1
|
Liu H, Dai H, Qiu F, Chen Y, Liang CL, Yang B, Gong N, Bromberg JS, Dai Z. Electrostimulation suppresses allograft rejection via promoting lymphatic regulatory T cell migration mediated by lymphotoxin - lymphotoxin receptor β signaling. Am J Transplant 2024; 24:2187-2198. [PMID: 38992495 DOI: 10.1016/j.ajt.2024.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024]
Abstract
Conventional immunosuppressants that suppress allograft rejection cause various side effects. Although regulatory T cells (Tregs) are essential for allograft survival, the limited efficacy of Treg therapy demands improvement. Thus, it is imperative to seek new approaches to enhancing Treg suppression. Low-intensity electrostimulation (ES) has been shown to exert antiinflammatory effects without causing major adverse reactions. However, it remains unknown whether and how ES regulates alloimmunity. Here, we found that regional ES delayed murine skin allograft rejection and promoted long-term allograft survival induced by an mTOR inhibitor, rapamycin. ES also extended islet allograft survival. Mechanistically, ES enhanced the expression of lymphotoxin α (LTα) on Tregs after transplantation. Blockade of lymphotoxin β receptor-mediated nonclassical NFκB signaling suppressed lymphatic Treg migration and largely reversed the effects of ES on allograft survival. Moreover, ES failed to extend allograft survival when recipients lacked LTα/lymph nodes or if transferred Tregs lacked LTα. Therefore, ES promoted the lymphatic migration of CD4+Foxp3+ Tregs by upregulating their surface expression of LTα. Finally, ES augmented expression of LTα on murine or human Tregs, but not conventional T cells, while promoting their calcium influx in vitro. This ES-mediated upregulation of LTα relied on calcium influx. Thus, our findings have unveiled novel mechanisms underlying ES-mediated immunoregulation.
Collapse
Affiliation(s)
- Huazhen Liu
- Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Helong Dai
- Department of Kidney Transplantation, Center of Organ Transplantation, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Feifei Qiu
- Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yuchao Chen
- Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chun-Ling Liang
- Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Bin Yang
- Department of Cardiovascular Sciences, College of Life Sciences University of Leicester, Leicester, UK
| | - Nianqiao Gong
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation of Ministry of Education, National Health Commission and Chinese Academy of Medical Sciences, Wuhan, Hubei, China.
| | - Jonathan S Bromberg
- Kidney and Pancreas Transplantation, Department of Surgery and Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| | - Zhenhua Dai
- Section of Immunology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Pezoldt J, Wiechers C, Zou M, Litovchenko M, Biocanin M, Beckstette M, Sitnik K, Palatella M, van Mierlo G, Chen W, Gardeux V, Floess S, Ebel M, Russeil J, Arampatzi P, Vafardanejad E, Saliba AE, Deplancke B, Huehn J. Postnatal expansion of mesenteric lymph node stromal cells towards reticular and CD34 + stromal cell subsets. Nat Commun 2022; 13:7227. [PMID: 36433946 PMCID: PMC9700677 DOI: 10.1038/s41467-022-34868-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
Gut-draining mesenteric lymph nodes (LN) provide the framework to shape intestinal adaptive immune responses. Based on the transcriptional signatures established by our previous work, the composition and immunomodulatory function of LN stromal cells (SC) vary according to location. Here, we describe the single-cell composition and development of the SC compartment within mesenteric LNs derived from postnatal to aged mice. We identify CD34+ SC and fibroblastic reticular stromal cell (FRC) progenitors as putative progenitors, both supplying the typical rapid postnatal mesenteric LN expansion. We further establish the location-specific chromatin accessibility and DNA methylation landscape of non-endothelial SCs and identify a microbiota-independent core epigenomic signature, showing characteristic differences between SCs from mesenteric and skin-draining peripheral LNs. The epigenomic landscape of SCs points to dynamic expression of Irf3 along the differentiation trajectories of FRCs. Accordingly, a mesenchymal stem cell line acquires a Cxcl9+ FRC molecular phenotype upon lentiviral overexpression of Irf3, and the relevance of Irf3 for SC biology is further underscored by the diminished proportion of Ccl19+ and Cxcl9+ FRCs in LNs of Irf3-/- mice. Together, our data constitute a comprehensive transcriptional and epigenomic map of mesenteric LNSC development in early life and dissect location-specific, microbiota-independent properties of non-endothelial SCs.
Collapse
Affiliation(s)
- Joern Pezoldt
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany ,grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Carolin Wiechers
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Mangge Zou
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Maria Litovchenko
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Marjan Biocanin
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Michael Beckstette
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany ,grid.512472.7Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine, Helmholtz Centre for Infection Research and Hannover Medical School, 30625 Hannover, Germany ,grid.7491.b0000 0001 0944 9128Genome Informatics Group, Bielefeld Institute for Bioinformatics Infrastructure, Department of Technology, Bielefeld University, 33615 Bielefeld, Germany
| | - Katarzyna Sitnik
- grid.6583.80000 0000 9686 6466Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Martina Palatella
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Guido van Mierlo
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Wanze Chen
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Vincent Gardeux
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Stefan Floess
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Maria Ebel
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Julie Russeil
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Panagiota Arampatzi
- grid.8379.50000 0001 1958 8658Core Unit Systems Medicine, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Ehsan Vafardanejad
- grid.498164.6Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), 97080 Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- grid.498164.6Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), 97080 Würzburg, Germany
| | - Bart Deplancke
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jochen Huehn
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany ,grid.10423.340000 0000 9529 9877Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
Anatomical Uniqueness of the Mucosal Immune System (GALT, NALT, iBALT) for the Induction and Regulation of Mucosal Immunity and Tolerance. MUCOSAL VACCINES 2020. [PMCID: PMC7149644 DOI: 10.1016/b978-0-12-811924-2.00002-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
8
|
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
|
9
|
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]
|
10
|
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]
|
11
|
Receptor Activator of NF-κB Orchestrates Activation of Antiviral Memory CD8 T Cells in the Spleen Marginal Zone. Cell Rep 2018; 21:2515-2527. [PMID: 29186688 PMCID: PMC5723674 DOI: 10.1016/j.celrep.2017.10.111] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/30/2017] [Accepted: 10/28/2017] [Indexed: 12/23/2022] Open
Abstract
The spleen plays an important role in protective immunity to bloodborne pathogens. Macrophages and dendritic cells (DCs) in the spleen marginal zone capture microbial antigens to trigger adaptive immune responses. Marginal zone macrophages (MZMs) can also act as a replicative niche for intracellular pathogens, providing a platform for mounting the immune response. Here, we describe a role for RANK in the coordinated function of antigen-presenting cells in the spleen marginal zone and triggering anti-viral immunity. Targeted deletion of RANK results in the selective loss of CD169+ MZMs, which provide a niche for viral replication, while RANK signaling in DCs promotes the recruitment and activation of anti-viral memory CD8 T cells. These studies reveal a role for the RANKL/RANK signaling axis in the orchestration of protective immune responses in the spleen marginal zone that has important implications for the host response to viral infection and induction of acquired immunity.
