of lymph nodes. Immunol Rev 2020; 289:62-83. [PMID: 30977201 DOI: 10.1111/imr.12746] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 01/22/2019] [Indexed: 12/28/2022]
Abstract
Secondary lymphoid organs like lymph nodes (LNs) are the main inductive sites for adaptive immune responses. Lymphocytes are constantly entering LNs, scanning the environment for their cognate antigen and get replenished by incoming cells after a certain period of time. As only a minor percentage of lymphocytes recognizes cognate antigen, this mechanism of permanent recirculation ensures fast and effective immune responses when necessary. Thus, homing, positioning, and activation as well as egress require precise regulation within LNs. In this review we discuss the mediators, including chemokines, cytokines, growth factors, and others that are involved in the formation of the LN anlage and subsequent functional organization of LNs. We highlight very recent findings in the fields of LN development, steady-state migration in LNs, and the intranodal processes during an adaptive immune response.
Collapse
Affiliation(s)
- Nadine Eckert
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Marc Permanyer
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Kai Yu
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Kathrin Werth
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
28
|
McFarland AP, Colonna M. Sense and immuno-sensibility: innate lymphoid cell niches and circuits. Curr Opin Immunol 2019; 62:9-14. [PMID: 31825814 DOI: 10.1016/j.coi.2019.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/11/2019] [Indexed: 01/05/2023]
Abstract
Tissue-resident lymphocytes that lack expression of rearranged antigen receptors and are lineage negative for classical T and B cell markers are collectively known as innate lymphoid cells (ILCs). The ILC family is remarkably heterogeneous and exhibits plasticity; however, mature ILCs can be grouped based on their steady state expression of distinct surface receptors and transcription factors as well as production of signature cytokines following activation. The study of ILC subsets in mouse and human tissues has revealed that the elicitation and magnitude of their effector functions are determined by a combination of extrinsic cues specific to the niches in which they reside. In this short review, we will summarize some recent findings related to tissue-specific signals that govern ILC responses and localization.
Collapse
Affiliation(s)
- Adelle P McFarland
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, United States.
| |
Collapse
|
29
|
Jeucken KCM, Koning JJ, Mebius RE, Tas SW. The Role of Endothelial Cells and TNF-Receptor Superfamily Members in Lymphoid Organogenesis and Function During Health and Inflammation. Front Immunol 2019; 10:2700. [PMID: 31824495 PMCID: PMC6879661 DOI: 10.3389/fimmu.2019.02700] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/04/2019] [Indexed: 01/02/2023] Open
Abstract
Lymph nodes (LNs) are crucial for the orchestration of immune responses. LN reactions depend on interactions between incoming and local immune cells, and stromal cells. To mediate these cellular interactions an organized vascular network within the LN exists. In general, the LN vasculature can be divided into two components: blood vessels, which include the specialized high endothelial venules that recruit lymphocytes from the bloodstream, and lymphatic vessels. Signaling via TNF receptor (R) superfamily (SF) members has been implicated as crucial for the development and function of LNs and the LN vasculature. In recent years the role of cell-specific signaling of TNFRSF members in different endothelial cell (EC) subsets and their roles in development and maintenance of lymphoid organs has been elucidated. Here, we discuss recent insights into EC-specific TNFRSF member signaling and highlight its importance in different EC subsets in LN organogenesis and function during health, and in lymphocyte activation and tertiary lymphoid structure formation during inflammation.
