1
|
Krimpenfort LT, Degn SE, Heesters BA. The follicular dendritic cell: At the germinal center of autoimmunity? Cell Rep 2024; 43:113869. [PMID: 38431843 DOI: 10.1016/j.celrep.2024.113869] [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: 11/09/2023] [Revised: 01/09/2024] [Accepted: 02/10/2024] [Indexed: 03/05/2024] Open
Abstract
Autoimmune diseases strain healthcare systems worldwide as their incidence rises, and current treatments put patients at risk for infections. An increased understanding of autoimmune diseases is required to develop targeted therapies that do not impair normal immune function. Many autoimmune diseases present with autoantibodies, which drive local or systemic inflammation. This indicates the presence of autoreactive B cells that have escaped tolerance. An important step in the development of autoreactive B cells is the germinal center (GC) reaction, where they undergo affinity maturation toward cognate self-antigen. Follicular dendritic cells (FDCs) perform the essential task of antigen presentation to B cells during the affinity maturation process. However, in recent years, it has become clear that FDCs play a much more active role in regulation of GC processes. Here, we evaluate the biology of FDCs in the context of autoimmune disease, with the goal of informing future therapeutic strategies.
Collapse
Affiliation(s)
- Luc T Krimpenfort
- Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Søren E Degn
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Balthasar A Heesters
- Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
2
|
De Martin A, Stanossek Y, Pikor NB, Ludewig B. Protective fibroblastic niches in secondary lymphoid organs. J Exp Med 2024; 221:e20221220. [PMID: 38038708 PMCID: PMC10691961 DOI: 10.1084/jem.20221220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023] Open
Abstract
Fibroblastic reticular cells (FRCs) are specialized fibroblasts of secondary lymphoid organs that provide the structural foundation of the tissue. Moreover, FRCs guide immune cells to dedicated microenvironmental niches where they provide lymphocytes and myeloid cells with homeostatic growth and differentiation factors. Inflammatory processes, including infection with pathogens, induce rapid morphological and functional adaptations that are critical for the priming and regulation of protective immune responses. However, adverse FRC reprogramming can promote immunopathological tissue damage during infection and autoimmune conditions and subvert antitumor immune responses. Here, we review recent findings on molecular pathways that regulate FRC-immune cell crosstalk in specialized niches during the generation of protective immune responses in the course of pathogen encounters. In addition, we discuss how FRCs integrate immune cell-derived signals to ensure protective immunity during infection and how therapies for inflammatory diseases and cancer can be developed through improved understanding of FRC-immune cell interactions.
Collapse
Affiliation(s)
- Angelina De Martin
- Institute of Immunobiology, Medical Research Center, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Yves Stanossek
- Institute of Immunobiology, Medical Research Center, Kantonsspital St.Gallen, St.Gallen, Switzerland
- Department of Otorhinolaryngology, Head and Neck Surgery, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Natalia Barbara Pikor
- Institute of Immunobiology, Medical Research Center, Kantonsspital St.Gallen, St.Gallen, Switzerland
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St.Gallen, St.Gallen, Switzerland
| |
Collapse
|
3
|
Araie H, Hosono N, Tsujikawa T, Kiyono Y, Okazawa H, Yamauchi T. Hematopoiesis in the spleen after engraftment in unrelated cord blood transplantation evaluated by 18F-FLT PET imaging. Int J Hematol 2023; 118:618-626. [PMID: 37782417 PMCID: PMC10615934 DOI: 10.1007/s12185-023-03658-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/29/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023]
Abstract
Cord blood is an important donor source for allogeneic hematopoietic stem cell transplantation (allo-HSCT), with its unique composition and quality of hematopoietic cells. The proliferation site and potency of infused hematopoietic stem cells in humans may vary between stem cell sources. We investigated this possibility in a prospective, exploratory study to assess hematopoietic dynamics using the radiopharmaceutical 3'-deoxy-3'-18F-fluorothymidine (18F-FLT), a thymidine analog used in positron emission tomography imaging, before allo-HSCT and on days 50 and 180 after allo-HSCT. We evaluated 11 patients with hematological malignancies who underwent allo-HSCT [five with peripheral blood stem cell transplantation (PBSCT) and six with unrelated cord blood transplantation (UCBT)]. Before allo-HSCT, 18F-FLT uptake did not differ between the two groups. At day 50, 18F-FLT uptake in the spleen was significantly greater in the UCBT group than in the PBSCT group (p = 0.0043), with no difference in whole-body bone marrow. At day 180, the differences in spleen uptake had diminished, and there were no differences between groups in whole-body bone marrow or the spleen, except for the sternum. The persistence of splenic hematopoiesis after engraftment in the UCBT group may reflect the complex systemic homing and proliferation mechanisms of cord blood hematopoietic cells.
Collapse
Affiliation(s)
- Hiroaki Araie
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Naoko Hosono
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan.
| | - Tetsuya Tsujikawa
- Department of Radiology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yasushi Kiyono
- Biomedical Imaging Research Center, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hidehiko Okazawa
- Biomedical Imaging Research Center, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Takahiro Yamauchi
- Department of Hematology and Oncology, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka Shimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| |
Collapse
|
4
|
Vallecillo-García P, Orgeur M, Comai G, Poehle-Kronawitter S, Fischer C, Gloger M, Dumas CE, Giesecke-Thiel C, Sauer S, Tajbakhsh S, Höpken UE, Stricker S. A local subset of mesenchymal cells expressing the transcription factor Osr1 orchestrates lymph node initiation. Immunity 2023; 56:1204-1219.e8. [PMID: 37160119 DOI: 10.1016/j.immuni.2023.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 12/05/2022] [Accepted: 04/13/2023] [Indexed: 05/11/2023]
Abstract
During development, lymph node (LN) initiation is coordinated by lymphoid tissue organizer (LTo) cells that attract lymphoid tissue inducer (LTi) cells at strategic positions within the embryo. The identity and function of LTo cells during the initial attraction of LTi cells remain poorly understood. Using lineage tracing, we demonstrated that a subset of Osr1-expressing cells was mesenchymal LTo progenitors. By investigating the heterogeneity of Osr1+ cells, we uncovered distinct mesenchymal LTo signatures at diverse anatomical locations, identifying a common progenitor of mesenchymal LTos and LN-associated adipose tissue. Osr1 was essential for LN initiation, driving the commitment of mesenchymal LTo cells independent of neural retinoic acid, and for LN-associated lymphatic vasculature assembly. The combined action of chemokines CXCL13 and CCL21 was required for LN initiation. Our results redefine the role and identity of mesenchymal organizer cells and unify current views by proposing a model of cooperative cell function in LN initiation.
Collapse
Affiliation(s)
| | - Mickael Orgeur
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, 75015 Paris, France
| | - Glenda Comai
- Institut Pasteur, Stem Cells & Development Unit, CNRS UMR 3738, Paris, France
| | | | - Cornelius Fischer
- Core Facility Genomics, Berlin Institute of Health at Charité, 10178 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115, Berlin, Germany
| | - Marleen Gloger
- Max Delbrück Center for Molecular Medicine, Department of Translational Tumor Immunology, 13125 Berlin, Germany; Uppsala University, Immunology Genetics and Pathology, 75237 Uppsala, Sweden
| | - Camille E Dumas
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | | | - Sascha Sauer
- Core Facility Genomics, Berlin Institute of Health at Charité, 10178 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115, Berlin, Germany
| | - Shahragim Tajbakhsh
- Institut Pasteur, Stem Cells & Development Unit, CNRS UMR 3738, Paris, France
| | - Uta E Höpken
- Max Delbrück Center for Molecular Medicine, Department of Microenvironmental Regulation in Autoimmunity and Cancer, 13125 Berlin, Germany
| | - Sigmar Stricker
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany.
| |
Collapse
|
5
|
Pærregaard SI, Wulff L, Schussek S, Niss K, Mörbe U, Jendholm J, Wendland K, Andrusaite AT, Brulois KF, Nibbs RJB, Sitnik K, Mowat AM, Butcher EC, Brunak S, Agace WW. The small and large intestine contain related mesenchymal subsets that derive from embryonic Gli1 + precursors. Nat Commun 2023; 14:2307. [PMID: 37085516 PMCID: PMC10121680 DOI: 10.1038/s41467-023-37952-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 04/06/2023] [Indexed: 04/23/2023] Open
Abstract
The intestinal lamina propria contains a diverse network of fibroblasts that provide key support functions to cells within their local environment. Despite this, our understanding of the diversity, location and ontogeny of fibroblasts within and along the length of the intestine remains incomplete. Here we show that the small and large intestinal lamina propria contain similar fibroblast subsets that locate in specific anatomical niches. Nevertheless, we find that the transcriptional profile of similar fibroblast subsets differs markedly between the small intestine and colon suggesting region specific functions. We perform in vivo transplantation and lineage-tracing experiments to demonstrate that adult intestinal fibroblast subsets, smooth muscle cells and pericytes derive from Gli1-expressing precursors present in embryonic day 12.5 intestine. Trajectory analysis of single cell RNA-seq datasets of E12.5 and adult mesenchymal cells suggest that adult smooth muscle cells and fibroblasts derive from distinct embryonic intermediates and that adult fibroblast subsets develop in a linear trajectory from CD81+ fibroblasts. Finally, we provide evidence that colonic subepithelial PDGFRαhi fibroblasts comprise several functionally distinct populations that originate from an Fgfr2-expressing fibroblast intermediate. Our results provide insights into intestinal stromal cell diversity, location, function, and ontogeny, with implications for intestinal development and homeostasis.
Collapse
Affiliation(s)
- Simone Isling Pærregaard
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | - Line Wulff
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | - Sophie Schussek
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | - Kristoffer Niss
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Urs Mörbe
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | - Johan Jendholm
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | | | - Anna T Andrusaite
- Institute of Infection, immunity and Inflammation, University of Glasgow, Glasgow, Scotland, UK
| | - Kevin F Brulois
- Laboratory of Immunology and Vascular Biology, Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Robert J B Nibbs
- Institute of Infection, immunity and Inflammation, University of Glasgow, Glasgow, Scotland, UK
| | - Katarzyna Sitnik
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | - Allan McI Mowat
- Institute of Infection, immunity and Inflammation, University of Glasgow, Glasgow, Scotland, UK
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
- The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System and the Palo Alto Veterans Institute for Research (PAVIR), Palo Alto, CA, USA
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - William W Agace
- Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark.