Collapse
|
12
|
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
|
13
|
Marrero L, Simkin J, Sammarco M, Muneoka K. Fibroblast reticular cells engineer a blastema extracellular network during digit tip regeneration in mice. ACTA ACUST UNITED AC 2017; 4:69-84. [PMID: 28616246 PMCID: PMC5469731 DOI: 10.1002/reg2.75] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/27/2017] [Accepted: 02/24/2017] [Indexed: 12/11/2022]
Abstract
The regeneration blastema which forms following amputation of the mouse digit tip is composed of undifferentiated cells bound together by an organized network of fibers. A monoclonal antibody (ER‐TR7) that identifies extracellular matrix (ECM) fibers produced by fibroblast reticular cells during lymphoid organogenesis was used to characterize the ECM of the digit, the blastema, and the regenerate. Digit fibroblast reticular cells produce an ER‐TR7+ ECM network associated with different tissues and represent a subset of loose connective tissue fibroblasts. During blastema formation there is an upregulation of matrix production that returns to its pre‐existing level and anatomical pattern in the endpoint regenerate. Co‐localization studies demonstrate a strong spatial correlation between the ER‐TR7 antigen and collagen type III (COL3) in histological sections. ER‐TR7 and COL3 are co‐induced in cultured digit fibroblasts following treatment with tumor necrosis factor alpha and a lymphotoxin beta receptor agonist. These results provide an initial characterization of the ECM during digit regeneration and identify a subpopulation of fibroblasts involved in producing the blastema provisional matrix that is remodeled during the regeneration response.
Collapse
Affiliation(s)
- Luis Marrero
- Department of Cell and Molecular Biology Tulane University New Orleans LA 70118 USA.,Department of Medicine Louisiana State University Health Sciences Center New Orleans LA 70112 USA
| | - Jennifer Simkin
- Department of Cell and Molecular Biology Tulane University New Orleans LA 70118 USA
| | - Mimi Sammarco
- Department of Cell and Molecular Biology Tulane University New Orleans LA 70118 USA
| | - Ken Muneoka
- Department of Cell and Molecular Biology Tulane University New Orleans LA 70118 USA.,Department of Veterinary Physiology & Pharmacology Texas A&M University College Station TX7 7843 USA
| |
Collapse
|
14
|
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
|
15
|
Barone F, Gardner DH, Nayar S, Steinthal N, Buckley CD, Luther SA. Stromal Fibroblasts in Tertiary Lymphoid Structures: A Novel Target in Chronic Inflammation. Front Immunol 2016; 7:477. [PMID: 27877173 PMCID: PMC5100680 DOI: 10.3389/fimmu.2016.00477] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/20/2016] [Indexed: 12/14/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are organized aggregates of lymphocytes, myeloid, and stromal cells that provide ectopic hubs for acquired immune responses. TLS share phenotypical and functional features with secondary lymphoid organs (SLO); however, they require persistent inflammatory signals to arise and are often observed at target sites of autoimmune disease, chronic infection, cancer, and organ transplantation. Over the past 10 years, important progress has been made in our understanding of the role of stromal fibroblasts in SLO development, organization, and function. A complex and stereotyped series of events regulate fibroblast differentiation from embryonic life in SLOs to lymphoid organ architecture observed in adults. In contrast, TLS-associated fibroblasts differentiate from postnatal, locally activated mesenchyme, predominantly in settings of inflammation and persistent antigen presentation. Therefore, there are critical differences in the cellular and molecular requirements that regulate SLO versus TLS development that ultimately impact on stromal and hematopoietic cell function. These differences may contribute to the pathogenic nature of TLS in the context of chronic inflammation and malignant transformation and offer a window of opportunity for therapeutic interventions in TLS associated pathologies.
Collapse
Affiliation(s)
- Francesca Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - David H Gardner
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Nathalie Steinthal
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Christopher D Buckley
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Sanjiv A Luther
- Department of Biochemistry, Center for Immunity and Infection, University of Lausanne , Lausanne , Switzerland
| |
Collapse
|
16
|
Bessman NJ, Sonnenberg GF. Emerging roles for antigen presentation in establishing host-microbiome symbiosis. Immunol Rev 2016; 272:139-50. [PMID: 27319348 PMCID: PMC4916850 DOI: 10.1111/imr.12425] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Trillions of beneficial bacteria inhabit the intestinal tract of healthy mammals from birth. Accordingly, mammalian hosts have evolved a series of complementary and redundant pathways to limit pathologic immune responses against these bacteria, while simultaneously protecting against enteric pathogen invasion. These pathways can be generically responsive to the presence of any commensal bacteria and innate in nature, as for IL-22-related pathways. Alternatively, specific bacterial antigens can drive a distinct set of adaptive immune cell responses, including IgA affinity maturation and secretion, and a recently described pathway of intestinal selection whereby MHCII(+) ILC3 deletes commensal bacteria-reactive CD4 T cells. These pathways can either promote or inhibit colonization by specific subsets of commensal bacteria, and cooperatively maintain intestinal homeostasis. In this review, we will highlight recent developments in understanding how these diverse pathways complement each other to cooperatively shape the symbiotic relationship between commensal bacteria and mammalian hosts.
Collapse
Affiliation(s)
- Nicholas J Bessman
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
| | - Gregory F Sonnenberg
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
17
|
Walsh MC, Lee J, Choi Y. Tumor necrosis factor receptor- associated factor 6 (TRAF6) regulation of development, function, and homeostasis of the immune system. Immunol Rev 2016; 266:72-92. [PMID: 26085208 DOI: 10.1111/imr.12302] [Citation(s) in RCA: 312] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tumor necrosis factor receptor (TNFR)-associated factor 6 (TRAF6) is an adapter protein that mediates a wide array of protein-protein interactions via its TRAF domain and a RING finger domain that possesses non-conventional E3 ubiquitin ligase activity. First identified nearly two decades ago as a mediator of interleukin-1 receptor (IL-1R)-mediated activation of NFκB, TRAF6 has since been identified as an actor downstream of multiple receptor families with immunoregulatory functions, including members of the TNFR superfamily, the Toll-like receptor (TLR) family, tumor growth factor-β receptors (TGFβR), and T-cell receptor (TCR). In addition to NFκB, TRAF6 may also direct activation of mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K), and interferon regulatory factor pathways. In the context of the immune system, TRAF6-mediated signals have proven critical for the development, homeostasis, and/or activation of B cells, T cells, and myeloid cells, including macrophages, dendritic cells, and osteoclasts, as well as for organogenesis of thymic and secondary lymphoid tissues. In multiple cellular contexts, TRAF6 function is essential not only for proper activation of the immune system but also for maintaining immune tolerance, and more recent work has begun to identify mechanisms of contextual specificity for TRAF6, involving both regulatory protein interactions, and messenger RNA regulation by microRNAs.