Collapse
Affiliation(s)
- Kim C M Jeucken
- Amsterdam Rheumatology and Immunology Center (ARC), Department of Rheumatology and Clinical Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jasper J Koning
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center (ARC), Department of Rheumatology and Clinical Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
30
|
Kaipainen A, Chen E, Chang L, Zhao B, Shin H, Stahl A, Fishman SJ, Mulliken JB, Folkman J, Huang S, Fannon M. Characterization of lymphatic malformations using primary cells and tissue transcriptomes. Scand J Immunol 2019; 90:e12800. [DOI: 10.1111/sji.12800] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/10/2019] [Accepted: 06/22/2019] [Indexed: 01/25/2023]
Affiliation(s)
- Arja Kaipainen
- Vascular Biology Program, Department of Surgery Harvard Medical School, Boston Children's Hospital Boston MA USA
| | - Emy Chen
- Vascular Biology Program, Department of Surgery Harvard Medical School, Boston Children's Hospital Boston MA USA
| | - Lynn Chang
- Vascular Biology Program, Department of Surgery Harvard Medical School, Boston Children's Hospital Boston MA USA
| | - Bing Zhao
- Department of Ophthalmology and Visual Sciences University of Kentucky Lexington KY USA
| | - Hainsworth Shin
- Department of Biomedical Engineering University of Kentucky Lexington KY USA
| | - Andreas Stahl
- Vascular Biology Program, Department of Surgery Harvard Medical School, Boston Children's Hospital Boston MA USA
| | - Steven J. Fishman
- Department of Surgery Harvard Medical School, Boston Children's Hospital Boston MA USA
| | - John B. Mulliken
- Department of Plastic and Oral Surgery, Department of Surgery Harvard Medical School, Boston Children's Hospital Boston MA USA
| | - Judah Folkman
- Vascular Biology Program, Department of Surgery Harvard Medical School, Boston Children's Hospital Boston MA USA
| | - Sui Huang
- Vascular Biology Program, Department of Surgery Harvard Medical School, Boston Children's Hospital Boston MA USA
| | - Michael Fannon
- Vascular Biology Program, Department of Surgery Harvard Medical School, Boston Children's Hospital Boston MA USA
- Department of Ophthalmology and Visual Sciences University of Kentucky Lexington KY USA
| |
Collapse
|
31
|
Denton AE, Carr EJ, Magiera LP, Watts AJB, Fearon DT. Embryonic FAP + lymphoid tissue organizer cells generate the reticular network of adult lymph nodes. J Exp Med 2019; 216:2242-2252. [PMID: 31324739 PMCID: PMC6780995 DOI: 10.1084/jem.20181705] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 05/17/2019] [Accepted: 06/27/2019] [Indexed: 01/03/2023] Open
Abstract
The induction of adaptive immunity is dependent on the structural organization of LNs, which is in turn governed by the stromal cells that underpin LN architecture. Using a novel fate-mapping mouse model, we trace the developmental origin of mesenchymal LN stromal cells (mLNSCs) to a previously undescribed embryonic fibroblast activation protein-α (FAP)+ progenitor. FAP+ cells of the LN anlagen express lymphotoxin β receptor (LTβR) and vascular cell adhesion molecule (VCAM), but not intercellular adhesion molecule (ICAM), suggesting they are early mesenchymal lymphoid tissue organizer (mLTo) cells. Clonal labeling shows that FAP+ progenitors locally differentiate into mLNSCs. This process is also coopted in nonlymphoid tissues in response to infection to facilitate the development of tertiary lymphoid structures, thereby mimicking the process of LN ontogeny in response to infection.
Collapse
Affiliation(s)
- Alice E Denton
- Lymphocyte Signaling and Development, Babraham Institute, Cambridge, UK .,Department of Medicine, University of Cambridge, Cambridge, UK.,Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Edward J Carr
- Lymphocyte Signaling and Development, Babraham Institute, Cambridge, UK.,Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lukasz P Magiera
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Andrew J B Watts
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Douglas T Fearon
- Department of Medicine, University of Cambridge, Cambridge, UK.,Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.,Weill Cornell Medicine and Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| |
Collapse
|
32
|
Díaz-Flores L, Gutiérrez R, Pino García M, González-Gómez M, Díaz-Flores L, Carrasco JL. Intussusceptive lymphangiogenesis in the sinuses of developing human foetal lymph nodes. Ann Anat 2019; 226:73-83. [PMID: 31279869 DOI: 10.1016/j.aanat.2019.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 03/28/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023]
Abstract
A meshwork of intraluminal processes in lymph node (LN) sinuses originates during LN development. Lymph flows through the meshwork, which has an important role in immunology and pathology. However, the formation mechanism of intraluminal processes has not been sufficiently studied. Our objective is to assess whether this mechanism is by intussusception, as occurs in transcapillary pillar formation in blood vessel intussusceptive angiogenesis. For this purpose, LNs with developing intrasinusal processes were used (human foetuses, 13-18GW) for serial histologic sections and immunohistochemical procedures. The studies showed (a) sinuses originating from lymphatic sacs around expanded LN anlagen, (b) intra-sinus structures (lined by anti-podoplanin+, VEGFR3+, Prox-1+, CD31+ lymphatic endothelial cells) with characteristics (in serial sections and 3D images) similar to those considered the hallmarks of intussusceptive angiogenesis, including pillars (≤2.5μm, with a collagen core), interstitial tissue structures (ITSs) or larger pillars (>2.5μm, with a more cellular core) and folds (that form pillars when spanning), and (c) remodelled and fused pillars, ITSs and folds, which formed meshworks, compartmentalizing the sinuses into small intercommunicating spaces (segmentation). In conclusion, intussusception participates in the formation of the meshwork of processes in LN sinuses during LN development. This mechanism is also of interest because it contributes to the general knowledge of intussusceptive lymphangiogenesis (which has received less attention than intussusception in blood vessels), provides a basis for further studies and supports a new role for vessel intussusception (formation of an intraluminal meshwork with known action in fluid filtering, cell interactions and immunology).