- Immunology Section, Lund University, Lund, 221 84, Sweden.
| |
Collapse
|
6
|
O’Neill HC, Lim HK. Skeletal stem/progenitor cells provide the niche for extramedullary hematopoiesis in spleen. Front Physiol 2023; 14:1148414. [PMID: 37007998 PMCID: PMC10063897 DOI: 10.3389/fphys.2023.1148414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/10/2023] [Indexed: 03/19/2023] Open
Abstract
In bone marrow, the niche which supports hematopoiesis and nurtures hematopoietic stem cells (HSCs) contains perivascular reticular cells representing a subset of skeletal stem/progenitor cells (SSPCs). These stromal cells which provide the niche are lost or become inadequate during stress, disease or ageing, such that HSCs leave bone marrow and enter spleen and other peripheral sites to initiate extramedullary hematopoiesis and particularly myelopoiesis. Spleen also maintains niches for HSCs under steady-state conditions, evident since neonatal and adult spleen contain HSCs in low number and provide low-level hematopoiesis. In spleen, HSCs are found in the sinusoidal-rich red pulp region also in the vicinity of perivascular reticular cells. These cells resemble to some extent the known stromal elements reflecting HSC niches in bone marrow, and are investigated here for their characteristics as a subset of SSPCs. The isolation of spleen stromal subsets and the generation of cell lines which support HSCs and myelopoiesis in vitro has led to the identification of perivascular reticular cells which are unique to spleen. Analysis of gene and marker expression, as well as differentiative potential, identifies an osteoprogenitor cell type, reflective of one of several subsets of SSPCs described previously in bone, bone marrow and adipose tissue. The combined information supports a model for HSC niches in spleen involving perivascular reticular cells as SSPCs having osteogenic, stroma-forming capacity. These associate with sinusoids in red pulp to form niches for HSCs and to support the differentiation of hematopoietic progenitors during extramedullary hematopoiesis.
Collapse
|
7
|
Huang J, Deng R, Li W, Jiang M, Xiang AP, Zhang X. Nestin+ Mesenchymal Precursors Generate Distinct Spleen Stromal Cell Subsets and Have Immunomodulatory Function. Int J Mol Sci 2022; 23:ijms231911819. [PMID: 36233119 PMCID: PMC9569994 DOI: 10.3390/ijms231911819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are known to be widespread in many tissues and possess a broad spectrum of immunoregulatory properties. They have been used in the treatment of a variety of inflammatory diseases; however, the therapeutic effects are still inconsistent owing to their heterogeneity. Spleen stromal cells have evolved to regulate the immune response at many levels as they are bathed in a complex inflammatory milieu during infection. Therefore, it is unknown whether they have stronger immunomodulatory effects than their counterparts derived from other tissues. Here, using a transgenic mouse model expressing GFP driven by the Nestin (Nes) promoter, Nes-GFP+ cells from bone marrow and spleen were collected. Artificial lymphoid reconstruction in vivo was performed. Cell phenotype, inhibition of T cell inflammatory cytokines, and in vivo therapeutic effects were assessed. We observed Nes-GFP+ cells colocalized with splenic stromal cells and further demonstrated that these Nes-GFP+ cells had the ability to establish ectopic lymphoid-like structures in vivo. Moreover, we showed that the Nes-GFP+ cells possessed the characteristics of MSCs. Spleen-derived Nes-GFP+ cells exhibited greater immunomodulatory ability in vitro and more remarkable therapeutic efficacy in inflammatory diseases, especially inflammatory bowel disease (IBD) than bone marrow-derived Nes-GFP+ cells. Overall, our data showed that Nes-GFP+ cells contributed to subsets of spleen stromal populations and possessed the biological characteristics of MSCs with a stronger immunoregulatory function and therapeutic potential than bone marrow-derived Nes-GFP+ cells.
Collapse
Affiliation(s)
- Jing Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Ronghai Deng
- Department of Organ Transplantation, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Weiqiang Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Meihua Jiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510080, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
- Correspondence: ; Tel.: +86-20-87335982
| |
Collapse
|
8
|
Zhai Y, Zhao Y, Zhang Y, He J, Tang M, Liu Y, Yang G, Xue P, Yao Y, He M, Xu Y, Qu W, Zhang Y. Lead suppresses interferon γ to induce splenomegaly via modification on splenic endothelial cells and lymphoid tissue organizer cells in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 244:114046. [PMID: 36057201 DOI: 10.1016/j.ecoenv.2022.114046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/21/2022] [Accepted: 08/29/2022] [Indexed: 06/15/2023]
Abstract
Splenomegaly is a symptom characterized by the presence of an enlarged spleen. The impact of environmental factors on splenomegaly is largely unknown. In this study, C57BL/6 mice were treated with 125 ppm or 1250 ppm lead (Pb) via drinking water for 8 wk, and the process of splenomegaly was evaluated. Treatment with 1250 ppm Pb, but not 125 ppm Pb, caused splenomegaly, which was associated with increased capacity for erythrocyte clearance. Intriguingly, Pb-caused splenomegaly was independent of lymphoid tissue inducer (LTi) cells, which produce lymphotoxins α and β (LTα/β) to activate endothelial cells and LT organizer (LTo) cells and drive the development of spleen physiologically. A direct action of Pb on endothelial cells and LTo cells did not impact their proliferation. On the other hand, during steady state, a tonic level of interferon (IFN)γ acted on endothelial cells and LTo cells to suppress splenomegaly, as IFNγ receptor (IFNγR)-deficient mice had enlarged spleens relative to wild-type mice; during Pb exposure, splenic IFNγ production was suppressed, thus leading to a loss of the inhibitory effect of IFNγ on splenomegaly. Mechanically, Pb acted on splenic CD4+ T cells to suppress IFNγ production, which impaired the Janus kinase (Jak)1/ signal transducer and activator of transcription (STAT)1 signaling in endothelial cells and LTo cells; the weakened Jak1/STAT1 signaling resulted in the enhanced nuclear factor-κB (NF-κB) signaling in endothelial cells and LTo cells, which drove their proliferation and caused splenomegaly. The present study reveals a previously unrecognized mechanism for the immunotoxicity of Pb, which may extend our current understanding for Pb toxicology.
Collapse
Affiliation(s)
- Yue Zhai
- School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Yifan Zhao
- School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Yufan Zhang
- School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Jinyi He
- School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Mengke Tang
- School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Yalin Liu
- School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Guangrui Yang
- School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Peng Xue
- School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Ye Yao
- School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Miao He
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yanyi Xu
- School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Weidong Qu
- School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Yubin Zhang
- School of Public Health and Key Laboratory of Public Health Safety, Ministry of Education, Fudan University, Shanghai 200032, China.
| |
Collapse
|
9
|
Lenti E, Genovese L, Bianchessi S, Maurizio A, Sain SB, di Lillo A, Mattavelli G, Harel I, Bernassola F, Hehlgans T, Pfeffer K, Crosti M, Abrignani S, Evans SM, Sitia G, Guimarães-Camboa N, Russo V, van de Pavert SA, Garcia-Manteiga JM, Brendolan A. Fate mapping and scRNA sequencing reveal origin and diversity of lymph node stromal precursors. Immunity 2022; 55:606-622.e6. [PMID: 35358427 DOI: 10.1016/j.immuni.2022.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/30/2021] [Accepted: 03/03/2022] [Indexed: 11/25/2022]
Abstract
Lymph node (LN) stromal cells play a crucial role in LN development and in supporting adaptive immune responses. However, their origin, differentiation pathways, and transcriptional programs are still elusive. Here, we used lineage-tracing approaches and single-cell transcriptome analyses to determine origin, transcriptional profile, and composition of LN stromal and endothelial progenitors. Our results showed that all major stromal cell subsets and a large proportion of blood endothelial cells originate from embryonic Hoxb6+ progenitors of the lateral plate mesoderm (LPM), whereas lymphatic endothelial cells arise from Pax3+ progenitors of the paraxial mesoderm (PXM). Single-cell RNA sequencing revealed the existence of different Cd34+ and Cxcl13+ stromal cell subsets and showed that embryonic LNs contain proliferating progenitors possibly representing the amplifying populations for terminally differentiated cells. Taken together, our work identifies the earliest embryonic sources of LN stromal and endothelial cells and demonstrates that stromal diversity begins already during LN development.
Collapse
Affiliation(s)
- Elisa Lenti
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Genovese
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Bianchessi
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Aurora Maurizio
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Simona Baghai Sain
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessia di Lillo
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Greta Mattavelli
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Itamar Harel
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 9190401, Israel
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Thomas Hehlgans
- Leibniz Institute of Immunotherapy (LIT), Chair for Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Klaus Pfeffer
- Institute of Medical, Microbiology and Hospital Hygiene, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
| | - Mariacristina Crosti
- INGM, Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy
| | - Sergio Abrignani
- INGM, Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy; Department of Clinical Science and Community Health (DISCCO), University of Milan, Milan, Italy
| | - Sylvia M Evans
- Skaggs School of Pharmacy, University of California at San Diego, La Jolla, CA 92093, USA
| | - Giovanni Sitia
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nuno Guimarães-Camboa
- Institute of Cardiovascular Regeneration, Goethe-University, Frankfurt 60590, Germany; German Center for Cardiovascular Research, Berlin (partner site Frankfurt Rhine-Main), Germany
| | - Vincenzo Russo
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serge A van de Pavert
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, INSERM, CNRS, Marseille, France
| | | | - Andrea Brendolan
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
10
|
Alexandre YO, Schienstock D, Lee HJ, Gandolfo LC, Williams CG, Devi S, Pal B, Groom JR, Cao W, Christo SN, Gordon CL, Starkey G, D'Costa R, Mackay LK, Haque A, Ludewig B, Belz GT, Mueller SN. A diverse fibroblastic stromal cell landscape in the spleen directs tissue homeostasis and immunity. Sci Immunol 2022; 7:eabj0641. [PMID: 34995096 DOI: 10.1126/sciimmunol.abj0641] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yannick O Alexandre
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Dominik Schienstock
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Hyun Jae Lee
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Luke C Gandolfo
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.,Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia
| | - Cameron G Williams
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Sapna Devi
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Bhupinder Pal
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
| | - Joanna R Groom
- Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Wang Cao
- Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Susan N Christo
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Claire L Gordon
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.,Department of Infectious Diseases, Austin Health, Melbourne, VIC, Australia
| | - Graham Starkey
- Liver and Intestinal Transplant Unit, Austin Health, Melbourne, VIC, Australia.,Department of Surgery, University of Melbourne, Austin Health, Melbourne, VIC, Australia
| | - Rohit D'Costa
- DonateLife Victoria, Carlton, VIC, Australia.,Department of Intensive Care Medicine, Melbourne Health, Melbourne, VIC, Australia
| | - Laura K Mackay
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Ashraful Haque
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Gabrielle T Belz
- Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,University of Queensland Diamantina Institute, University of Queensland, Brisbane, QLD, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
11
|
El Shikh MEM, El Sayed R, Aly NAR, Prediletto E, Hands R, Fossati-Jimack L, Bombardieri M, Lewis MJ, Pitzalis C. Follicular dendritic cell differentiation is associated with distinct synovial pathotype signatures in rheumatoid arthritis. Front Med (Lausanne) 2022; 9:1013660. [PMID: 36465908 PMCID: PMC9709129 DOI: 10.3389/fmed.2022.1013660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
Follicular dendritic cells (FDCs) fundamentally contribute to the formation of synovial ectopic lymphoid-like structures in rheumatoid arthritis (RA) which is associated with poor clinical prognosis. Despite this critical role, regulation of FDC development in the RA synovium and its correlation with synovial pathotype differentiation remained largely unknown. Here, we demonstrate that CNA.42+ FDCs distinctively express the pericyte/fibroblast-associated markers PDGFR-β, NG2, and Thy-1 in the synovial perivascular space but not in established follicles. In addition, synovial RNA-Seq analysis revealed that expression of the perivascular FDC markers was strongly correlated with PDGF-BB and fibroid synovitis, whereas TNF-α/LT-β was significantly associated with lymphoid synovitis and expression of CR1, CR2, and FcγRIIB characteristic of mature FDCs in lymphoid follicles. Moreover, PDGF-BB induced CNA.42+ FDC differentiation and CXCL13 secretion from NG2+ synovial pericytes, and together with TNF-α/LT-β conversely regulated early and late FDC differentiation genes in unsorted RA synovial fibroblasts (RASF) and this was confirmed in flow sorted stromal cell subsets. Furthermore, RASF TNF-αR expression was upregulated by TNF-α/LT-β and PDGF-BB; and TNF-α/LT-β-activated RASF retained ICs and induced B cell activation in in vitro germinal center reactions typical of FDCs. Additionally, FDCs trapped peptidyl citrulline, and strongly correlated with IL-6 expression, and plasma cell, B cell, and T cell infiltration of the RA synovium. Moreover, synovial FDCs were significantly associated with RA disease activity and radiographic features of tissue damage. To the best of our knowledge, this is the first report describing the reciprocal interaction between PDGF-BB and TNF-α/LT-β in synovial FDC development and evolution of RA histological pathotypes. Selective targeting of this interplay could inhibit FDC differentiation and potentially ameliorate RA in clinically severe and drug-resistant patients.