Collapse
Affiliation(s)
- Matthew C Walsh
- Institute for Immunology and Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - JangEun Lee
- Institute for Immunology and Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yongwon Choi
- Institute for Immunology and Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
18
|
Context-Dependent Development of Lymphoid Stroma from Adult CD34+ Adventitial Progenitors. Cell Rep 2016; 14:2375-88. [DOI: 10.1016/j.celrep.2016.02.033] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/30/2015] [Accepted: 02/02/2016] [Indexed: 01/31/2023] Open
|
19
|
Hoorweg K, Narang P, Li Z, Thuery A, Papazian N, Withers DR, Coles MC, Cupedo T. A Stromal Cell Niche for Human and Mouse Type 3 Innate Lymphoid Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:4257-4263. [PMID: 26378073 DOI: 10.4049/jimmunol.1402584] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 08/22/2015] [Indexed: 11/19/2022]
Abstract
Adaptive immunity critically depends on the functional compartmentalization of secondary lymphoid organs. Mesenchymal stromal cells create and maintain specialized niches that support survival, activation, and expansion of T and B cells, and integrated analysis of lymphocytes and their niche has been instrumental in understanding adaptive immunity. Lymphoid organs are also home to type 3 innate lymphoid cells (ILC3), innate effector cells essential for barrier immunity. However, a specialized stromal niche for ILC3 has not been identified. A novel lineage-tracing approach now identifies a subset of murine fetal lymphoid tissue organizer cells that gives rise exclusively to adult marginal reticular cells. Moreover, both cell types are conserved from mice to humans and colocalize with ILC3 in secondary lymphoid tissues throughout life. In sum, we provide evidence that fetal stromal organizers give rise to adult marginal reticular cells and form a dedicated stromal niche for innate ILC3 in adaptive lymphoid organs.
Collapse
Affiliation(s)
- Kerim Hoorweg
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Priyanka Narang
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, UK
| | - Zhi Li
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, UK
| | - Anne Thuery
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, UK
| | - Natalie Papazian
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - David R Withers
- Medical Research Council Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, U.K
| | - Mark C Coles
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, UK
| | - Tom Cupedo
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
20
|
Buckley CD, Barone F, Nayar S, Bénézech C, Caamaño J. Stromal Cells in Chronic Inflammation and Tertiary Lymphoid Organ Formation. Annu Rev Immunol 2015; 33:715-45. [DOI: 10.1146/annurev-immunol-032713-120252] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Christopher D. Buckley
- Rheumatology Research Group, Center for Translational Inflammation Research, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham B15 2WD, United Kingdom
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| | - Francesca Barone
- Rheumatology Research Group, Center for Translational Inflammation Research, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham B15 2WD, United Kingdom
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| | - Saba Nayar
- Rheumatology Research Group, Center for Translational Inflammation Research, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham B15 2WD, United Kingdom
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| | - Cecile Bénézech
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| | - Jorge Caamaño
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| |
Collapse
|
21
|
Zhao L, Chen J, Liu L, Gao J, Guo B, Zhu B. Essential role of TNF-alpha in development of spleen fibroblastic reticular cells. Cell Immunol 2015; 293:130-6. [PMID: 25618740 DOI: 10.1016/j.cellimm.2015.01.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/09/2015] [Accepted: 01/10/2015] [Indexed: 01/05/2023]
Abstract
TNF-alpha plays an important role in the development of secondary lymphoid tissues. Earlier studies showed that fibroblastic reticular cells express TNF-alpha receptor, suggesting that TNF-alpha may affect the development of FRCs. To test this, we analyzed the development and function of FRCs in wild-type or TNF-alpha knockout mice. We found that GP38 expression was down-regulated in the spleen of TNF-alpha knockout mice. Chemokines, mainly secreted by GP38(+) FRCs, were also down-regulated. Additionally, we found that absence of TNF-alpha decreased the homing ability to direct T cells to the spleen. However, absence of TNF-alpha did not affect the development of lymph nodes FRCs. These data reveal that TNF-alpha plays an important role in the development of spleen FRCs. Absence of TNF-alpha could cause abnormality of spleen FRCs, thereby weakening the homing ability of T cells to localize to the spleen T cell zone.
Collapse
Affiliation(s)
- Lintao Zhao
- Institution of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| | - Junying Chen
- Institution of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| | - Lina Liu
- Institution of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| | - Jianbao Gao
- Institution of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| | - Bo Guo
- Institution of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing, China; Department of Immunology, Third Military Medical University, Chongqing, China.
| | - Bo Zhu
- Institution of Oncology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
22
|
Šedý J, Bekiaris V, Ware CF. Tumor necrosis factor superfamily in innate immunity and inflammation. Cold Spring Harb Perspect Biol 2014; 7:a016279. [PMID: 25524549 DOI: 10.1101/cshperspect.a016279] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The tumor necrosis factor superfamily (TNFSF) and its corresponding receptor superfamily (TNFRSF) form communication pathways required for developmental, homeostatic, and stimulus-responsive processes in vivo. Although this receptor-ligand system operates between many different cell types and organ systems, many of these proteins play specific roles in immune system function. The TNFSF and TNFRSF proteins lymphotoxins, LIGHT (homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for herpes virus entry mediator [HVEM], a receptor expressed by T lymphocytes), lymphotoxin-β receptor (LT-βR), and HVEM are used by embryonic and adult innate lymphocytes to promote the development and homeostasis of lymphoid organs. Lymphotoxin-expressing innate-acting B cells construct microenvironments in lymphoid organs that restrict pathogen spread and initiate interferon defenses. Recent results illustrate how the communication networks formed among these cytokines and the coreceptors B and T lymphocyte attenuator (BTLA) and CD160 both inhibit and activate innate lymphoid cells (ILCs), innate γδ T cells, and natural killer (NK) cells. Understanding the role of TNFSF/TNFRSF and interacting proteins in innate cells will likely reveal avenues for future therapeutics for human disease.
Collapse
Affiliation(s)
- John Šedý
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| | - Vasileios Bekiaris
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| | - Carl F Ware
- Laboratory of Molecular Immunology, Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, California 92037
| |
Collapse
|
23
|
TNFα-dependent development of lymphoid tissue in the absence of RORγt⁺ lymphoid tissue inducer cells. Mucosal Immunol 2014; 7:602-14. [PMID: 24129162 PMCID: PMC4264842 DOI: 10.1038/mi.2013.79] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 08/28/2013] [Accepted: 09/09/2013] [Indexed: 02/04/2023]
Abstract
Lymphoid tissue often forms within sites of chronic inflammation. Here we report that expression of the proinflammatory cytokine tumor necrosis factor α (TNFα) drives development of lymphoid tissue in the intestine. Formation of this ectopic lymphoid tissue was not dependent on the presence of canonical RORgt(+) lymphoid tissue-inducer (LTi) cells, because animals expressing increased levels of TNFα but lacking RORgt(+) LTi cells (TNF/Rorc(gt)(-/-) mice) developed lymphoid tissue in inflamed areas. Unexpectedly, such animals developed several lymph nodes (LNs) that were structurally and functionally similar to those of wild-type animals. TNFα production by F4/80(+) myeloid cells present within the anlagen was important for the activation of stromal cells during the late stages of embryogenesis and for the activation of an organogenic program that allowed the development of LNs. Our results show that lymphoid tissue organogenesis can occur in the absence of LTi cells and suggest that interactions between TNFα-expressing myeloid cells and stromal cells have an important role in secondary lymphoid organ formation.