Collapse
Affiliation(s)
- Lucio Díaz-Flores
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, Tenerife, Spain.
| | - Ricardo Gutiérrez
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| | | | - Miriam González-Gómez
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| | - Lucio Díaz-Flores
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| | - José Luis Carrasco
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| |
Collapse
|
33
|
Nayar S, Campos J, Smith CG, Iannizzotto V, Gardner DH, Mourcin F, Roulois D, Turner J, Sylvestre M, Asam S, Glaysher B, Bowman SJ, Fearon DT, Filer A, Tarte K, Luther SA, Fisher BA, Buckley CD, Coles MC, Barone F. Immunofibroblasts are pivotal drivers of tertiary lymphoid structure formation and local pathology. Proc Natl Acad Sci U S A 2019; 116:13490-13497. [PMID: 31213547 PMCID: PMC6613169 DOI: 10.1073/pnas.1905301116] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Resident fibroblasts at sites of infection, chronic inflammation, or cancer undergo phenotypic and functional changes to support leukocyte migration and, in some cases, aggregation into tertiary lymphoid structures (TLS). The molecular programming that shapes these changes and the functional requirements of this population in TLS development are unclear. Here, we demonstrate that external triggers at mucosal sites are able to induce the progressive differentiation of a population of podoplanin (pdpn)-positive stromal cells into a network of immunofibroblasts that are able to support the earliest phases of TLS establishment. This program of events, that precedes lymphocyte infiltration in the tissue, is mediated by paracrine and autocrine signals mainly regulated by IL13. This initial fibroblast network is expanded and stabilized, once lymphocytes are recruited, by the local production of the cytokines IL22 and lymphotoxin. Interfering with this regulated program of events or depleting the immunofibroblasts in vivo results in abrogation of local pathology, demonstrating the functional role of immunofibroblasts in supporting TLS maintenance in the tissue and suggesting novel therapeutic targets in TLS-associated diseases.
Collapse
Affiliation(s)
- Saba Nayar
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, B15 2WB Birmingham, United Kingdom
- bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, B15 2TT, Birmingham, UK
| | - Joana Campos
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, B15 2WB Birmingham, United Kingdom
- bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, B15 2TT, Birmingham, UK
| | - Charlotte G Smith
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, B15 2WB Birmingham, United Kingdom
- bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, B15 2TT, Birmingham, UK
| | - Valentina Iannizzotto
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, B15 2WB Birmingham, United Kingdom
- bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, B15 2TT, Birmingham, UK
| | - David H Gardner
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, B15 2WB Birmingham, United Kingdom
- bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, B15 2TT, Birmingham, UK
| | - Frédéric Mourcin
- UMR INSERM U1236, Université Rennes 1, Etablissement Français du Sang, 35043 Rennes, France
| | - David Roulois
- UMR INSERM U1236, Université Rennes 1, Etablissement Français du Sang, 35043 Rennes, France
| | - Jason Turner
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, B15 2WB Birmingham, United Kingdom
- bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, B15 2TT, Birmingham, UK
| | - Marvin Sylvestre
- UMR INSERM U1236, Université Rennes 1, Etablissement Français du Sang, 35043 Rennes, France
| | - Saba Asam
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, B15 2WB Birmingham, United Kingdom
- bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, B15 2TT, Birmingham, UK
| | - Bridget Glaysher
- Centre for Immunology and Infection, Department of Biology, Hull York Medical School, University of York, YO10 5DD York, United Kingdom
| | - Simon J Bowman
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, B15 2WB Birmingham, United Kingdom
- bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, B15 2TT, Birmingham, UK
| | - Douglas T Fearon
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, CB2 0RE Cambridge, United Kingdom
| | - Andrew Filer
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, B15 2WB Birmingham, United Kingdom
- bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, B15 2TT, Birmingham, UK
| | - Karin Tarte
- UMR INSERM U1236, Université Rennes 1, Etablissement Français du Sang, 35043 Rennes, France
| | - Sanjiv A Luther
- Department of Biochemistry, Center of Immunity and Infection, University of Lausanne, 1066 Epalinges, Switzerland
| | - Benjamin A Fisher
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, B15 2WB Birmingham, United Kingdom
- bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, B15 2TT, Birmingham, UK
| | - Christopher D Buckley
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, B15 2WB Birmingham, United Kingdom
- bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, B15 2TT, Birmingham, UK
| | - Mark C Coles
- Centre for Immunology and Infection, Department of Biology, Hull York Medical School, University of York, YO10 5DD York, United Kingdom;
| | - Francesca Barone