Collapse
|
12
|
Abd El-Aleem SA, Saber EA, Aziz NM, El-Sherif H, Abdelraof AM, Djouhri L. Follicular dendritic cells. J Cell Physiol 2021; 237:2019-2033. [PMID: 34918359 DOI: 10.1002/jcp.30662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 11/08/2022]
Abstract
Follicular dendritic cells (FDCs) are unique accessory immune cells that contribute to the regulation of humoral immunity. They are multitasker cells essential for the organization and maintenance of the lymphoid architecture, induction of germinal center reaction, production of B memory cells, and protection from autoimmune disorders. They perform their activities through both antigen-driven and chemical signaling to B cells. FDCs play a crucial role in the physiological regulation of the immune response. Dis-regulation of this immune response results when FDCs retain antigens for years. This provides a constant antigenic stimulation for B cells resulting in the development of immune disorders. Antigen trapped on FDCs is resistant to therapeutic intervention causing chronicity and recurrences. Beyond their physiological immunoregulatory functions, FDCs are involved in the pathogenesis of several immune-related disorders including HIV/AIDS, prion diseases, chronic inflammatory, and autoimmune disorders. FDCs have also been recently implicated in rare neoplasms of lymphoid and hematopoietic tissues. Understanding FDC biology is essential for better control of humoral immunity and opens the gate for therapeutic management of FDC-mediated immune disorders. Thus, the biology of FDCs has become a hot research area in the last couple of decades. In this review, we aim to provide a comprehensive overview of FDCs and their role in physiological and pathological conditions.
Collapse
Affiliation(s)
| | - Entesar Ali Saber
- Department of Histology and Cell Biology, Minia University, Minya, Egypt.,Department of Pharmacy, Deraya University, New Minia City, Egypt
| | - Neven M Aziz
- Department of Pharmacy, Deraya University, New Minia City, Egypt.,Department of Physiology, Minia Faculty of Medicine, Minia, Egypt
| | - Hani El-Sherif
- Department of Pharmacy, Deraya University, New Minia City, Egypt
| | - Asmaa M Abdelraof
- Public Health, Community, Environmental and Occupational Department, Faculty of Medicine, Beni-Suef University, Beni Suef, Egypt
| | - Laiche Djouhri
- Department of Physiology, College of Medicine (QU Health), Qatar University, Doha, Qatar
| |
Collapse
|
13
|
Koning JJ, Rajaraman A, Reijmers RM, Konijn T, Pan J, Ware CF, Butcher EC, Mebius RE. Development of follicular dendritic cells in lymph nodes depends on retinoic acid-mediated signaling. Development 2021; 148:dev199713. [PMID: 34528674 PMCID: PMC8572003 DOI: 10.1242/dev.199713] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 09/09/2021] [Indexed: 11/20/2022]
Abstract
Specialized stromal cells occupy and help define B- and T-cell domains, which are crucial for proper functioning of our immune system. Signaling through lymphotoxin and TNF receptors is crucial for the development of different stromal subsets, which are thought to arise from a common precursor. However, mechanisms that control the selective generation of the different stromal phenotypes are not known. Using in vitro cultures of embryonic mouse stromal cells, we show that retinoic acid-mediated signaling is important for the differentiation of precursors towards the Cxcl13pos follicular dendritic cell (FDC) lineage, and also blocks lymphotoxin-mediated Ccl19pos fibroblastic reticular cell lineage differentiation. Accordingly, at the day of birth we observe the presence of Cxcl13posCcl19neg/low and Cxcl13neg/lowCcl19pos cells within neonatal lymph nodes. Furthermore, ablation of retinoic acid receptor signaling in stromal precursors early after birth reduces Cxcl13 expression, and complete blockade of retinoic acid signaling prevents the formation of FDC networks in lymph nodes.
Collapse
Affiliation(s)
- Jasper J. Koning
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Infection and Immunity Institute, 1081HZ Amsterdam, the Netherlands
| | - Anusha Rajaraman
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA 94304, USA
| | - Rogier M. Reijmers
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Infection and Immunity Institute, 1081HZ Amsterdam, the Netherlands
| | - Tanja Konijn
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Infection and Immunity Institute, 1081HZ Amsterdam, the Netherlands
| | - Junliang Pan
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA 94304, USA
| | - Carl F. Ware
- Infectious and Inflammatory Diseases Research Center, Laboratory of Molecular Immunology, Sanford Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Eugene C. Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA 94304, USA
- The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Reina E. Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Infection and Immunity Institute, 1081HZ Amsterdam, the Netherlands
| |
Collapse
|
14
|
Correa-Gallegos D, Jiang D, Rinkevich Y. Fibroblasts as confederates of the immune system. Immunol Rev 2021; 302:147-162. [PMID: 34036608 DOI: 10.1111/imr.12972] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 12/18/2022]
Abstract
Fibroblastic stromal cells are as diverse, in origin and function, as the niches they fashion in the mammalian body. This cellular variety impacts the spectrum of responses elicited by the immune system. Fibroblast influence on the immune system keeps evolving our perspective on fibroblast roles and functions beyond just a passive structural part of organs. This review discusses the foundations of fibroblastic stromal-immune crosstalk, under the scope of stromal heterogeneity as a basis for tissue-specific tutoring of the immune system. Focusing on the skin as a relevant immunological organ, we detail the complex interactions between distinct fibroblast populations and immune cells that occur during homeostasis, injury repair, scarring, and disease. We further review the relevance of fibroblastic stromal cell heterogeneity and how this heterogeneity is central to regulate the immune system from its inception during embryonic development into adulthood.
Collapse
Affiliation(s)
- Donovan Correa-Gallegos
- Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München, Munich, Germany
| | - Dongsheng Jiang
- Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München, Munich, Germany
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
15
|
Bellomo A, Gentek R, Golub R, Bajénoff M. Macrophage-fibroblast circuits in the spleen. Immunol Rev 2021; 302:104-125. [PMID: 34028841 DOI: 10.1111/imr.12979] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/30/2021] [Accepted: 04/30/2021] [Indexed: 12/22/2022]
Abstract
Macrophages are an integral part of all organs in the body, where they contribute to immune surveillance, protection, and tissue-specific homeostatic functions. This is facilitated by so-called niches composed of macrophages and their surrounding stroma. These niches structurally anchor macrophages and provide them with survival factors and tissue-specific signals that imprint their functional identity. In turn, macrophages ensure appropriate functioning of the niches they reside in. Macrophages thus form reciprocal, mutually beneficial circuits with their cellular niches. In this review, we explore how this concept applies to the spleen, a large secondary lymphoid organ whose primary functions are to filter the blood and regulate immunity. We first outline the splenic micro-anatomy, the different populations of splenic fibroblasts and macrophages and their respective contribution to protection of and key physiological processes occurring in the spleen. We then discuss firmly established and potential cellular circuits formed by splenic macrophages and fibroblasts, with an emphasis on the molecular cues underlying their crosstalk and their relevance to splenic functionality. Lastly, we conclude by considering how these macrophage-fibroblast circuits might be impaired by aging, and how understanding these changes might help identify novel therapeutic avenues with the potential of restoring splenic functions in the elderly.
Collapse
Affiliation(s)
- Alicia Bellomo
- CIRI, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Univ Lyon, Lyon, France
| | - Rebecca Gentek
- Centre for Inflammation Research & Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Rachel Golub
- Inserm U1223, Institut Pasteur, Paris, France.,Lymphopoiesis Unit, Institut Pasteur, Paris, France
| | - Marc Bajénoff
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| |
Collapse
|
16
|
Kwang D, Tjin G, Purton LE. Regulation of murine B lymphopoiesis by stromal cells. Immunol Rev 2021; 302:47-67. [PMID: 34002391 DOI: 10.1111/imr.12973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/21/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022]
Abstract
B lymphocytes are crucial for the body's humoral immune response, secreting antibodies generated against foreign antigens to fight infection. Adult murine B lymphopoiesis is initiated in the bone marrow and additional maturation occurs in the spleen. In both these organs, B lymphopoiesis involves interactions with numerous different non-hematopoietic cells, also known as stromal or microenvironment cells, which provide migratory, maturation, and survival signals. A variety of conditional knockout and transgenic mouse models have been used to identify the roles of distinct microenvironment cell types in the regulation of B lymphopoiesis. These studies have revealed that mesenchymal lineage cells and endothelial cells comprise the non-hematopoietic microenvironment cell types that support B lymphopoiesis in the bone marrow. In the spleen, various types of stromal cells and endothelial cells contribute to B lymphocyte maturation. More recently, comprehensive single cell RNA-seq studies have also been used to identify clusters of stromal cell types in the bone marrow and spleen, which will aid in further identifying key regulators of B lymphopoiesis. Here, we review the different types of microenvironment cells and key extrinsic regulators that are known to be involved in the regulation of murine B lymphopoiesis in the bone marrow and spleen.