Collapse
|
24
|
Regulation of the adaptive immune system by innate lymphoid cells. Curr Opin Immunol 2014; 27:75-82. [PMID: 24594491 DOI: 10.1016/j.coi.2014.01.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 01/16/2014] [Accepted: 01/23/2014] [Indexed: 12/11/2022]
Abstract
Innate lymphoid cells (ILCs) are a group of lymphocytes that promote rapid cytokine-dependent innate immunity, inflammation and tissue repair. In addition, a growing body of evidence suggests ILCs can influence adaptive immune cell responses. During fetal development a subset of ILCs orchestrate the generation and maturation of secondary lymphoid tissues. Following birth, ILCs continue to modulate adaptive immune cell responses indirectly through interactions with stromal cells in lymphoid tissues and epithelial cells at barrier surfaces. In this review we summarize the current understanding of how ILCs modulate the magnitude and quality of adaptive immune cell responses, and in particular focus on recent evidence suggesting that ILCs can also directly regulate CD4(+) T cells. Further, we discuss the implications that these pathways may have on human health and disease.
Collapse
|
25
|
Benezech C, Caamano JH. Generation of lymph node-fat pad chimeras for the study of lymph node stromal cell origin. J Vis Exp 2013:e50952. [PMID: 24378826 DOI: 10.3791/50952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The stroma is a key component of the lymph node structure and function. However, little is known about its origin, exact cellular composition and the mechanisms governing its formation. Lymph nodes are always encapsulated in adipose tissue and we recently demonstrated the importance of this relation for the formation of lymph node stroma. Adipocyte precursor cells migrate into the lymph node during its development and upon engagement of the Lymphotoxin-b receptor switch off adipogenesis and differentiate into lymphoid stromal cells (Bénézech et al.). Based on the lymphoid stroma potential of adipose tissue, we present a method using a lymph node/fat pad chimera that allows the lineage tracing of lymph node stromal cell precursors. We show how to isolate newborn lymph nodes and EYFP(+) embryonic adipose tissue and make a LN/ EYFP(+) fat pad chimera. After transfer under the kidney capsule of a host mouse, the lymph node incorporates local adipose tissue precursor cells and finishes its formation. Progeny analysis of EYFP(+) fat pad cells in the resulting lymph nodes can be performed by flow-cytometric analysis of enzymatically digested lymph nodes or by immunofluorescence analysis of lymph nodes cryosections. By using fat pads from different knockout mouse models, this method will provide an efficient way of analyzing the origin of the different lymph node stromal cell populations.
Collapse
Affiliation(s)
- Cecile Benezech
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham
| | | |
Collapse
|
26
|
Generation of tissue organoids by compaction reaggregation. Methods Mol Biol 2013. [PMID: 24318818 DOI: 10.1007/978-1-60327-292-6_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Cellular reaggregation methods are commonly used to generate tissue organoids for use in biological studies. Using a modified method termed "compaction reaggregation," it is possible to establish reaggregates of reproducible size from defined input cell numbers with ease and without specialist equipment. Importantly, this method is suitable for the study of tissues that have proved refractory to reaggregation by other methods. With the option of juxtaposing cell populations, this method is useful for studies of tissue organization and structure.
Collapse
|
27
|
Coles M, Veiga-Fernandes H. Insight into lymphoid tissue morphogenesis. Immunol Lett 2013; 156:46-53. [PMID: 23954810 DOI: 10.1016/j.imlet.2013.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 07/25/2013] [Accepted: 08/05/2013] [Indexed: 11/17/2022]
Abstract
Secondary lymphoid organs (SLO) are crucial structures for immune-surveillance and rapid immune responses allowing resident lymphocytes to encounter antigen-presenting cells that carry antigens from peripheral tissues. These structures develop during embryonic life through a tightly regulated process that involves interactions between haematopoietic and mesenchymal cells. Importantly, this morphogenesis potential is maintained throughout life since in chronic inflammatory conditions novel "tertiary lymphoid organs" can be generated by processes that are reminiscent of embryonic SLO development. In this review we will discuss early events in SLO morphogenesis, focusing on haematopoietic and mesenchymal cell subsets implicated on the development of lymphoid organs.
Collapse
Affiliation(s)
- Mark Coles
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York YO10 5DD, UK.
| | | |
Collapse
|
28
|
Wege AK, Huber B, Wimmer N, Männel DN, Hehlgans T. LTβR expression on hematopoietic cells regulates acute inflammation and influences maturation of myeloid subpopulations. Innate Immun 2013; 20:461-70. [PMID: 23940077 DOI: 10.1177/1753425913497242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 06/14/2013] [Indexed: 01/13/2023] Open
Abstract
Lymphotoxin beta-receptor (LTβR) is involved in the formation and maintenance of secondary lymphoid structures, as well as in the regulation of inflammatory responses. Because LTβR lymphoid structure formation continues to develop in infants, we compared two different chimera models: one using adult mice and the other using a transplantation model of neonatal mice. To elucidate the function of LTβR on lymphoid and non-lymphoid cells, we generated bone marrow chimeras on the wild type C57Bl/6 and the LTβR-deficient (LTβR(-/-)) background, and reconstituted the mice with bone marrow cells reciprocally. These chimeric mice were analyzed in the experimental model of acute dextran sulfate sodium-induced colitis. Interestingly, both models revealed not only equal reconstitution levels but also similar immunological responses: LTβR expression on stromal cells is essential for lymph node formation, whereas LTBR on hematopoietic cells is crucial for a decrease in inflammation. In addition, mice lacking LTβR on hematopoietic cells revealed (a) an increase of immature granulocytic cells in the spleen and (b) a reduced proportion of myeloid cells in peripheral blood and spleen expressing CD11b(+)Ly6C(+)Ly6G(-) (myeloid-derived suppressor cells expression profile). In conclusion, LTβR expression on hematopoietic cells seems to be involved in the down-regulation of acute inflammatory reactions paralleled by the appearance of immature myeloid cells.
Collapse
Affiliation(s)
- Anja K Wege
- Institute of Immunology, University of Regensburg, Regensburg, Germany Clinic of Gynecology and Obstetrics, Caritas Hospital St. Josef, University of Regensburg, Regensburg, Germany
| | - Barbara Huber
- Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Nadin Wimmer
- Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Daniela N Männel
- Institute of Immunology, University of Regensburg, Regensburg, Germany
| | - Thomas Hehlgans
- Institute of Immunology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
29
|
Lawton JC, Benson RA, Garside P, Brewer JM. Using lymph node transplantation as an approach to image cellular interactions between the skin and draining lymph nodes during parasitic infections. Parasitol Int 2013; 63:165-70. [PMID: 23892176 PMCID: PMC3863950 DOI: 10.1016/j.parint.2013.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 07/16/2013] [Indexed: 12/24/2022]
Abstract
The growing use of protozoan parasites expressing fluorescent reporter genes, together with advances in microscopy, is enabling visualisation of their behaviour and functions within the host from the very earliest stages of infection with previously unparalleled spatiotemporal resolution. These developments have begun to provide novel insights, which are informing our understanding of where host immune responses may be initiated, which cells are involved and the types of response that are elicited. Here we will review some of these recent observations that highlight the importance of cellular communication between the site of infection and the draining lymph node (dLN) in establishing infection and immunity. We also highlight a number of remaining challenges and unknowns that arise through our inability to follow and fate map the journey of a single cell between spatially separated tissue sites. In response to these challenges, we review a recently described experimental strategy that extends the spatial and temporal limits of previous imaging approaches, most significantly allowing longitudinal analysis of cellular migration between the skin and draining lymph nodes in vivo, without the requirement for invasive surgery. Role of skin and LN communication in establishing infection and immunity A technique to extend the spatial and temporal limits of tissue imaging This approach does not require invasive surgery for imaging. We can follow the journey of a single cell between spatially separated tissue sites. We outline future applications for this approach.