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, B15 2WB Birmingham, United Kingdom;
- bNIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, B15 2TT, Birmingham, UK
| |
Collapse
|
34
|
Camara A, Cordeiro OG, Alloush F, Sponsel J, Chypre M, Onder L, Asano K, Tanaka M, Yagita H, Ludewig B, Flacher V, Mueller CG. Lymph Node Mesenchymal and Endothelial Stromal Cells Cooperate via the RANK-RANKL Cytokine Axis to Shape the Sinusoidal Macrophage Niche. Immunity 2019; 50:1467-1481.e6. [DOI: 10.1016/j.immuni.2019.05.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 04/06/2019] [Accepted: 05/15/2019] [Indexed: 12/15/2022]
|
35
|
Barron AMS, Mantero JC, Ho JD, Nazari B, Horback KL, Bhawan J, Lafyatis R, Lam C, Browning JL. Perivascular Adventitial Fibroblast Specialization Accompanies T Cell Retention in the Inflamed Human Dermis. THE JOURNAL OF IMMUNOLOGY 2018; 202:56-68. [PMID: 30510068 DOI: 10.4049/jimmunol.1801209] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Perivascular accumulation of lymphocytes can be a prominent histopathologic feature of various human inflammatory skin diseases. Select examples include systemic sclerosis, spongiotic dermatitis, and cutaneous lupus. Although a large body of work has described various aspects of the endothelial and vascular smooth muscle layers in these diseases, the outer adventitial compartment is poorly explored. The goal of the current study was to characterize perivascular adventitial fibroblast states in inflammatory human skin diseases and relate these states to perivascular lymphocyte accumulation. In normal skin, adventitial fibroblasts are distinguished by CD90 expression, and dense perivascular lymphocytic infiltrates are uncommon. In systemic sclerosis, this compartment expands, but lymphocyte infiltrates remain sparse. In contrast, perivascular adventitial fibroblast expression of VCAM1 is upregulated in spongiotic dermatitis and lupus and is associated with a dense perivascular T cell infiltrate. VCAM1 expression marks transitioned fibroblasts that show some resemblance to the reticular stromal cells in secondary lymphoid organs. Expanded adventitial compartments with perivascular infiltrates similar to the human settings were not seen in the inflamed murine dermis. This species difference may hinder the dissection of aspects of perivascular adventitial pathology. The altered perivascular adventitial compartment and its associated reticular network form a niche for lymphocytes and appear to be fundamental in the development of an inflammatory pattern.
Collapse
Affiliation(s)
- Alexander M S Barron
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| | - Julio C Mantero
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| | - Jonathan D Ho
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Banafsheh Nazari
- Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118
| | - Katharine L Horback
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| | - Jag Bhawan
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Robert Lafyatis
- Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118.,Division of Rheumatology and Clinical Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213
| | - Christina Lam
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Jeffrey L Browning
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118; .,Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118
| |
Collapse
|
36
|
Koning JJ, Mebius RE. Complexity of Lymphoid Tissue Organizers: A Response to Onder and Ludewig. Trends Immunol 2018; 39:951-952. [DOI: 10.1016/j.it.2018.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 02/02/2023]
|
37
|
Onder L, Ludewig B. A Fresh View on Lymph Node Organogenesis. Trends Immunol 2018; 39:775-787. [DOI: 10.1016/j.it.2018.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/07/2018] [Accepted: 08/07/2018] [Indexed: 01/18/2023]
|
38
|
Zhu G, Nemoto S, Mailloux AW, Perez-Villarroel P, Nakagawa R, Falahat R, Berglund AE, Mulé JJ. Induction of Tertiary Lymphoid Structures With Antitumor Function by a Lymph Node-Derived Stromal Cell Line. Front Immunol 2018; 9:1609. [PMID: 30061886 PMCID: PMC6054958 DOI: 10.3389/fimmu.2018.01609] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/28/2018] [Indexed: 02/03/2023] Open
Abstract
Tertiary lymphoid structures (TLSs) associate with better prognosis in certain cancer types, but their underlying formation and immunological benefit remain to be determined. We established a mouse model of TLSs to study their contribution to antitumor immunity. Because the stroma in lymph nodes (sLN) participates in architectural support, lymphogenesis, and lymphocyte recruitment, we hypothesized that TLSs can be created by sLN. We selected a sLN line with fibroblast morphology that expressed sLN surface markers and lymphoid chemokines. The subcutaneous injection of the sLN line successfully induced TLSs that attracted infiltration of host immune cell subsets. Injection of MC38 tumor lysate-pulsed dendritic cells activated TLS-residing lymphocytes to demonstrate specific cytotoxicity. The presence of TLSs suppressed MC38 tumor growth in vivo by improving antitumor activity of tumor-infiltrating lymphocytes with downregulated immune checkpoint proteins (PD-1 and Tim-3). Future engineering of sLN lines may allow for further enhancements of TLS functions and immune cell compositions.