Collapse
Affiliation(s)
- Diannita Kwang
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Vic., Australia
| | - Gavin Tjin
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Vic., Australia
| | - Louise E Purton
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Vic., Australia.,Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, Vic., Australia
| |
Collapse
|
17
|
Wu H, Liu P, Xie XR, Chi JS, Li H, Xu CX. Inflammatory pseudotumor-like follicular dendritic cell sarcoma: Literature review of 67 cases. World J Meta-Anal 2021; 9:1-11. [DOI: 10.13105/wjma.v9.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/02/2021] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammatory pseudotumor (IPT)-like follicular dendritic cell (FDC) sarcoma is rare. The 2017 World Health Organization classification of tumors of hematopoietic and lymphoid tissues noted that data on its clinical outcome are limited, but that the tumor appears to be indolent. The aim of this study was to summarize the clinical characteristics, treatment outcomes, and prognostic factors for IPT-like FDC sarcoma. A literature review was conducted on retrospective analyses of clinical data and prognostic information on IPT-like FDC sarcoma reported between 2001 and 2020. A total of 67 cases of IPT-like FDC sarcoma were retrieved from the literature, documenting that it occurs predominantly in middle-aged adults, with a marked female predilection. Six patients had a separate malignancy and five had an autoimmune disease. Typically involving the spleen and/or liver, it may also selectively involve the abdomen, gastrointes-tinal tract, pancreas, retroperitoneum, and mesentery. Necrosis, hemorrhage, noncaseating epithelioid granulomas, and fibrinoid deposits in blood vessel walls are often present. The neoplastic cells are predominantly positive for follicular dendritic cell markers such as cluster of differentiation 21 (CD21), CD23, CD35 and CNA.42 and are consistently Epstein-Barr virus (EBV)-positive. Mitoses were very rare in most cases. Most patients were treated by surgery alone. Disease status at the time of last follow-up was known for 57 patients with follow-up time ranging from 2 to 144 mo. Local and/or distant recurrence after initial treatment was seen in 15.8% of the patients. The 1- and 5-year progression-free survival for the entire group was 91.5% and 56.1%, respectively. Kaplan-Meier and multivariate analyses showed that age, sex, tumor size, and pathological features were not risk factors for disease progression. IPT-like FDC sarcoma appears to be mildly aggressive and requires annual surveillance. Surgery is the most effective treatment modality, and the role of adjuvant chemotherapy for postoperative management is unclear. EBV is likely to play an important role in the etiology of IPT-like FDC sarcoma.
Collapse
Affiliation(s)
- Hao Wu
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Peng Liu
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Xiao-Ran Xie
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Jing-Shu Chi
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Huan Li
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Can-Xia Xu
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| |
Collapse
|
18
|
Cao J, O'Day DR, Pliner HA, Kingsley PD, Deng M, Daza RM, Zager MA, Aldinger KA, Blecher-Gonen R, Zhang F, Spielmann M, Palis J, Doherty D, Steemers FJ, Glass IA, Trapnell C, Shendure J. A human cell atlas of fetal gene expression. Science 2020; 370:370/6518/eaba7721. [PMID: 33184181 DOI: 10.1126/science.aba7721] [Citation(s) in RCA: 360] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022]
Abstract
The gene expression program underlying the specification of human cell types is of fundamental interest. We generated human cell atlases of gene expression and chromatin accessibility in fetal tissues. For gene expression, we applied three-level combinatorial indexing to >110 samples representing 15 organs, ultimately profiling ~4 million single cells. We leveraged the literature and other atlases to identify and annotate hundreds of cell types and subtypes, both within and across tissues. Our analyses focused on organ-specific specializations of broadly distributed cell types (such as blood, endothelial, and epithelial), sites of fetal erythropoiesis (which notably included the adrenal gland), and integration with mouse developmental atlases (such as conserved specification of blood cells). These data represent a rich resource for the exploration of in vivo human gene expression in diverse tissues and cell types.
Collapse
Affiliation(s)
- Junyue Cao
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Diana R O'Day
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Hannah A Pliner
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Paul D Kingsley
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Mei Deng
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Riza M Daza
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Michael A Zager
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Center for Data Visualization, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Kimberly A Aldinger
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Ronnie Blecher-Gonen
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Malte Spielmann
- Human Molecular Genomics Group, Max Planck Institute for Molecular Genetics, Berlin, Germany.,Institute of Human Genetics, University of Lübeck, Lübeck, Germany
| | - James Palis
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Dan Doherty
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA.,Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Ian A Glass
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA.,Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Cole Trapnell
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA. .,Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA. .,Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.,Howard Hughes Medical Institute, Seattle, WA, USA
| |
Collapse
|
19
|
Cozzitorto C, Mueller L, Ruzittu S, Mah N, Willnow D, Darrigrand JF, Wilson H, Khosravinia D, Mahmoud AA, Risolino M, Selleri L, Spagnoli FM. A Specialized Niche in the Pancreatic Microenvironment Promotes Endocrine Differentiation. Dev Cell 2020; 55:150-162.e6. [PMID: 32857951 PMCID: PMC7720791 DOI: 10.1016/j.devcel.2020.08.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/11/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022]
Abstract
The interplay between pancreatic epithelium and the surrounding microenvironment is pivotal for pancreas formation and differentiation as well as adult organ homeostasis. The mesenchyme is the main component of the embryonic pancreatic microenvironment, yet its cellular identity is broadly defined, and whether it comprises functionally distinct cell subsets is not known. Using genetic lineage tracing, transcriptome, and functional studies, we identified mesenchymal populations with different roles during pancreatic development. Moreover, we showed that Pbx transcription factors act within the mouse pancreatic mesenchyme to define a pro-endocrine specialized niche. Pbx directs differentiation of endocrine progenitors into insulin- and glucagon-positive cells through non-cell-autonomous regulation of ECM-integrin interactions and soluble molecules. Next, we measured functional conservation between mouse and human pancreatic mesenchyme by testing identified mesenchymal factors in an iPSC-based differentiation model. Our findings provide insights into how lineage-specific crosstalk between epithelium and neighboring mesenchymal cells underpin the generation of different pancreatic cell types.
Collapse
Affiliation(s)
- Corinna Cozzitorto
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Department of Ophthalmology & Department of Anatomy, Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Laura Mueller
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Silvia Ruzittu
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Nancy Mah
- Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - David Willnow
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Jean-Francois Darrigrand
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Heather Wilson
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Daniel Khosravinia
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Amir-Ala Mahmoud
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Department of Orofacial Sciences & Department of Anatomy, University of California, San Francisco, 513 Parnassus Ave, HSW 710, San Francisco, CA 94143, USA
| | - Maurizio Risolino
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Department of Orofacial Sciences & Department of Anatomy, University of California, San Francisco, 513 Parnassus Ave, HSW 710, San Francisco, CA 94143, USA
| | - Licia Selleri
- Program in Craniofacial Biology, Institute for Human Genetics, Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, Department of Orofacial Sciences & Department of Anatomy, University of California, San Francisco, 513 Parnassus Ave, HSW 710, San Francisco, CA 94143, USA
| | - Francesca M Spagnoli
- Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin 13125, Germany; Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK.
| |
Collapse
|
20
|
Bellomo A, Mondor I, Spinelli L, Lagueyrie M, Stewart BJ, Brouilly N, Malissen B, Clatworthy MR, Bajénoff M. Reticular Fibroblasts Expressing the Transcription Factor WT1 Define a Stromal Niche that Maintains and Replenishes Splenic Red Pulp Macrophages. Immunity 2020; 53:127-142.e7. [PMID: 32562599 DOI: 10.1016/j.immuni.2020.06.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/20/2020] [Accepted: 06/04/2020] [Indexed: 12/21/2022]
Abstract
Located within red pulp cords, splenic red pulp macrophages (RPMs) are constantly exposed to the blood flow, clearing senescent red blood cells (RBCs) and recycling iron from hemoglobin. Here, we studied the mechanisms underlying RPM homeostasis, focusing on the involvement of stromal cells as these cells perform anchoring and nurturing macrophage niche functions in lymph nodes and liver. Microscopy revealed that RPMs are embedded in a reticular meshwork of red pulp fibroblasts characterized by the expression of the transcription factor Wilms' Tumor 1 (WT1) and colony stimulating factor 1 (CSF1). Conditional deletion of Csf1 in WT1+ red pulp fibroblasts, but not white pulp fibroblasts, drastically altered the RPM network without altering circulating CSF1 levels. Upon RPM depletion, red pulp fibroblasts transiently produced the monocyte chemoattractants CCL2 and CCL7, thereby contributing to the replenishment of the RPM network. Thus, red pulp fibroblasts anchor and nurture RPM, a function likely conserved in humans.
Collapse
Affiliation(s)
- Alicia Bellomo
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | | | | | | - Benjamin J Stewart
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Nicolas Brouilly
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Institut de Biologie du Développement de Marseille, Marseille, France
| | | | - Menna R Clatworthy
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Marc Bajénoff
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.
| |
Collapse
|
21
|
Benabid A, Peduto L. Mesenchymal perivascular cells in immunity and disease. Curr Opin Immunol 2020; 64:50-55. [PMID: 32387900 PMCID: PMC7597593 DOI: 10.1016/j.coi.2020.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/12/2020] [Accepted: 03/14/2020] [Indexed: 12/13/2022]
Abstract
The mesenchymal microenvironment is increasingly recognized as a major player in immunity. Here we focus on mesenchymal cells located within or in proximity to the blood vessels wall, which include pericytes, adventitial fibroblasts and mesenchymal stromal cells. We discuss recent evidence that these cells play a role in tissue homeostasis, immunity and inflammatory pathologies by multiple mechanisms, including vascular modulation, leucocyte migration, activation or survival in the perivascular space and differentiation into specialized 'effector' mesenchymal cells essential for tissue repair and immunity, such as myofibroblasts and lymphoid stromal cells. When dysregulated, these responses contribute to inflammatory and fibrotic diseases.
Collapse
Affiliation(s)
- Adam Benabid
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Inserm U1224, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Lucie Peduto
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Inserm U1224, Paris, France.
| |
Collapse
|
22
|
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: 184] [Impact Index Per Article: 46.0] [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
|
23
|
Knop L, Deiser K, Bank U, Witte A, Mohr J, Philipsen L, Fehling HJ, Müller AJ, Kalinke U, Schüler T. IL-7 derived from lymph node fibroblastic reticular cells is dispensable for naive T cell homeostasis but crucial for central memory T cell survival. Eur J Immunol 2020; 50:846-857. [PMID: 32043573 DOI: 10.1002/eji.201948368] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/23/2020] [Accepted: 02/07/2020] [Indexed: 01/20/2023]
Abstract
The survival of peripheral T cells is dependent on their access to peripheral LNs (pLNs) and stimulation by IL-7. In pLNs fibroblastic reticular cells (FRCs) and lymphatic endothelial cells (LECs) produce IL-7 suggesting their contribution to the IL-7-dependent survival of T cells. However, IL-7 production is detectable in multiple organs and is not restricted to pLNs. This raises the question whether pLN-derived IL-7 is required for the maintenance of peripheral T cell homeostasis. Here, we show that numbers of naive T cells (TN ) remain unaffected in pLNs and spleen of mice lacking Il7 gene activity in pLN FRCs, LECs, or both. In contrast, frequencies of central memory T cells (TCM ) are reduced in FRC-specific IL-7 KO mice. Thus, steady state IL-7 production by pLN FRCs is critical for the maintenance of TCM , but not TN , indicating that both T cell subsets colonize different ecological niches in vivo.