Collapse
Affiliation(s)
- Jennifer C Lawton
- Wellcome Trust Centre For Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | | | | | | |
Collapse
|
30
|
Chai Q, Onder L, Scandella E, Gil-Cruz C, Perez-Shibayama C, Cupovic J, Danuser R, Sparwasser T, Luther SA, Thiel V, Rülicke T, Stein JV, Hehlgans T, Ludewig B. Maturation of lymph node fibroblastic reticular cells from myofibroblastic precursors is critical for antiviral immunity. Immunity 2013; 38:1013-24. [PMID: 23623380 PMCID: PMC7111182 DOI: 10.1016/j.immuni.2013.03.012] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 03/29/2013] [Indexed: 01/11/2023]
Abstract
The stromal scaffold of the lymph node (LN) paracortex is built by fibroblastic reticular cells (FRCs). Conditional ablation of lymphotoxin-β receptor (LTβR) expression in LN FRCs and their mesenchymal progenitors in developing LNs revealed that LTβR-signaling in these cells was not essential for the formation of LNs. Although T cell zone reticular cells had lost podoplanin expression, they still formed a functional conduit system and showed enhanced expression of myofibroblastic markers. However, essential immune functions of FRCs, including homeostatic chemokine and interleukin-7 expression, were impaired. These changes in T cell zone reticular cell function were associated with increased susceptibility to viral infection. Thus, myofibroblasic FRC precursors are able to generate the basic T cell zone infrastructure, whereas LTβR-dependent maturation of FRCs guarantees full immunocompetence and hence optimal LN function during infection. Novel transgenic mouse model that targets FRCs in adult lymph nodes FRC-specific ablation of the LTβR did not abrogate LN development Myofibroblastic FRC precursors generate the basic infrastructure of the adult LN LTβR-mediated FRC maturation is critical for the maintenance of immunocompentence
Collapse
Affiliation(s)
- Qian Chai
- Institute of Immunobiology, Kantonal Hospital St. Gallen, 9007 St. Gallen, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Brendolan A, Caamaño JH. Mesenchymal cell differentiation during lymph node organogenesis. Front Immunol 2012; 3:381. [PMID: 23248630 PMCID: PMC3522075 DOI: 10.3389/fimmu.2012.00381] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 11/29/2012] [Indexed: 12/31/2022] Open
Abstract
Secondary lymphoid tissues such as lymph nodes are essential for the interactions between antigen presenting cells and lymphocytes that result in adaptive immune responses that protect the host against invading pathogens. The specialized architecture of these organs facilitates the cognate interactions between antigen-loaded dendritic cells and lymphocytes expressing their specific receptor as well as B-T cell interactions that are at the core of long lasting adaptive immune responses. Lymph nodes develop during embryogenesis as a result of a series of cross-talk interactions between a hematopoietically derived cell lineage called lymphoid tissue inducer cells and stromal cells of mesenchymal origin to form the anlagen of these organs. This review will present an overview of the different signaling pathways and maturation steps that mesenchymal cells undergo during the process of lymph node formation such as cell specification, priming, and maturation to become lymphoid tissue stromal organizer cells.
Collapse
Affiliation(s)
- Andrea Brendolan
- Division of Molecular Oncology, San Raffaele Scientific Institute Milan, Italy
| | | |
Collapse
|
32
|
Bénézech C, Mader E, Desanti G, Khan M, Nakamura K, White A, Ware CF, Anderson G, Caamaño JH. Lymphotoxin-β receptor signaling through NF-κB2-RelB pathway reprograms adipocyte precursors as lymph node stromal cells. Immunity 2012; 37:721-34. [PMID: 22940098 PMCID: PMC3809035 DOI: 10.1016/j.immuni.2012.06.010] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 06/20/2012] [Accepted: 06/30/2012] [Indexed: 01/08/2023]
Abstract
Lymph node development during embryogenesis involves lymphotoxin-β receptor engagement and subsequent differentiation of a poorly defined population of mesenchymal cells into lymphoid tissue organizer cells. Here, we showed that embryonic mesenchymal cells with characteristics of adipocyte precursors present in the microenvironment of lymph nodes gave rise to lymph node organizer cells. Signaling through the lymphotoxin-β receptor controlled the fate of adipocyte precursor cells by blocking adipogenesis and instead promoting lymphoid tissue stromal cell differentiation. This effect involved activation of the NF-κB2-RelB signaling pathway and inhibition of the expression of the key adipogenic factors Pparγ and Cebpα. In vivo organogenesis assays show that embryonic and adult adipocyte precursor cells can migrate into newborn lymph nodes and differentiate into a variety of lymph node stromal cells. Thus, we propose that adipose tissues act as a source of lymphoid stroma for lymph nodes and other lymphoid structures associated with fat.
Collapse
Affiliation(s)
- Cécile Bénézech
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Emma Mader
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Guillaume Desanti
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Mahmood Khan
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Kyoko Nakamura
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Andrea White
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Carl F. Ware
- Infectious and Inflammatory Disease Center, Sanford Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Graham Anderson
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Jorge H. Caamaño
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
33
|
Mueller CG, Hess E. Emerging Functions of RANKL in Lymphoid Tissues. Front Immunol 2012; 3:261. [PMID: 22969763 PMCID: PMC3432452 DOI: 10.3389/fimmu.2012.00261] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 08/01/2012] [Indexed: 12/21/2022] Open
Abstract
The tumor necrosis factor superfamily (TNFSF) members play pivotal roles in embryonic development of lymphoid tissue and their homeostasis. RANKL (Receptor activator of NF-κB ligand, also called TRANCE, TNFSF11) is recognized as an important player in bone homeostasis and lymphoid tissue formation. In its absence bone mass control is deregulated and lymph nodes fail to develop. While its function in bone is well described, there is still little functional insight into the action of RANKL in lymphoid tissue development and homeostasis. Here we provide an overview of the known functions of RANKL, its signaling receptor RANK and its decoy receptor OPG from the perspective of lymphoid tissue development and immune activation in the mouse. Expressed by the hematopoietic lymphoid tissue inducing (LTi) cells and the mesenchymal lymphoid tissue organizer (LTo) cells, RANKL was shown to stimulate Lymphotoxin (LT) expression and to be implicated in LTi cell accumulation. Our recent finding that RANKL also triggers proliferation of adult lymph node stroma suggests that RANKL may furthermore directly activate LTo cells. Beyond bone, the RANKL-RANK-OPG triad plays important roles in immunobiology that are waiting to be unraveled.