Collapse
Affiliation(s)
- Genyuan Zhu
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Satoshi Nemoto
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Adam W Mailloux
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | | | - Ryosuke Nakagawa
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Rana Falahat
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Anders E Berglund
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, United States
| | - James J Mulé
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States.,Cutaneous Oncology Program, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
39
|
Dorraji SE, Hovd AMK, Kanapathippillai P, Bakland G, Eilertsen GØ, Figenschau SL, Fenton KA. Mesenchymal stem cells and T cells in the formation of Tertiary Lymphoid Structures in Lupus Nephritis. Sci Rep 2018; 8:7861. [PMID: 29777158 PMCID: PMC5959845 DOI: 10.1038/s41598-018-26265-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/09/2018] [Indexed: 12/13/2022] Open
Abstract
Tertiary lymphoid structures (TLS) develop in the kidneys of lupus-prone mice and systemic lupus erythematosus (SLE) patients with lupus nephritis (LN). Here we investigated the presence of mesenchymal stem cells (MSCs) in the development of TLS in murine LN, as well as the role of human MSCs as lymphoid tissue organizer (LTo) cells on the activation of CD4+ T cells from three groups of donors including Healthy, SLE and LN patients. Mesenchymal stem like cells were detected within the pelvic wall and TLS in kidneys of lupus-prone mice. An increase in LTβ, CXCL13, CCL19, VCAM1 and ICAM1 gene expressions were detected during the development of murine LN. Human MSCs stimulated with the pro-inflammatory cytokines TNF-α and IL-1β significantly increased the expression of CCL19, VCAM1, ICAM1, TNF-α, and IL-1β. Stimulated MSCs induced proliferation of CD4+ T cells, but an inhibitory effect was observed when in co-culture with non-stimulated MSCs. A contact dependent increase in Th2 and Th17 subsets were observed for T cells from the Healthy group after co-culture with stimulated MSCs. Our data suggest that tissue-specific or/and migratory MSCs could have pivotal roles as LTo cells in accelerating early inflammatory processes and initiating the formation of kidney specific TLS in chronic inflammatory conditions.
Collapse
Affiliation(s)
- S Esmaeil Dorraji
- RNA and Molecular Pathology Research Group, Institute of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Aud-Malin K Hovd
- RNA and Molecular Pathology Research Group, Institute of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Premasany Kanapathippillai
- RNA and Molecular Pathology Research Group, Institute of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Gunnstein Bakland
- University Hospital of Northern Norway, Tromsø, Norway.,Molecular Inflammatory Research Group, Institute of Clinical Medicine, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Gro Østli Eilertsen
- University Hospital of Northern Norway, Tromsø, Norway.,Molecular Inflammatory Research Group, Institute of Clinical Medicine, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Stine L Figenschau
- RNA and Molecular Pathology Research Group, Institute of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Kristin A Fenton
- RNA and Molecular Pathology Research Group, Institute of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
40
|
Wang Z, Chai Q, Zhu M. Differential Roles of LTβR in Endothelial Cell Subsets for Lymph Node Organogenesis and Maturation. THE JOURNAL OF IMMUNOLOGY 2018; 201:69-76. [PMID: 29760194 DOI: 10.4049/jimmunol.1701080] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 04/26/2018] [Indexed: 11/19/2022]
Abstract
Cellular cross-talk mediated by lymphotoxin αβ-lymphotoxin β receptor (LTβR) signaling plays a critical role in lymph node (LN) development. Although the major role of LTβR signaling has long been considered to occur in mesenchymal lymphoid tissue organizer cells, a recent study using a VE-cadherincreLtbrfl/fl mouse model suggested that endothelial LTβR signaling contributes to the formation of LNs. However, the detailed roles of LTβR in different endothelial cells (ECs) in LN development remain unknown. Using various cre transgenic mouse models (Tekcre , a strain targeting ECs, and Lyve1cre , mainly targeting lymphatic ECs), we observed that specific LTβR ablation in Tekcre+ or Lyve1cre+ cells is not required for LN formation. Moreover, double-cre-mediated LTβR depletion does not interrupt LN formation. Nevertheless, TekcreLtbrfl/fl mice exhibit reduced lymphoid tissue inducer cell accumulation at the LN anlagen and impaired LN maturation. Interestingly, a subset of ECs (VE-cadherin+Tekcre-low/neg ECs) was found to be enriched in transcripts related to hematopoietic cell recruitment and transendothelial migration, resembling LN high ECs in adult animals. Furthermore, endothelial Tek was observed to negatively regulate hematopoietic cell transmigration. Taken together, our data suggest that although Tekcre+ endothelial LTβR is required for the accumulation of hematopoietic cells and full LN maturation, LTβR in VE-cadherin+Tekcre-low/neg ECs in embryos might represent a critical portal-determining factor for LN formation.