Collapse
Affiliation(s)
- Laura Knop
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Katrin Deiser
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Ute Bank
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Amelie Witte
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Juliane Mohr
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Lars Philipsen
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Hans J Fehling
- Institute of Immunology, University Clinics Ulm, Ulm, Germany
| | - Andreas J Müller
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.,Intravital Microscopy in Infection and Immunity, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ulrich Kalinke
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Medical School Hannover, Institute for Experimental Infection Research, Hannover, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
24
|
Thierry GR, Gentek R, Bajenoff M. Remodeling of reactive lymph nodes: Dynamics of stromal cells and underlying chemokine signaling. Immunol Rev 2020; 289:42-61. [PMID: 30977194 DOI: 10.1111/imr.12750] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 01/28/2019] [Accepted: 01/31/2019] [Indexed: 12/19/2022]
Abstract
Lymph nodes (LNs) are secondary immune organs dispersed throughout the body. They are primarily composed of lymphocytes, "transient passengers" that are only present for a few hours. During this time, they extensively interact with a meshwork of stromal cells. Although these cells constitute less than 5% of all LN cells, they are integral to LN function: Stromal cells create a three-dimensional network that provides a rigid backbone for the transport of lymph and generates "roads" for lymphocyte migration. Beyond structural support, the LN stroma also produces survival signals for lymphocytes and provides nutrients, soluble factors, antigens, and immune cells collectively required for immune surveillance and the generation of adaptive immune responses. A unique feature of LNs is their ability to considerably and rapidly change size: the volume and cellularity of inflamed LNs can increase up to 20-fold before returning to homeostatic levels. This cycle will be repeated many times during life and is accommodated by stromal cells. The dynamics underlying this dramatic remodeling are subject of this review. We will first introduce the main types of LN stromal cells and explain their known functions. We will then discuss how these cells enable LN growth during immune responses, with a particular focus on underlying cellular mechanisms and molecular cues. Similarly, we will elaborate on stromal dynamics mediating the return to LN homeostasis, a process that is mechanistically much less understood than LN expansion.
Collapse
Affiliation(s)
- Guilhem R Thierry
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, Marseille, France
| | - Rebecca Gentek
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, Marseille, France
| | - Marc Bajenoff
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, Marseille, France
| |
Collapse
|
25
|
Transcription factor Tlx1 marks a subset of lymphoid tissue organizer-like mesenchymal progenitor cells in the neonatal spleen. Sci Rep 2019; 9:20408. [PMID: 31892733 PMCID: PMC6938487 DOI: 10.1038/s41598-019-56984-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022] Open
Abstract
The spleen is comprised of spatially distinct compartments whose functions, such as immune responses and removal of aged red blood cells, are tightly controlled by the non-hematopoietic stromal cells that provide regionally-restricted signals to properly activate hematopoietic cells residing in each area. However, information regarding the ontogeny and relationships of the different stromal cell types remains limited. Here we have used in vivo lineage tracing analysis and in vitro mesenchymal stromal cell assays and found that Tlx1, a transcription factor essential for embryonic spleen organogenesis, marks neonatal stromal cells that are selectively localized in the spleen and retain mesenchymal progenitor potential to differentiate into mature follicular dendritic cells, fibroblastic reticular cells and marginal reticular cells. Furthermore, by establishing a novel three-dimensional cell culture system that enables maintenance of Tlx1-expressing cells in vitro, we discovered that signals from the lymphotoxin β receptor and TNF receptor promote differentiation of these cells to express MAdCAM-1, CCL19 and CXCL13, representative functional molecules expressed by different subsets of mature stromal cells in the spleen. Taken together, these findings indicate that mesenchymal progenitor cells expressing Tlx1 are a subset of lymphoid tissue organizer-like cells selectively found in the neonatal spleen.
Collapse
|
26
|
O'Neill HC, Lim HK, Periasamy P, Kumarappan L, Tan JKH, O'Neill TJ. Transplanted spleen stromal cells with osteogenic potential support ectopic myelopoiesis. PLoS One 2019; 14:e0223416. [PMID: 31584977 PMCID: PMC6777786 DOI: 10.1371/journal.pone.0223416] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 09/20/2019] [Indexed: 12/24/2022] Open
Abstract
Spleen stromal lines which support in vitro hematopoiesis are investigated for their lineage origin and hematopoietic support function in vivo. Marker expression and gene profiling identify a lineage relationship with mesenchymal stem cells and perivascular reticular cells described recently in bone marrow. Stromal lines commonly express Cxcl12, Pdgfra and Pdgfr typical of bone marrow derived perivascular reticular cells but reflect a unique cell type in terms of other gene and marker expression. Their classification as osteoprogenitors is confirmed through ability to undergo osteogenic, but not adipogenic or chondrogenic differentiation. Some stromal lines were shown to form ectopic niches for HSCs following engraftment under the kidney capsule of NOD/SCID mice. The presence of myeloid cells and a higher representation of a specific dendritic-like cell type over other myeloid cells within grafts was consistent with previous in vitro evidence of hematopoietic support capacity. These studies reinforce the role of perivascular/perisinusoidal reticular cells in hematopoiesis and implicate such cells as niches for hematopoiesis in spleen.
Collapse
Affiliation(s)
- Helen C O'Neill
- Clem Jones Research Centre for Regenerative Medicine, Bond University, Gold Coast, Queensland, Australia
| | - Hong K Lim
- Clem Jones Research Centre for Regenerative Medicine, Bond University, Gold Coast, Queensland, Australia
| | - Pravin Periasamy
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory, Australia.,Department of Microbiology, Yoo Long School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lavanya Kumarappan
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Jonathan K H Tan
- Clem Jones Research Centre for Regenerative Medicine, Bond University, Gold Coast, Queensland, Australia
| | - Terence J O'Neill
- Big Data Centre, Bond Business School, Bond University, Gold Coast, Queensland, Australia
| |
Collapse
|
27
|
Denton AE, Carr EJ, Magiera LP, Watts AJB, Fearon DT. Embryonic FAP + lymphoid tissue organizer cells generate the reticular network of adult lymph nodes. J Exp Med 2019; 216:2242-2252. [PMID: 31324739 PMCID: PMC6780995 DOI: 10.1084/jem.20181705] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 05/17/2019] [Accepted: 06/27/2019] [Indexed: 01/03/2023] Open
Abstract
The induction of adaptive immunity is dependent on the structural organization of LNs, which is in turn governed by the stromal cells that underpin LN architecture. Using a novel fate-mapping mouse model, we trace the developmental origin of mesenchymal LN stromal cells (mLNSCs) to a previously undescribed embryonic fibroblast activation protein-α (FAP)+ progenitor. FAP+ cells of the LN anlagen express lymphotoxin β receptor (LTβR) and vascular cell adhesion molecule (VCAM), but not intercellular adhesion molecule (ICAM), suggesting they are early mesenchymal lymphoid tissue organizer (mLTo) cells. Clonal labeling shows that FAP+ progenitors locally differentiate into mLNSCs. This process is also coopted in nonlymphoid tissues in response to infection to facilitate the development of tertiary lymphoid structures, thereby mimicking the process of LN ontogeny in response to infection.
Collapse
Affiliation(s)
- Alice E Denton
- Lymphocyte Signaling and Development, Babraham Institute, Cambridge, UK .,Department of Medicine, University of Cambridge, Cambridge, UK.,Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Edward J Carr
- Lymphocyte Signaling and Development, Babraham Institute, Cambridge, UK.,Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lukasz P Magiera
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Andrew J B Watts
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Douglas T Fearon
- Department of Medicine, University of Cambridge, Cambridge, UK.,Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.,Weill Cornell Medicine and Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| |
Collapse
|
28
|
Lenti E, Bianchessi S, Proulx ST, Palano MT, Genovese L, Raccosta L, Spinelli A, Drago D, Andolfo A, Alfano M, Petrova TV, Mukenge S, Russo V, Brendolan A. Therapeutic Regeneration of Lymphatic and Immune Cell Functions upon Lympho-organoid Transplantation. Stem Cell Reports 2019; 12:1260-1268. [PMID: 31155505 PMCID: PMC6565831 DOI: 10.1016/j.stemcr.2019.04.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 12/14/2022] Open
Abstract
Lymph nodes (LNs) are secondary lymphoid tissues that play a critical role in filtering the lymph and promoting adaptive immune responses. Surgical resection of LNs, radiation therapy, or infections may damage lymphatic vasculature and compromise immune functions. Here, we describe the generation of functional synthetic lympho-organoids (LOs) using LN stromal progenitors and decellularized extracellular matrix-based scaffolds, two basic constituents of secondary lymphoid tissues. We show that upon transplantation at the site of resected LNs, LOs become integrated into the endogenous lymphatic vasculature and efficiently restore lymphatic drainage and perfusion. Upon immunization, LOs support the activation of antigen-specific immune responses, thus acquiring properties of native lymphoid tissues. These findings provide a proof-of-concept strategy for the development of functional lympho-organoids suitable for restoring lymphatic and immune cell functions.
Collapse
Affiliation(s)
- Elisa Lenti
- Unit of Lymphoid Organ Development, Division of Experimental Oncology, DIBIT-1 3A2, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Silvia Bianchessi
- Unit of Lymphoid Organ Development, Division of Experimental Oncology, DIBIT-1 3A2, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Steven T Proulx
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, 8093 Zurich, Switzerland
| | - Maria Teresa Palano
- Unit of Lymphoid Organ Development, Division of Experimental Oncology, DIBIT-1 3A2, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Luca Genovese
- Unit of Lymphoid Organ Development, Division of Experimental Oncology, DIBIT-1 3A2, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Laura Raccosta
- Unit of Immuno-Biotherapy of Melanoma and Solid Tumors, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Antonello Spinelli
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Denise Drago
- ProMiFa, Protein Microsequencing Facility, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Annapaola Andolfo
- ProMiFa, Protein Microsequencing Facility, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne, and Ludwig Institute for Cancer Research, 1066 Lausanne, Switzerland
| | - Sylvain Mukenge
- Department of Hepatobiliary Surgery, IRCCS San Raffaele Hospital, 20132 Milan, Italy
| | - Vincenzo Russo
- Unit of Immuno-Biotherapy of Melanoma and Solid Tumors, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Andrea Brendolan
- Unit of Lymphoid Organ Development, Division of Experimental Oncology, DIBIT-1 3A2, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy.
| |
Collapse
|
29
|
Origin and differentiation trajectories of fibroblastic reticular cells in the splenic white pulp. Nat Commun 2019; 10:1739. [PMID: 30988302 PMCID: PMC6465367 DOI: 10.1038/s41467-019-09728-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 03/27/2019] [Indexed: 12/12/2022] Open
Abstract
The splenic white pulp is underpinned by poorly characterized stromal cells that demarcate distinct immune cell microenvironments. Here we establish fibroblastic reticular cell (FRC)-specific fate-mapping in mice to define their embryonic origin and differentiation trajectories. Our data show that all reticular cell subsets descend from multipotent progenitors emerging at embryonic day 19.5 from periarterial progenitors. Commitment of FRC progenitors is concluded during the first week of postnatal life through occupation of niches along developing central arterioles. Single cell transcriptomic analysis facilitated deconvolution of FRC differentiation trajectories and indicated that perivascular reticular cells function both as adult lymphoid organizer cells and mural cell progenitors. The lymphotoxin-β receptor-independent sustenance of postnatal progenitor stemness unveils that systemic immune surveillance in the splenic white pulp is governed through subset specification of reticular cells from a multipotent periarterial progenitor cell. In sum, the finding that discrete signaling events in perivascular niches determine the differentiation trajectories of reticular cell networks explains the development of distinct microenvironmental niches in secondary and tertiary lymphoid tissues that are crucial for the induction and regulation of innate and adaptive immune processes. The white pulp of spleen is an important immune structure dynamically modulated during development and immune responses. Here the authors define, using multi-color lineage tracing and single-cell transcriptome analysis, the subset distribution and differentiation trajectory of fibroblastic reticular cells to serve structural insights for splenic white pulps.