Collapse
Affiliation(s)
- Christopher G Mueller
- CNRS, Laboratory of Therapeutic Immunology and Chemistry, Institut de Biologie Moléculaire et Cellulaire, University of Strasbourg Strasbourg, France
| | | |
Collapse
|
34
|
Ferreira M, Domingues RG, Veiga-Fernandes H. Stroma cell priming in enteric lymphoid organ morphogenesis. Front Immunol 2012; 3:219. [PMID: 22837761 PMCID: PMC3402974 DOI: 10.3389/fimmu.2012.00219] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 07/07/2012] [Indexed: 12/18/2022] Open
Abstract
The lymphoid system is equipped with a network of specialized platforms located at strategic sites, which grant strict immune-surveillance and efficient immune responses. The development of these peripheral secondary lymphoid organs (SLO) occurs mainly in utero, while tertiary lymphoid structures can form in adulthood generally in response to persistent infection and inflammation. Regardless of the lymphoid tissue and intrinsic cellular and molecular differences, it is now well established that the recruitment of fully functional lymphoid tissue inducer (LTi) cells to presumptive lymphoid organ sites, and their consequent close and reciprocal interaction with resident stroma cells, are central to SLO formation. In contrast, the nature of events that initially prime resident sessile stroma cells to recruit and retain LTi cells remains poorly understood. Recently, new findings revealed early phases of SLO development putting emphasis on mesenchymal and lymphoid tissue initiator cells. Herein we discuss the main tenets of enteric lymphoid organs genesis and focus in the most recent findings that open new perspectives to the understanding of the early phases of lymphoid morphogenesis.
Collapse
Affiliation(s)
- Manuela Ferreira
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Lisboa, Portugal
| | | | | |
Collapse
|
35
|
Gibson VB, Benson RA, Bryson KJ, McInnes IB, Rush CM, Grassia G, Maffia P, Jenkinson EJ, White AJ, Anderson G, Brewer JM, Garside P. A novel method to allow noninvasive, longitudinal imaging of the murine immune system in vivo. Blood 2012; 119:2545-51. [PMID: 22271449 PMCID: PMC3398133 DOI: 10.1182/blood-2011-09-378356] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In vivo imaging has revolutionized understanding of the spatiotemporal complexity that subserves the generation of successful effector and regulatory immune responses. Until now, invasive surgery has been required for microscopic access to lymph nodes (LNs), making repeated imaging of the same animal impractical and potentially affecting lymphocyte behavior. To allow longitudinal in vivo imaging, we conceived the novel approach of transplanting LNs into the mouse ear pinna. Transplanted LNs maintain the structural and cellular organization of conventional secondary lymphoid organs. They participate in lymphocyte recirculation and exhibit the capacity to receive and respond to local antigenic challenge. The same LN could be repeatedly imaged through time without the requirement for surgical exposure, and the dynamic behavior of the cells within the transplanted LN could be characterized. Crucially, the use of blood vessels as fiducial markers also allowed precise re-registration of the same regions for longitudinal imaging. Thus, we provide the first demonstration of a method for repeated, noninvasive, in vivo imaging of lymphocyte behavior.
Collapse
Affiliation(s)
- Vivienne B. Gibson
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Robert A. Benson
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Karen J. Bryson
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Iain B. McInnes
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Catherine M. Rush
- Microbiology and Immunology, School of Veterinary and Biomedical Science, James Cook University, Townsville, Australia
| | - Gianluca Grassia
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Department of Experimental Pharmacology, University of Naples, Naples, Italy
| | - Pasquale Maffia
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
- Department of Experimental Pharmacology, University of Naples, Naples, Italy
| | - Eric J. Jenkinson
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, Medical School, University of Birmingham, Edgbaston, United Kingdom
| | - Andrea J. White
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, Medical School, University of Birmingham, Edgbaston, United Kingdom
| | - Graham Anderson
- Medical Research Council Centre for Immune Regulation, School of Immunity and Infection, Medical School, University of Birmingham, Edgbaston, United Kingdom
| | - James M. Brewer
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Paul Garside
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
36
|
Hess E, Duheron V, Decossas M, Lézot F, Berdal A, Chea S, Golub R, Bosisio MR, Bridal SL, Choi Y, Yagita H, Mueller CG. RANKL induces organized lymph node growth by stromal cell proliferation. THE JOURNAL OF IMMUNOLOGY 2011; 188:1245-54. [PMID: 22210913 DOI: 10.4049/jimmunol.1101513] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RANK and its ligand RANKL play important roles in the development and regulation of the immune system. We show that mice transgenic for Rank in hair follicles display massive postnatal growth of skin-draining lymph nodes. The proportions of hematopoietic and nonhematopoietic stromal cells and their organization are maintained, with the exception of an increase in B cell follicles. The hematopoietic cells are not activated and respond to immunization by foreign Ag and adjuvant. We demonstrate that soluble RANKL is overproduced from the transgenic hair follicles and that its neutralization normalizes lymph node size, inclusive area, and numbers of B cell follicles. Reticular fibroblastic and vascular stromal cells, important for secondary lymphoid organ formation and organization, express RANK and undergo hyperproliferation, which is abrogated by RANKL neutralization. In addition, they express higher levels of CXCL13 and CCL19 chemokines, as well as MAdCAM-1 and VCAM-1 cell-adhesion molecules. These findings highlight the importance of tissue-derived cues for secondary lymphoid organ homeostasis and identify RANKL as a key molecule for controlling the plasticity of the immune system.
Collapse
Affiliation(s)
- Estelle Hess
- Centre National de la Recherche Scientifique, Laboratoire d'Immunopathologie et Chimie Thérapeutique, Unité Propre de Recherche 9021, Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, 67084 Strasbourg, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Koning JJ, Mebius RE. Interdependence of stromal and immune cells for lymph node function. Trends Immunol 2011; 33:264-70. [PMID: 22153930 DOI: 10.1016/j.it.2011.10.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 09/15/2011] [Accepted: 10/31/2011] [Indexed: 01/09/2023]
Abstract
Lymph nodes are strategically located throughout the body to allow lymphocytes to efficiently encounter their cognate antigen and become activated. The structure of the lymph nodes is such that B and T lymphocytes each have their own microdomain. This structure is provided by lymph node stromal cells, which also provide the lymphocytes with a scaffold upon which to migrate. Here, we discuss how stromal cells differentiate from mesenchymal precursor cells in response to the interaction with lymphocytes, while these stromal cells in turn provide necessary survival factors for the lymphocytes. We propose that during immune reactions, the interactions of stromal and immune cells are similarly important for controlling the expanding lymphocyte pool.