Collapse
Affiliation(s)
- Zhongnan Wang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; and.,College of Life Sciences, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Chai
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; and
| | - Mingzhao Zhu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; and .,College of Life Sciences, University of the Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
41
|
Prados A, Muñoz-Fernández R, Fernandez-Rubio P, Olivares EG. Characterization of mesenchymal stem/stromal cells with lymphoid tissue organizer cell potential in tonsils from children. Eur J Immunol 2018; 48:829-843. [PMID: 29435977 DOI: 10.1002/eji.201746963] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 12/15/2017] [Accepted: 02/07/2018] [Indexed: 12/18/2022]
Abstract
Lymphoid tissue organizer (LTo) cells, identified in mouse and human embryos, are thought to be precursors of stromal cells in secondary lymphoid organs. Whether LTo cells are present in human adults, however remains unknown. We obtained 15 stromal cell lines from tonsils from children who underwent tonsillectomy, and studied the antigen phenotype of these tonsil stromal cell (TSC) lines by flow cytometry and RT-PCR. Cell lines met the minimal criteria proposed by the International Society for Cellular Therapy to define human mesenchymal stem/stromal cells (MSCs): plastic-adherent capacity; expression of CD73, CD90 and CD105, lack of CD45, CD19 and HLA-DR; and capacity to differentiate into adipocytes, osteoblasts and chondrocytes. Furthermore, our TSC lines exhibited an antigen phenotype and functional characteristics very similar to those seen in murine embryo LTo cells: they expressed chemokines CCL19, CCL21 and CXCL13, cytokines TRANCE and IL-7, and adhesion molecules ICAM-1, mucosal addressin cell adhesion molecule (MadCAM)-1 and VCAM-1. The expression of LTo cell-associated markers and functions were upregulated by lymphotoxin (LT)α1β2 and TNF, two cytokines involved in the development and maturation of secondary lymphoid tissues. Our results show that TSCs are tonsil MSCs that differentiate into LTo-like cells in response to the effects of these cytokines.
Collapse
Affiliation(s)
- Alejandro Prados
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Spain
| | - Raquel Muñoz-Fernández
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Spain
| | - Pablo Fernandez-Rubio
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Spain
| | - Enrique G Olivares
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Spain.,Unidad de Gestión Clínica Laboratorios, Complejo Hospitalario Universitario de Granada, Spain
| |
Collapse
|
42
|
Di Carlo SE, Peduto L. The perivascular origin of pathological fibroblasts. J Clin Invest 2018; 128:54-63. [PMID: 29293094 DOI: 10.1172/jci93558] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The ability to repair tissues is essential for the survival of organisms. In chronic settings, the failure of the repair process to terminate results in overproduction of collagen, a pathology known as fibrosis, which compromises organ recovery and impairs function. The origin of the collagen-overproducing cell has been debated for years. Here we review recent insights gained from the use of lineage tracing approaches in several organs. The resulting evidence points toward specific subsets of tissue-resident mesenchymal cells, mainly localized in a perivascular position, as the major source for collagen-producing cells after injury. We discuss these findings in view of the functional heterogeneity of mesenchymal cells of the perivascular niche, which have essential vascular, immune, and regenerative functions that need to be preserved for efficient repair.
Collapse
|
43
|
Leukocyte-Stromal Interactions Within Lymph Nodes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:1-22. [PMID: 30155619 DOI: 10.1007/978-3-319-78127-3_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lymph nodes play a crucial role in the formation and initiation of immune responses, allowing lymphocytes to efficiently scan for foreign antigens and serving as rendezvous points for leukocyte-antigen interactions. Here we describe the major stromal subsets found in lymph nodes, including fibroblastic reticular cells, lymphatic endothelial cells, blood endothelial cells, marginal reticular cells, follicular dendritic cells and other poorly defined subsets such as integrin alpha-7+ pericytes. We focus on biomedically relevant interactions with T cells, B cells and dendritic cells, describing pro-survival mechanisms of support for these cells, promotion of their migration and tolerance-inducing mechanisms that help keep the body free of autoimmune-mediated damage.