Collapse
|
30
|
Short C, Lim HK, Tan J, O'Neill HC. Targeting the Spleen as an Alternative Site for Hematopoiesis. Bioessays 2019; 41:e1800234. [PMID: 30970171 DOI: 10.1002/bies.201800234] [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: 11/22/2018] [Revised: 02/25/2019] [Indexed: 12/21/2022]
Abstract
Bone marrow is the main site for hematopoiesis in adults. It acts as a niche for hematopoietic stem cells (HSCs) and contains non-hematopoietic cells that contribute to stem cell dormancy, quiescence, self-renewal, and differentiation. HSC also exist in resting spleen of several species, although their contribution to hematopoiesis under steady-state conditions is unknown. The spleen can however undergo extramedullary hematopoiesis (EMH) triggered by physiological stress or disease. With the loss of bone marrow niches in aging and disease, the spleen as an alternative tissue site for hematopoiesis is an important consideration for future therapy, particularly during HSC transplantation. In terms of harnessing the spleen as a site for hematopoiesis, here the remarkable regenerative capacity of the spleen is considered with a view to forming additional or ectopic spleen tissue through cell engraftment. Studies in mice indicate the potential for such grafts to support the influx of hematopoietic cells leading to the development of normal spleen architecture. An important goal will be the formation of functional ectopic spleen tissue as an aid to hematopoietic recovery following clinical treatments that impact bone marrow. For example, expansion or replacement of niches could be considered where myeloablation ahead of HSC transplantation compromises treatment outcomes.
Collapse
Affiliation(s)
- Christie Short
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, 4229, Australia
| | - Hong K Lim
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, 4229, Australia
| | - Jonathan Tan
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, 4229, Australia
| | - Helen C O'Neill
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, 4229, Australia
| |
Collapse
|
31
|
Srivastava S, Salter AI, Liggitt D, Yechan-Gunja S, Sarvothama M, Cooper K, Smythe KS, Dudakov JA, Pierce RH, Rader C, Riddell SR. Logic-Gated ROR1 Chimeric Antigen Receptor Expression Rescues T Cell-Mediated Toxicity to Normal Tissues and Enables Selective Tumor Targeting. Cancer Cell 2019; 35:489-503.e8. [PMID: 30889382 PMCID: PMC6450658 DOI: 10.1016/j.ccell.2019.02.003] [Citation(s) in RCA: 212] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 12/17/2018] [Accepted: 02/06/2019] [Indexed: 01/07/2023]
Abstract
Many potential targets for CAR-T cells in solid tumors are expressed in some normal tissues, raising concern for off-tumor toxicity. Following lymphodepletion, CAR-T cells targeting the tumor-associated antigen ROR1 lysed tumors in mice but induced lethal bone marrow failure due to recognition of ROR1+ stromal cells. To improve selectivity, we engineered T cells with synthetic Notch (synNotch) receptors specific for EpCAM or B7-H3, which are expressed on ROR1+ tumor cells but not ROR1+ stromal cells. SynNotch receptors induced ROR1 CAR expression selectively within the tumor, resulting in tumor regression without toxicity when tumor cells were segregated from, but not when co-localized with, normal ROR1+ cells. This strategy, thus, permits safe targeting of tumors that are sufficiently separated from normal cells.
Collapse
Affiliation(s)
- Shivani Srivastava
- Program in Immunology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, D3-100, Seattle, WA 98109-1024, USA.
| | - Alexander I Salter
- Program in Immunology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, D3-100, Seattle, WA 98109-1024, USA
| | - Denny Liggitt
- Department of Comparative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Sushma Yechan-Gunja
- Program in Immunology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, D3-100, Seattle, WA 98109-1024, USA
| | - Megha Sarvothama
- Program in Immunology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, D3-100, Seattle, WA 98109-1024, USA
| | - Kirsten Cooper
- Program in Immunology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, D3-100, Seattle, WA 98109-1024, USA
| | - Kimberly S Smythe
- Program in Immunology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, D3-100, Seattle, WA 98109-1024, USA
| | - Jarrod A Dudakov
- Program in Immunology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, D3-100, Seattle, WA 98109-1024, USA; Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Robert H Pierce
- Program in Immunology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, D3-100, Seattle, WA 98109-1024, USA
| | - Christoph Rader
- Department of Immunology and Microbiology, Scripps Research Institute, Jupiter, FL 33458, USA
| | - Stanley R Riddell
- Program in Immunology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, D3-100, Seattle, WA 98109-1024, USA; Department of Immunology, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
32
|
Zhang W, Yu Q, Liu H, Li B. Resident Prrx1 lineage stromal cells promote T cell survival in the spleen. J Mol Cell Biol 2019; 11:182-184. [PMID: 30624687 PMCID: PMC6392099 DOI: 10.1093/jmcb/mjy073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/23/2018] [Indexed: 12/04/2022] Open
Affiliation(s)
- Wanyao Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Yu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
33
|
Bogdanova D, Takeuchi A, Ozawa M, Kanda Y, Rahman MA, Ludewig B, Kinashi T, Katakai T. Essential Role of Canonical NF-κB Activity in the Development of Stromal Cell Subsets in Secondary Lymphoid Organs. THE JOURNAL OF IMMUNOLOGY 2018; 201:3580-3586. [PMID: 30397032 DOI: 10.4049/jimmunol.1800539] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 10/11/2018] [Indexed: 11/19/2022]
Abstract
Organized tissue structure in the secondary lymphoid organs (SLOs) tightly depends on the development of fibroblastic stromal cells (FSCs) of mesenchymal origin; however, the mechanisms of this relationship are poorly understood. In this study, we specifically inactivated the canonical NF-κB pathway in FSCs in vivo by conditionally inducing IκBα mutant in a Ccl19-IκBSR mouse system in which NF-κB activity is likely to be suppressed in fetal FSC progenitors. Given that NF-κB activation in fetal FSCs is essential for SLO development, the animals were expected to lack SLOs. However, all SLOs were preserved in Ccl19-IκBSR mice. Instead, the T cell area was severely disturbed by the lack of CCL21-expressing FSCs, whereas the follicles and associated FSC networks were formed. Fate mapping revealed that IκBSR-expressing cells constituted only a small fraction of stromal compartment outside the follicles. Taken together, our findings indicate an essential role of the canonical NF-κB pathway activity in the development of three FSC subsets common to SLOs and suggest transient or stochastic CCL19 expression in FSC progenitors and a compensatory differentiation program of follicular FSCs.
Collapse
Affiliation(s)
- Dana Bogdanova
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Arata Takeuchi
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Madoka Ozawa
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yasuhiro Kanda
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - M Azizur Rahman
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Burkhard Ludewig
- Institute of Immunobiology, CH-9007 St. Gallen, Switzerland; and
| | - Tatsuo Kinashi
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Tomoya Katakai
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan;
| |
Collapse
|
34
|
Schaeuble K, Britschgi MR, Scarpellino L, Favre S, Xu Y, Koroleva E, Lissandrin TKA, Link A, Matloubian M, Ware CF, Nedospasov SA, Tumanov AV, Cyster JG, Luther SA. Perivascular Fibroblasts of the Developing Spleen Act as LTα1β2-Dependent Precursors of Both T and B Zone Organizer Cells. Cell Rep 2018; 21:2500-2514. [PMID: 29186687 DOI: 10.1016/j.celrep.2017.10.119] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 09/08/2017] [Accepted: 10/30/2017] [Indexed: 12/24/2022] Open
Abstract
T and B cell compartmentalization is a hallmark of secondary lymphoid organs and is maintained by chemokine-expressing stromal cells. How this stromal cell network initially develops and differentiates into two distinct subsets is poorly known, especially for the splenic white pulp (WP). Here, we show that perivascular fibroblast precursors are triggered by LTα1β2 signals to expand, express CCL19/21, and then differentiate into two functionally distinct fibroblast subsets responsible for B and T cell clustering and WP compartmentalization. Failure to express or sense CCL19 leads to impaired T zone development, while lack of B cells or LTα1β2 leads to an earlier and stronger impairment in WP development. We therefore propose that WP development proceeds in multiple steps, with LTα1β2+ B cells acting as major inducer cells driving the expansion and gradual differentiation of perivascular fibroblasts into T and B zone organizer cells.
Collapse
Affiliation(s)
- Karin Schaeuble
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Mirjam R Britschgi
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Leo Scarpellino
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Stéphanie Favre
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Ying Xu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Ekaterina Koroleva
- University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | | | - Alexander Link
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Mehrdad Matloubian
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Carl F Ware
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sergei A Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilov Street, Moscow 119991, Russia
| | - Alexei V Tumanov
- University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sanjiv A Luther
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland.
| |
Collapse
|
35
|
A retinoic acid-dependent stroma-leukemia crosstalk promotes chronic lymphocytic leukemia progression. Nat Commun 2018; 9:1787. [PMID: 29725010 PMCID: PMC5934403 DOI: 10.1038/s41467-018-04150-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 03/14/2018] [Indexed: 12/19/2022] Open
Abstract
In chronic lymphocytic leukemia (CLL), the non-hematopoietic stromal microenvironment plays a critical role in promoting tumor cell recruitment, activation, survival, and expansion. However, the nature of the stromal cells and molecular pathways involved remain largely unknown. Here, we demonstrate that leukemic B lymphocytes induce the activation of retinoid acid synthesis and signaling in the microenvironment. Inhibition of RA-signaling in stromal cells causes deregulation of genes associated with adhesion, tissue organization and chemokine secretion including the B-cell chemokine CXCL13. Notably, reducing retinoic acid precursors from the diet or inhibiting RA-signaling through retinoid-antagonist therapy prolong survival by preventing dissemination of leukemia cells into lymphoid tissues. Furthermore, mouse and human leukemia cells could be distinguished from normal B-cells by their increased expression of Rarγ2 and RXRα, respectively. These findings establish a role for retinoids in murine CLL pathogenesis, and provide new therapeutic strategies to target the microenvironment and to control disease progression. The stromal microenvironment plays a key role in the expansion of chronic lymphocytic leukemia. Here, the authors use the Eµ-TCL1 mouse model to show that leukemic B-cells induce the activation of retinoic acid synthesis in stromal cells of the lymphoid microenvironment, and that impacting on retinoic acid signalling via diet or chemical inhibition prolonged survival by preventing leukemia dissemination and accumulation in lymphoid tissues.
Collapse
|
36
|
Golub R, Tan J, Watanabe T, Brendolan A. Origin and Immunological Functions of Spleen Stromal Cells. Trends Immunol 2018; 39:503-514. [PMID: 29567327 DOI: 10.1016/j.it.2018.02.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 01/20/2023]
Abstract
The mammalian spleen is a peripheral lymphoid organ that plays a central role in host defense. Consequently, the lack of spleen is often associated with immunodeficiency and increased risk of overwhelming infections. Growing evidence suggests that non-hematopoietic stromal cells are central players in spleen development, organization, and immune functions. In addition to its immunological role, the spleen also provides a site for extramedullary hematopoiesis (EMH) in response to injuries. A deeper understanding of the biology of stromal cells is therefore essential to fully comprehend how these cells modulate the immune system during normal and pathological conditions. Here, we review the specificities of the different mouse spleen stromal cell subsets and complement the murine studies with human data when available.