Collapse
Affiliation(s)
- Jasper J Koning
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
38
|
Milićević NM, Klaperski K, Nohroudi K, Milićević Ž, Bieber K, Baraniec B, Blessenohl M, Kalies K, Ware CF, Westermann J. TNF receptor-1 is required for the formation of splenic compartments during adult, but not embryonic life. THE JOURNAL OF IMMUNOLOGY 2010; 186:1486-94. [PMID: 21187446 DOI: 10.4049/jimmunol.1000740] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lymphotoxin β-receptor (LTβR) and TNF receptor-1 (TNFR1) are important for the development of secondary lymphoid organs during embryonic life. The significance of LTβR and TNFR1 for the formation of lymphoid tissue during adult life is not well understood. Immunohistochemistry, morphometry, flow cytometry, and laser microdissection were used to compare wild-type, LTβR(-/-), TNFR1(-/-) spleens with splenic tissue that has been newly formed 8 wk after avascular implantation into adult mice. During ontogeny, LTβR is sufficient to induce formation of the marginal zone, similar-sized T and B cell zones, and a mixed T/B cell zone that completely surrounded the T cell zone. Strikingly, in adult mice, the formation of splenic compartments required both LTβR and TNFR1 expression, demonstrating that the molecular requirements for lymphoid tissue formation are different during embryonic and adult life. Thus, interfering with the TNFR1 pathway offers the possibility to selectively block the formation of ectopic lymphoid tissue and at the same time to spare secondary lymphoid organs such as spleen and lymph nodes. This opens a new perspective for the treatment of autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Novica M Milićević
- Institute of Histology and Embryology, Faculty of Medicine, University of Beograd, 11000 Beograd, Serbia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Secondary lymphoid organs are important locations for the initiation of adaptive immune responses. They develop before birth, and their formation requires interaction between lymphotoxin-α₁ß₂-expressing lymphoid-tissue inducer cells and lymphotoxin-ß receptor-expressing stromal organizer cells. Here, we discuss new insights into the earliest phases of peripheral lymph node and Peyer's patch formation that occur before lymphotoxin-ß receptor signalling and suggest a role for the developing nervous system. In addition, we discuss the differing requirements for the postnatal formation of mucosa-associated lymphoid tissues and tertiary lymphoid structures that develop at sites of chronic inflammation.
Collapse
|
40
|
Bénézech C, White A, Mader E, Serre K, Parnell S, Pfeffer K, Ware CF, Anderson G, Caamaño JH. Ontogeny of stromal organizer cells during lymph node development. THE JOURNAL OF IMMUNOLOGY 2010; 184:4521-30. [PMID: 20237296 DOI: 10.4049/jimmunol.0903113] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The development of secondary lymphoid organs, such as lymph nodes (LNs), in the embryo results from the reciprocal action between lymphoid tissue inducer (LTi) cells and stromal cells. However, the initial events inducing LN anlagen formation before the LTi stromal cells cross-talk interactions take place are not fully elucidated. In this study, we show that the inguinal LN anlagen in mouse embryos developed from mesenchymal cells surrounding the lymph sacs, spherical structures of endothelial cells that bud from veins. Using inguinal and mesenteric LNs (mLNs), we provide evidence supporting a two-step maturation model for stromal cells: first, ICAM-1(-)VCAM-1(-) mesenchymal precursor cells become ICAM-1(int)VCAM-1(int) cells, in a process independent of LTi cells and lymphotoxin beta receptor (LTbetaR) signaling. The second step involves the maturation of ICAM-1(int)VCAM-1(int) cells to ICAM-1(high)VCAM-1(high) mucosal addressin cell adhesion molecule-1(+) organizer cells and depends on both LTi cells and LTbetaR. Addition of alphaLTbetaR agonist to LN organ cultures was sufficient to induce ICAM-1(int)VCAM-1(int) cells to mature. In LtbetaR(-/-) embryos, both inguinal and mLN stromal cells showed a block at the ICAM-1(int)VCAM-1(int) stage, and, contrary to inguinal LNs, mLNs persist longer and contained LTi cells, which correlated with the sustained gene expression of Il-7, Cxcl13, and, to a lesser degree, Ccl21. Taken together, these results highlight the importance of the signals and cellular interactions that induce the maturation of stromal cells and ultimately lead to the formation of lymphoid tissues.
Collapse
Affiliation(s)
- Cécile Bénézech
- School of Immunity and Infection, Institute for BioMedical Research-Medical Research Council Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Chappaz S, Finke D. The IL-7 signaling pathway regulates lymph node development independent of peripheral lymphocytes. THE JOURNAL OF IMMUNOLOGY 2010; 184:3562-9. [PMID: 20207995 DOI: 10.4049/jimmunol.0901647] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lymph node (LN) organogenesis is initiated by the interaction between hematopoietic lymphoid tissue inducer (LTi) cells and the mesenchymal organizer cells. Mice in which the IL-7 signaling pathway has been disrupted have a severe defect in LN development; however, the reasons underlying this defect are as yet unknown. In this study, we show that the overexpression of thymic stromal lymphopoietin (TSLP) increased LTi cell numbers and restored LN development in IL-7(-/-) and RAG2(-/-) gamma(c)(-/-) mice. The TSLP-mediated LN restoration was strictly dependent on LTi cells and independent of lymphocyte colonization. Increased LTi cell numbers in the LN anlagen of RAG2(-/-) gamma(c)(-/-) TSLP transgenic mice were associated with the restoration of organizer cells, suggesting that LTi cell number is a critical parameter for LN organogenesis. Our results shed light on the minimal cellular requirement for LN development during ontogeny. We show that the presence of LTi and organizer cells, but not of peripheral lymphocytes, is critical for LN development and persistence and further suggest that the IL-7 signaling pathway regulates LN organogenesis by controlling the size of the LTi cell pool.
Collapse
Affiliation(s)
- Stéphane Chappaz
- Department of Biomedicine, University of Basel, Basel, Switzerland.
| | | |
Collapse
|
42
|
Coles M, Kioussis D, Veiga-Fernandes H. Cellular and Molecular Requirements in Lymph Node and Peyer's Patch Development. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 92:177-205. [DOI: 10.1016/s1877-1173(10)92008-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
43
|
|
44
|
Sheridan JM, Taoudi S, Medvinsky A, Blackburn CC. A novel method for the generation of reaggregated organotypic cultures that permits juxtaposition of defined cell populations. Genesis 2009; 47:346-51. [PMID: 19370754 DOI: 10.1002/dvg.20505] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cellular reaggregation methods have been used to generate in vitro organotypic cultures as a means to elucidate the cellular and molecular requirements of organogenesis. However, reproducibility from experiment to experiment has remained problematic and furthermore, current protocols do not support reaggregation of many important tissues. Here, using the thymus as a model organ, we present a novel reaggregation method termed "compaction reaggregation" that offers improved kinetics of reaggregation and greatly improved efficiency. Using compaction reaggregation we have been able to reaggregate the aorta-gonad- mesonephros region, a tissue that previously proved refractory to commonly used reaggregation methods, enabling the study of hematopoietic stem cell emergence and expansion. Additionally, compaction reaggregation permits the juxtaposition of different cell layers within the aggregated structure thus providing the means to study inductive interactions between different cell populations in vitro.