Collapse
|
44
|
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: 165] [Impact Index Per Article: 20.6] [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
|
45
|
Lymphatic Endothelial Cells Control Initiation of Lymph Node Organogenesis. Immunity 2017; 47:80-92.e4. [DOI: 10.1016/j.immuni.2017.05.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 04/14/2017] [Accepted: 05/26/2017] [Indexed: 10/19/2022]
|
46
|
Agace WW, McCoy KD. Regionalized Development and Maintenance of the Intestinal Adaptive Immune Landscape. Immunity 2017; 46:532-548. [PMID: 28423335 DOI: 10.1016/j.immuni.2017.04.004] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/03/2017] [Accepted: 04/04/2017] [Indexed: 12/14/2022]
Abstract
The intestinal immune system has the daunting task of protecting us from pathogenic insults while limiting inflammatory responses against the resident commensal microbiota and providing tolerance to food antigens. This role is particularly impressive when one considers the vast mucosal surface and changing landscape that the intestinal immune system must monitor. In this review, we highlight regional differences in the development and composition of the adaptive immune landscape of the intestine and the impact of local intrinsic and environmental factors that shape this process. To conclude, we review the evidence for a critical window of opportunity for early-life exposures that affect immune development and alter disease susceptibility later in life.
Collapse
Affiliation(s)
- William W Agace
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark (DTU), 2800 Kongens Lyngby, Denmark; Immunology Section, Department of Experimental Medical Science, Lund University, BMC D14, Sölvegatan 19, 221 84 Lund, Sweden.
| | - Kathy D McCoy
- Department of Physiology and Pharmacology and Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
47
|
Irshad S, Flores-Borja F, Lawler K, Monypenny J, Evans R, Male V, Gordon P, Cheung A, Gazinska P, Noor F, Wong F, Grigoriadis A, Fruhwirth GO, Barber PR, Woodman N, Patel D, Rodriguez-Justo M, Owen J, Martin SG, Pinder SE, Gillett CE, Poland SP, Ameer-Beg S, McCaughan F, Carlin LM, Hasan U, Withers DR, Lane P, Vojnovic B, Quezada SA, Ellis P, Tutt ANJ, Ng T. RORγt + Innate Lymphoid Cells Promote Lymph Node Metastasis of Breast Cancers. Cancer Res 2017; 77:1083-1096. [PMID: 28082403 DOI: 10.1158/0008-5472.can-16-0598] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 12/09/2016] [Accepted: 12/10/2016] [Indexed: 11/16/2022]
Abstract
Cancer cells tend to metastasize first to tumor-draining lymph nodes, but the mechanisms mediating cancer cell invasion into the lymphatic vasculature remain little understood. Here, we show that in the human breast tumor microenvironment (TME), the presence of increased numbers of RORγt+ group 3 innate lymphoid cells (ILC3) correlates with an increased likelihood of lymph node metastasis. In a preclinical mouse model of breast cancer, CCL21-mediated recruitment of ILC3 to tumors stimulated the production of the CXCL13 by TME stromal cells, which in turn promoted ILC3-stromal interactions and production of the cancer cell motile factor RANKL. Depleting ILC3 or neutralizing CCL21, CXCL13, or RANKL was sufficient to decrease lymph node metastasis. Our findings establish a role for RORγt+ILC3 in promoting lymphatic metastasis by modulating the local chemokine milieu of cancer cells in the TME. Cancer Res; 77(5); 1083-96. ©2017 AACR.
Collapse
Affiliation(s)
- Sheeba Irshad
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
| | - Fabian Flores-Borja
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Katherine Lawler
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
- Institute for Mathematical and Molecular Biomedicine, King's College London, London, United Kingdom
| | - James Monypenny
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Rachel Evans
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Victoria Male
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
| | - Peter Gordon
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Anthony Cheung
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Patrycja Gazinska
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
| | - Farzana Noor
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
| | - Felix Wong
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Anita Grigoriadis
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
| | - Gilbert O Fruhwirth
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
- Leukocyte Dynamics Group, Beatson Advanced Imaging Resource, CRUK Beatson Institute, Glasgow, United Kingdom
| | - Paul R Barber
- Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, United Kingdom
| | - Natalie Woodman
- King's Health Partners Cancer Biobank, King's College London, London, United Kingdom
| | - Dominic Patel
- International Center for Infectiology Research, University of Lyon, Lyon, France
| | | | - Julie Owen
- King's Health Partners Cancer Biobank, King's College London, London, United Kingdom
| | - Stewart G Martin
- Division of Cancer and Stem Cells, Department of Clinical Oncology, School of Medicine, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Sarah E Pinder
- King's Health Partners Cancer Biobank, King's College London, London, United Kingdom
- Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, London, United Kingdom
| | - Cheryl E Gillett
- King's Health Partners Cancer Biobank, King's College London, London, United Kingdom
- Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, London, United Kingdom
| | - Simon P Poland
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Simon Ameer-Beg
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Frank McCaughan
- Department of Asthma, Allergy, and Lung Biology, King's College London, London, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Leo M Carlin
- Leukocyte Dynamics Group, Beatson Advanced Imaging Resource, CRUK Beatson Institute, Glasgow, United Kingdom
| | - Uzma Hasan
- International Center for Infectiology Research, University of Lyon, Lyon, France
| | - David R Withers
- MRC Centre for Immune Regulation, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Peter Lane
- MRC Centre for Immune Regulation, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Borivoj Vojnovic
- Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, United Kingdom
| | - Sergio A Quezada
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London, United Kingdom
| | - Paul Ellis
- Department of Medical Oncology, Guy's and St Thomas Foundation Trust, London, United Kingdom
| | - Andrew N J Tutt
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
- ICR, BCN Research Unit, Toby Robins Research Centre, London, United Kingdom
| | - Tony Ng
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom.