Collapse
Affiliation(s)
- Rachel Golub
- Unit for Lymphopoiesis, Immunology Department, INSERM U1223, University Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Institut Pasteur, Paris, France.
| | - Jonathan Tan
- Clem Jones Research Centre for Regenerative Medicine, Faculty of Health Sciences & Medicine, Bond University, Gold Coast, Australia
| | - Takeshi Watanabe
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Andrea Brendolan
- Unit of Lymphoid Organ Development, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
37
|
Prados A, Muñoz-Fernández R, Fernandez-Rubio P, Olivares EG. Characterization of mesenchymal stem/stromal cells with lymphoid tissue organizer cell potential in tonsils from children. Eur J Immunol 2018; 48:829-843. [PMID: 29435977 DOI: 10.1002/eji.201746963] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 12/15/2017] [Accepted: 02/07/2018] [Indexed: 12/18/2022]
Abstract
Lymphoid tissue organizer (LTo) cells, identified in mouse and human embryos, are thought to be precursors of stromal cells in secondary lymphoid organs. Whether LTo cells are present in human adults, however remains unknown. We obtained 15 stromal cell lines from tonsils from children who underwent tonsillectomy, and studied the antigen phenotype of these tonsil stromal cell (TSC) lines by flow cytometry and RT-PCR. Cell lines met the minimal criteria proposed by the International Society for Cellular Therapy to define human mesenchymal stem/stromal cells (MSCs): plastic-adherent capacity; expression of CD73, CD90 and CD105, lack of CD45, CD19 and HLA-DR; and capacity to differentiate into adipocytes, osteoblasts and chondrocytes. Furthermore, our TSC lines exhibited an antigen phenotype and functional characteristics very similar to those seen in murine embryo LTo cells: they expressed chemokines CCL19, CCL21 and CXCL13, cytokines TRANCE and IL-7, and adhesion molecules ICAM-1, mucosal addressin cell adhesion molecule (MadCAM)-1 and VCAM-1. The expression of LTo cell-associated markers and functions were upregulated by lymphotoxin (LT)α1β2 and TNF, two cytokines involved in the development and maturation of secondary lymphoid tissues. Our results show that TSCs are tonsil MSCs that differentiate into LTo-like cells in response to the effects of these cytokines.
Collapse
Affiliation(s)
- Alejandro Prados
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Spain
| | - Raquel Muñoz-Fernández
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Spain
| | - Pablo Fernandez-Rubio
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Spain
| | - Enrique G Olivares
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Facultad de Medicina, Universidad de Granada, Spain.,Unidad de Gestión Clínica Laboratorios, Complejo Hospitalario Universitario de Granada, Spain
| |
Collapse
|
38
|
Vojkovics D, Kellermayer Z, Kajtár B, Roncador G, Vincze Á, Balogh P. Nkx2-3-A Slippery Slope From Development Through Inflammation Toward Hematopoietic Malignancies. Biomark Insights 2018; 13:1177271918757480. [PMID: 29449776 PMCID: PMC5808962 DOI: 10.1177/1177271918757480] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 01/10/2018] [Indexed: 12/11/2022] Open
Abstract
The development of peripheral lymphoid tissues from the mesoderm is the result of a complex convergence combining lymphohematopoietic differentiation with the local specification of nonhematopoietic mesenchymal components. Although the various transcriptional regulators with fate-determining effects in diversifying the mobile leukocyte subsets have been thoroughly studied and identified, the tissue-specific determinants promoting the regional differentiation of resident mesenchyme are less understood. Of these factors, various members of the NK-class Nkx paralogues have emerged as key regulators for the organogenesis of spleen and mucosal lymphoid tissues, and recent data have also indicated their involvement in various pathological events, including gut inflammation and hematopoietic malignancies. Here, we summarize available data on the roles of Nkx2-3 in lymphoid tissue development and discuss its possible value as a developmental marker and disease-associated pathogenic trait.
Collapse
Affiliation(s)
- Dóra Vojkovics
- Department of Immunology and Biotechnology, Medical School, University of Pécs, Pécs, Hungary.,Lymphoid Organogenesis Research Group, Szentágothai János Research Center, University of Pécs, Pécs, Hungary
| | - Zoltán Kellermayer
- Department of Immunology and Biotechnology, Medical School, University of Pécs, Pécs, Hungary.,Lymphoid Organogenesis Research Group, Szentágothai János Research Center, University of Pécs, Pécs, Hungary
| | - Béla Kajtár
- Department of Pathology, Medical School, University of Pécs, Pécs, Hungary
| | | | - Áron Vincze
- 1st Department of Internal Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Balogh
- Department of Immunology and Biotechnology, Medical School, University of Pécs, Pécs, Hungary.,Lymphoid Organogenesis Research Group, Szentágothai János Research Center, University of Pécs, Pécs, Hungary
| |
Collapse
|
39
|
Tan JKH, Watanabe T. Determinants of postnatal spleen tissue regeneration and organogenesis. NPJ Regen Med 2018; 3:1. [PMID: 29367882 PMCID: PMC5770394 DOI: 10.1038/s41536-018-0039-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/01/2018] [Accepted: 01/02/2018] [Indexed: 12/27/2022] Open
Abstract
The spleen is an organ that filters the blood and is responsible for generating blood-borne immune responses. It is also an organ with a remarkable capacity to regenerate. Techniques for splenic auto-transplantation have emerged to take advantage of this characteristic and rebuild spleen tissue in individuals undergoing splenectomy. While this procedure has been performed for decades, the underlying mechanisms controlling spleen regeneration have remained elusive. Insights into secondary lymphoid organogenesis and the roles of stromal organiser cells and lymphotoxin signalling in lymph node development have helped reveal similar requirements for spleen regeneration. These factors are now considered in the regulation of embryonic and postnatal spleen formation, and in the establishment of mature white pulp and marginal zone compartments which are essential for spleen-mediated immunity. A greater understanding of the cellular and molecular mechanisms which control spleen development will assist in the design of more precise and efficient tissue grafting methods for spleen regeneration on demand. Regeneration of organs which harbour functional white pulp tissue will also offer novel opportunities for effective immunotherapy against cancer as well as infectious diseases.
Collapse
Affiliation(s)
- Jonathan K. H. Tan
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD 4229 Australia
| | - Takeshi Watanabe
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507 Japan
- The Tazuke Kofukai Medical Research Institute/Kitano Hospital, Osaka, 530-8480 Japan
| |
Collapse
|
40
|
Buechler MB, Turley SJ. A short field guide to fibroblast function in immunity. Semin Immunol 2017; 35:48-58. [PMID: 29198601 DOI: 10.1016/j.smim.2017.11.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/06/2017] [Indexed: 12/27/2022]
Abstract
Fibroblasts in secondary lymphoid organs, or fibroblastic reticular cells (FRC), are gate-keepers of immune responses. Here, we frame how these cells regulate immune responses via a three-part scheme in which FRC can setup, support or suppress immune responses. We also review how fibroblasts from non-lymphoid tissues influence immunity and highlight how they resemble and differ from FRC. Overall, we aim to focus attention on the emerging roles of lymphoid tissue and non-lymphoid tissue fibroblasts in control of innate and adaptive immunity.
Collapse
Affiliation(s)
- Matthew B Buechler
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, United States
| | - Shannon J Turley
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, United States.
| |
Collapse
|
41
|
Kellermayer Z, Vojkovics D, Balogh P. Innate lymphoid cells and their stromal microenvironments. Immunol Lett 2017; 189:3-9. [PMID: 28414182 DOI: 10.1016/j.imlet.2017.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/12/2017] [Indexed: 12/31/2022]
Abstract
In addition to the interaction between antigen presenting cells, T and B lymphocytes, recent studies have revealed important roles for a diverse set of auxiliary cells that profoundly influence the induction and regulation of immune responses against pathogens. Of these the stromal cells composed of various non-hematopoietic constituents are crucial for the creation and maintenance of specialized semi-static three-dimensional lymphoid tissue microenvironment, whereas the more recently described innate lymphoid cells are generated by the diversification of committed lymphoid precursor cells independently from clonally rearranged antigen receptor genes. Recent findings have revealed important contributions by innate lymphoid cells in inflammation and protection against pathogens in a tissue-specific manner. Importantly, lymphoid stromal cells also influence the onset of immune responses in tissue-specific fashion, raising the possibility of tissue-specific stromal - innate lymphoid cell collaboration. In this review we summarize the main features and interactions between these two cells types, with particular emphasis on ILC type 3 cells and their microenvironmental partners.
Collapse
Affiliation(s)
- Zoltán Kellermayer
- Department of Immunology and Biotechnology, Szentágothai Research Center, University of Pécs, Hungary; Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs, Hungary
| | - Dóra Vojkovics
- Department of Immunology and Biotechnology, Szentágothai Research Center, University of Pécs, Hungary; Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs, Hungary
| | - Péter Balogh
- Department of Immunology and Biotechnology, Szentágothai Research Center, University of Pécs, Hungary; Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs, Hungary.
| |
Collapse
|
42
|
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: 2.1] [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
|
43
|
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: 14.5] [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
|
44
|
Heesters BA, van der Poel CE, Das A, Carroll MC. Antigen Presentation to B Cells. Trends Immunol 2016; 37:844-854. [PMID: 27793570 DOI: 10.1016/j.it.2016.10.003] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 10/06/2016] [Accepted: 10/06/2016] [Indexed: 12/20/2022]
Abstract
Unlike T cells that recognize digested peptides, B cells recognize their cognate antigen in its native form. The B cell receptor used in recognition can also be secreted to bind to antigens and initiate multiple effector functions such as phagocytosis, complement activation, or neutralization of receptors. While B cells can interact with soluble antigens, it is now clear that the presentation of membrane-bound antigen plays a crucial role in B cell activation, and in particular during affinity-maturation, the process during which high-affinity B cells are selected. In this review we discuss how native antigen is presented to B cells and its impact at several stages of B cell responses.
Collapse
Affiliation(s)
- Balthasar A Heesters
- Program in Cellular and Molecular Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Current address: Department of Cell Biology and Histology, Academic Medical Center, Amsterdam, 1105 AZ, The Netherlands
| | - Cees E van der Poel
- Program in Cellular and Molecular Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Abhishek Das
- Program in Cellular and Molecular Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Michael C Carroll
- Program in Cellular and Molecular Medicine, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
45
|
Sato K, Honda SI, Shibuya A, Shibuya K. Improved protocol for the isolation of naïve follicular dendritic cells. Mol Immunol 2016; 78:140-145. [PMID: 27639061 DOI: 10.1016/j.molimm.2016.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/10/2016] [Accepted: 09/09/2016] [Indexed: 10/21/2022]
Abstract
Follicular dendritic cells (FDCs) in lymphoid organs play an important role in the humoral immune response. However, because the isolation of FDCs is difficult due to their very small population size and fragility under mechanical and chemical stresses, the genetic and biochemical characteristics of FDCs remain unclear. Previously, we identified FDCs as ICAM-1+ cells in the CD45- non-hematopoietic cell fraction from naïve mouse spleen after cell separation by means of digestion with a combination of enzymes. In the present study, using a new combination of enzymes, we found that FDCs are highly enriched in the CD45-ICAM-1+CD21/35+ cell fraction. CD45-ICAM-1+CD21/35+ cells in the mouse spleen retained an antigen administered in vivo for more than 7 days. Moreover, CD45-ICAM-1+CD21/35+ cells isolated from the spleen of mice administered with a cognate antigen enhanced the survival and proliferation of antigen-specific B cells in vitro. Our improved protocol for the isolation of naïve FDCs will be useful for the analysis of FDCs in vitro and in vivo.