Collapse
Affiliation(s)
- Julie M Sheridan
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, University of Edinburgh, United Kingdom
| | | | | | | |
Collapse
|
45
|
Ruddle NH, Akirav EM. Secondary lymphoid organs: responding to genetic and environmental cues in ontogeny and the immune response. THE JOURNAL OF IMMUNOLOGY 2009; 183:2205-12. [PMID: 19661265 DOI: 10.4049/jimmunol.0804324] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Secondary lymphoid organs (SLOs) include lymph nodes, spleen, Peyer's patches, and mucosal tissues such as the nasal-associated lymphoid tissue, adenoids, and tonsils. Less discretely anatomically defined cellular accumulations include the bronchus-associated lymphoid tissue, cryptopatches, and isolated lymphoid follicles. All SLOs serve to generate immune responses and tolerance. SLO development depends on the precisely regulated expression of cooperating lymphoid chemokines and cytokines such as LTalpha, LTbeta, RANKL, TNF, IL-7, and perhaps IL-17. The relative importance of these factors varies between the individual lymphoid organs. Participating in the process are lymphoid tissue initiator, lymphoid tissue inducer, and lymphoid tissue organizer cells. These cells and others that produce crucial cytokines maintain SLOs in the adult. Similar signals regulate the transition from inflammation to ectopic or tertiary lymphoid tissues.
Collapse
Affiliation(s)
- Nancy H Ruddle
- Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, CT 06520--8089, USA
| | | |
Collapse
|
46
|
Vondenhoff MF, Greuter M, Goverse G, Elewaut D, Dewint P, Ware CF, Hoorweg K, Kraal G, Mebius RE. LTbetaR signaling induces cytokine expression and up-regulates lymphangiogenic factors in lymph node anlagen. THE JOURNAL OF IMMUNOLOGY 2009; 182:5439-45. [PMID: 19380791 DOI: 10.4049/jimmunol.0801165] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The formation of lymph nodes is a complex process crucially controlled through triggering of LTbetaR on mesenchymal cells by LTalpha(1)beta(2) expressing lymphoid tissue inducer (LTi) cells. This leads to the induction of chemokines to attract more hematopoietic cells and adhesion molecules to retain them. In this study, we show that the extravasation of the first hematopoietic cells at future lymph node locations occurs independently of LTalpha and that these cells, expressing TNF-related activation-induced cytokine (TRANCE), are the earliest LTi cells. By paracrine signaling the first expression of LTalpha(1)beta(2) is induced. Subsequent LTbetaR triggering on mesenchymal cells leads to their differentiation to stromal organizers, which now also start to express TRANCE, IL-7, as well as VEGF-C, in addition to the induced adhesion molecules and chemokines. Both TRANCE and IL-7 will further induce the expression of LTalpha(1)beta(2) on newly arrived immature LTi cells, resulting in more LTbetaR triggering, generating a positive feedback loop. Thus, LTbetaR triggering by LTi cells during lymph node development creates a local environment to which hematopoietic precursors are attracted and where they locally differentiate into fully mature, LTalpha(1)beta(2) expressing, LTi cells. Furthermore, the same signals may regulate lymphangiogenesis to the lymph node through induction of VEGF-C.
Collapse
Affiliation(s)
- Mark F Vondenhoff
- Department of Molecular Cell Biology and Immunology, VU (Vrije Universiteit) University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Vondenhoff MF, van de Pavert SA, Dillard ME, Greuter M, Goverse G, Oliver G, Mebius RE. Lymph sacs are not required for the initiation of lymph node formation. Development 2009; 136:29-34. [PMID: 19060331 DOI: 10.1242/dev.028456] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The lymphatic vasculature drains lymph fluid from the tissue spaces of most organs and returns it to the blood vasculature for recirculation. Before reaching the circulatory system, antigens and pathogens transported by the lymph are trapped by the lymph nodes. As proposed by Florence Sabin more than a century ago and recently validated, the mammalian lymphatic vasculature has a venous origin and is derived from primitive lymph sacs scattered along the embryonic body axis. Also as proposed by Sabin, it has been generally accepted that lymph nodes originate from those embryonic primitive lymph sacs. However, we now demonstrate that the initiation of lymph node development does not require lymph sacs. We show that lymph node formation is initiated normally in E14.5 Prox1-null mouse embryos devoid of lymph sacs and lymphatic vasculature, and in E17.5 Prox1 conditional mutant embryos, which have defective lymph sacs. However, subsequent clustering of hematopoietic cells within these developing lymph nodes is less efficient.
Collapse
Affiliation(s)
- Mark F Vondenhoff
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, PO box 7057, 1007 MB Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
48
|
Glanville SH, Bekiaris V, Jenkinson EJ, Lane PJL, Anderson G, Withers DR. Transplantation of embryonic spleen tissue reveals a role for adult non-lymphoid cells in initiating lymphoid tissue organization. Eur J Immunol 2009; 39:280-9. [PMID: 19089813 PMCID: PMC3149129 DOI: 10.1002/eji.200838724] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
In this report we describe a transplantation system where embryonic spleens are grafted into adult hosts. This model can be used to analyze the cellular and molecular requirements for the development and organization of splenic microenvironments. Whole embryonic day 15 (ED15) spleens, grafted under the kidney capsule of adult mice, were colonized by host-derived lymphocytes and DC and developed normal splenic architecture. Grafts were also able to form germinal centers in response to T-dependent antigen. Using this system we demonstrated that adult host-derived lymphotoxin (LT) α was sufficient for the development of ED15 LTα−/− grafts. Grafting of ED15 LTα−/− spleens into RAG−/− hosts followed by transfer of LT α−/− splenocytes revealed no requirement for lymphocyte-derived LT α in the induction of CCL21 or the development of T-zone stroma. These data suggest that interactions between adult lymphoid-tissue inducer-like cells and embryonic stromal cells initiated T-zone development. Furthermore, adult lymphoid tissue inducer-like cells were shown to develop from bone marrow-derived progenitors. The model described here demonstrates a method of transferring whole splenic microenvironments and dissecting the stromal and hematopoietic signals involved in spleen development and organization.
Collapse
Affiliation(s)
- Stephanie H Glanville
- MRC Centre for Immune Regulation, Institute for Biomedical Research, Birmingham Medical School, Birmingham, UK
| | | | | | | | | | | |
Collapse
|
49
|
Taoudi S, Gonneau C, Moore K, Sheridan JM, Blackburn CC, Taylor E, Medvinsky A. Extensive Hematopoietic Stem Cell Generation in the AGM Region via Maturation of VE-Cadherin+CD45+ Pre-Definitive HSCs. Cell Stem Cell 2008; 3:99-108. [DOI: 10.1016/j.stem.2008.06.004] [Citation(s) in RCA: 206] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Revised: 04/23/2008] [Accepted: 06/09/2008] [Indexed: 11/16/2022]
|
50
|
Abstract
In contrast to our understanding of murine lymphoid organogenesis, detailed knowledge on the mechanisms of human lymph node development is virtually lacking. This is mainly due to the obvious difficulties that accompany research using human fetal organs. In this review we will highlight current knowledge on human lymph node and Peyer's patch development and will temporally align observations made in humans with data available from murine studies. In the final paragraphs we will put this knowledge in the context of human malignancies in which interactions between lymphocytes and stroma, resembling those seen in lymphoid organs, are recapitulated.
Collapse
|