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London, United Kingdom
| |
Collapse
|
48
|
CD34+ mesenchymal cells are a major component of the intestinal stem cells niche at homeostasis and after injury. Proc Natl Acad Sci U S A 2017; 114:E506-E513. [PMID: 28074039 DOI: 10.1073/pnas.1620059114] [Citation(s) in RCA: 229] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The intestinal epithelium is continuously renewed by intestinal epithelial stem cells (IESCs) positioned at the base of each crypt. Mesenchymal-derived factors are essential to maintain IESCs; however, the cellular composition and development of such mesenchymal niche remains unclear. Here, we identify pericryptal CD34+ Gp38+ αSMA- mesenchymal cells closely associated with Lgr5+ IESCs. We demonstrate that CD34+ Gp38+ cells are the major intestinal producers of the niche factors Wnt2b, Gremlin1, and R-spondin1, and are sufficient to promote maintenance of Lgr5+ IESCs in intestinal organoids, an effect mainly mediated by Gremlin1. CD34+ Gp38+ cells develop after birth in the intestinal submucosa and expand around the crypts during the third week of life in mice, independently of the microbiota. We further show that pericryptal CD34+gp38+ cells are rapidly activated by intestinal injury, up-regulating niche factors Gremlin1 and R-spondin1 as well as chemokines, proinflammatory cytokines, and growth factors with key roles in gut immunity and tissue repair, including IL-7, Ccl2, Ptgs2, and Amphiregulin. Our results indicate that CD34+ Gp38+ mesenchymal cells are programmed to develop in the intestine after birth to constitute a specialized microenvironment that maintains IESCs at homeostasis and contribute to intestinal inflammation and repair after injury.
Collapse
|
49
|
Tan JKH, Watanabe T. Stromal Cell Subsets Directing Neonatal Spleen Regeneration. Sci Rep 2017; 7:40401. [PMID: 28067323 PMCID: PMC5220291 DOI: 10.1038/srep40401] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 12/06/2016] [Indexed: 01/08/2023] Open
Abstract
Development of lymphoid tissue is determined by interactions between stromal lymphoid tissue organiser (LTo) and hematopoietic lymphoid tissue inducer (LTi) cells. A failure for LTo to receive appropriate activating signals during embryogenesis through lymphotoxin engagement leads to a complete cessation of lymph node (LN) and Peyer's patch development, identifying LTo as a key stromal population for lymphoid tissue organogenesis. However, little is known about the equivalent stromal cells that induce spleen development. Here, by dissociating neonatal murine spleen stromal tissue for re-aggregation and transplant into adult mouse recipients, we have identified a MAdCAM-1+CD31+CD201+ spleen stromal organizer cell-type critical for new tissue formation. This finding provides an insight into the regulation of post-natal spleen tissue organogenesis, and could be exploited in the development of spleen regenerative therapies.
Collapse
Affiliation(s)
- Jonathan K H Tan
- AK Project, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan.,Division of Biomedical Science, Research School of Biology, The Australian National University, Canberra 0200, Australia
| | - Takeshi Watanabe
- AK Project, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
50
|
Abstract
Tissue mechanical disruption is often not sufficient to disrupt cell-to-cell interactions; this is particularly relevant for stromal cells that are embedded within the extracellular matrix. For this reason, different enzyme combinations have been described to enable the isolation of single-cell populations, particularly stromal cells. This chapter aims to describe different methods used for enzymatic digestion of stromal cell and leukocyte populations from secondary and tertiary lymphoid organs. Collagenase D and P and collagenase D and dispase protocols provide a good yield of stromal cells, while a collagenase dispase-only protocol should be used if the main aim of the technique is to retrieve leukocyte populations. However, for isolation of both stroma and leukocyte populations the collagenase D and P protocol would provide the best results. Protocols for these techniques and illustrative results from flow cytometry analysis can be found in this chapter.
Collapse
Affiliation(s)
- Saba Nayar
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
| | - Joana Campos
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
| | - Nathalie Steinthal
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK
| | - Francesca Barone
- Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, College of Medical & Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, B15 2WB, UK.
| |
Collapse
|