Collapse
Affiliation(s)
- Kazuki Sato
- Department of Immunology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Shin-Ichiro Honda
- Department of Immunology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki 305-8575, Japan; Life Science Center of Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| | - Kazuko Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
46
|
Prados A, Kollias G, Koliaraki V. CollagenVI-Cre mice: A new tool to target stromal cells in secondary lymphoid organs. Sci Rep 2016; 6:33027. [PMID: 27604178 PMCID: PMC5015111 DOI: 10.1038/srep33027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/16/2016] [Indexed: 12/26/2022] Open
Abstract
Stromal cells in secondary lymphoid organs (SLOs) are non-hematopoietic cells involved in the regulation of adaptive immune responses. Three major stromal populations have been identified in adult SLOs: fibroblastic reticular cells (FRCs), follicular dendritic cells (FDCs) and marginal reticular cells (MRCs). The properties of these individual populations are not clearly defined, mainly due to the lack of appropriate genetic tools, especially for MRCs. Here, we analyzed stromal cell targeting in SLOs from a transgenic mouse strain that expresses Cre recombinase under the CollagenVI promoter, using lineage tracing approaches. We show that these mice target specifically MRCs and FDCs, but not FRCs in Peyer’s patches and isolated lymphoid follicles in the intestine. In contrast, stromal cells in lymph nodes and the spleen do not express the transgene, which renders ColVI-cre mice ideal for the specific targeting of stromal cells in the gut-associated lymphoid tissue (GALT). This funding further supports the hypothesis of organ-specific stromal precursors in SLOs. Interestingly, in all tissues analyzed, there was also high specificity for perivascular cells, which have been proposed to act as FDC precursors. Taken together, ColVI-Cre mice are a useful new tool for the dissection of MRC- and FDC-specific functions and plasticity in the GALT.
Collapse
Affiliation(s)
- Alejandro Prados
- Biomedical Sciences Research Center "Alexander Fleming", 16672 Vari, Greece
| | - George Kollias
- Biomedical Sciences Research Center "Alexander Fleming", 16672 Vari, Greece.,Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Vasiliki Koliaraki
- Biomedical Sciences Research Center "Alexander Fleming", 16672 Vari, Greece
| |
Collapse
|
47
|
Koning JJ, Konijn T, Lakeman KA, O'Toole T, Kenswil KJG, Raaijmakers MHGP, Michurina TV, Enikolopov G, Mebius RE. Nestin-Expressing Precursors Give Rise to Both Endothelial as well as Nonendothelial Lymph Node Stromal Cells. THE JOURNAL OF IMMUNOLOGY 2016; 197:2686-94. [PMID: 27574301 DOI: 10.4049/jimmunol.1501162] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 08/02/2016] [Indexed: 01/01/2023]
Abstract
During embryogenesis, lymph nodes form through intimate interaction between lymphoid tissue inducer and lymphoid tissue organizer (LTo) cells. Shortly after birth in mice, specialized stromal cell subsets arise that organize microenvironments within the lymph nodes; however, their direct precursors have not yet been identified. In the bone marrow, mesenchymal stem cells are labeled with GFP in nestin-GFP mice, and we show that during all stages of development, nestin(+) cells are present within lymph nodes of these mice. At day of birth, both mesenchymal CD31(-) and endothelial CD31(+) LTo cells were GFP(+), and only the population of CD31(-) LTo cells contained mesenchymal precursors. These CD31(-)nestin(+) cells are found in the T and B cell zones or in close association with high endothelial venules in adult lymph nodes. Fate mapping of nestin(+) cells unambiguously revealed the contribution of nestin(+) precursor cells to the mesenchymal as well as the endothelial stromal populations within lymph nodes. However, postnatal tamoxifen induced targeting of nestin(+) cells in nes-creER mice showed that most endothelial cells and only a minority of the nonendothelial cells were labeled. Overall our data show that nestin(+) cells contribute to all subsets of the complex stromal populations that can be found in lymph nodes.
Collapse
Affiliation(s)
- Jasper J Koning
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands
| | - Tanja Konijn
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands
| | - Kim A Lakeman
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands
| | - Tom O'Toole
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands
| | - Keane J G Kenswil
- Department of Hematology, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands; Erasmus Stem Cell Institute, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Marc H G P Raaijmakers
- Department of Hematology, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands; Erasmus Stem Cell Institute, Erasmus University Medical Center, 3015 GE Rotterdam, the Netherlands
| | - Tatyana V Michurina
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794; Center for Developmental Genetics, Stony Brook University, Stony Brook, NY 11794; Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724; and Department of Nano-, Bio-, Information, and Cognitive Sciences, Moscow Institute of Physics and Technology, 123182 Moscow, Russia
| | - Grigori Enikolopov
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794; Center for Developmental Genetics, Stony Brook University, Stony Brook, NY 11794; Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724; and Department of Nano-, Bio-, Information, and Cognitive Sciences, Moscow Institute of Physics and Technology, 123182 Moscow, Russia
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands;
| |
Collapse
|
48
|
Wang S, Xia P, Chen Y, Huang G, Xiong Z, Liu J, Li C, Ye B, Du Y, Fan Z. Natural Killer-like B Cells Prime Innate Lymphocytes against Microbial Infection. Immunity 2016; 45:131-44. [PMID: 27421702 DOI: 10.1016/j.immuni.2016.06.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/06/2016] [Accepted: 06/15/2016] [Indexed: 02/05/2023]
Abstract
Natural killer (NK) cells and non-cytotoxic interferon-γ (IFN-γ)-producing group I innate lymphoid cells (ILC1s) produce large amounts of IFN-γ and cause activation of innate and adaptive immunity. However, how NKs and ILC1s are primed during infection remains elusive. Here we have shown that a lymphocyte subpopulation natural killer-like B (NKB) cells existed in spleen and mesenteric lymph nodes (MLNs). NKBs had unique features that differed from T and B cells, and produced interleukin-18 (IL-18) and IL-12 at an early phase of infection. NKB cells played a critical role in eradication of microbial infection via secretion of IL-18 and IL-12. Moreover, IL-18 deficiency abrogated the antibacterial effect of NKBs. Upon bacterial challenge, NKB precursors (NKBPs) rapidly differentiated to NKBs that activated NKs and ILC1s against microbial infection. Our findings suggest that NKBs might be exploited to develop effective therapies for treatment of infectious diseases.
Collapse
Affiliation(s)
- Shuo Wang
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Pengyan Xia
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Chen
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guanling Huang
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhen Xiong
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jing Liu
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Chong Li
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Buqing Ye
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ying Du
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zusen Fan
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
49
|
Zewdu R, Risolino M, Barbulescu A, Ramalingam P, Butler JM, Selleri L. Spleen hypoplasia leads to abnormal stress hematopoiesis in mice with loss of Pbx homeoproteins in splenic mesenchyme. J Anat 2016; 229:153-69. [PMID: 27075259 PMCID: PMC5341595 DOI: 10.1111/joa.12479] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2016] [Indexed: 01/01/2023] Open
Abstract
The spleen plays critical roles in immunity and also provides a permissive microenvironment for hematopoiesis. Previous studies have reported that the TALE-class homeodomain transcription factor Pbx1 is essential in hematopoietic stem and progenitor cells (HSPCs) for stem cell maintenance and progenitor expansion. However, the role of Pbx1 in the hematopoietic niche has not been investigated. Here we explored the effects that genetic perturbation of the splenic mesenchymal niche has on hematopoiesis upon loss of members of the Pbx family of homeoproteins. Splenic mesenchyme-specific inactivation of Pbx1 (SKO) on a Pbx2- or Pbx3-deficient genetic background (DKO) resulted in abnormal development of the spleen, which is dysmorphic and severely hypoplastic. This phenotype, in turn, affected the number of HSPCs in the fetal and adult spleen at steady state, as well as markedly impairing the kinetics of hematopoietic regeneration in adult mice after sub-lethal and lethal myelosuppressive irradiation. Spleens of mice with compound Pyx deficiency 8 days following sublethal irradiation displayed significant downregulation of multiple cytokine-encoding genes, including KitL/SCF, Cxcl12/SDF-1, IL-3, IL-4, GM-CSF/Csf2 IL-10, and Igf-1, compared with controls. KitL/SCF and Cxcl12/SDF-1 were recently shown to play key roles in the splenic niche in response to various haematopoietic stresses such as myeloablation, blood loss, or pregnancy. Our results demonstrate that, in addition to their intrinsic roles in HSPCs, non-cell autonomous functions of Pbx factors within the splenic niche contribute to the regulation of hematopoiesis, at least in part via the control of KitL/SCF and Cxcl12/SDF-1. Furthermore, our study establishes that abnormal spleen development and hypoplasia have deleterious effects on the efficiency of hematopoietic recovery after bone marrow injury.
Collapse
Affiliation(s)
- Rediet Zewdu
- Department of Cell and Developmental BiologyWeill Cornell MedicineNew YorkNYUSA
- Present address: Huntsman Cancer Institute University of UtahSalt Lake CityUTUSA
| | - Maurizio Risolino
- Department of Cell and Developmental BiologyWeill Cornell MedicineNew YorkNYUSA
- Program in Craniofacial BiologyDepartment of Orofacial Sciences & Department of AnatomyUniversity of California San FranciscoSan FranciscoCAUSA
| | | | | | - Jason M. Butler
- Department of Genetic MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Licia Selleri
- Department of Cell and Developmental BiologyWeill Cornell MedicineNew YorkNYUSA
- Program in Craniofacial BiologyDepartment of Orofacial Sciences & Department of AnatomyUniversity of California San FranciscoSan FranciscoCAUSA
| |
Collapse
|
50
|
Kranich J, Krautler NJ. How Follicular Dendritic Cells Shape the B-Cell Antigenome. Front Immunol 2016; 7:225. [PMID: 27446069 PMCID: PMC4914831 DOI: 10.3389/fimmu.2016.00225] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 05/26/2016] [Indexed: 12/21/2022] Open
Abstract
Follicular dendritic cells (FDCs) are stromal cells residing in primary follicles and in germinal centers of secondary and tertiary lymphoid organs (SLOs and TLOs). There, they play a crucial role in B-cell activation and affinity maturation of antibodies. FDCs have the unique capacity to bind and retain native antigen in B-cell follicles for long periods of time. Therefore, FDCs shape the B-cell antigenome (the sum of all B-cell antigens) in SLOs and TLOs. In this review, we discuss recent findings that explain how this stromal cell type can arise in almost any tissue during TLO formation and, furthermore, focus on the mechanisms of antigen capture and retention involved in the generation of long-lasting antigen depots displayed on FDCs.
Collapse
Affiliation(s)
- Jan Kranich
- Institute for Immunology, Ludwig Maximilian University Munich, Munich, Germany
| | | |
Collapse
|