1
|
Fujii S, Sugino N, Miura Y. The Supportive Role of Lymph Node Mesenchymal Stromal Cells in Follicular Lymphoma Involves the PITX1-hTERT-Podoplanin Axis. Stem Cells Dev 2025; 34:201-213. [PMID: 40130551 DOI: 10.1089/scd.2025.0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025] Open
Abstract
The microenvironment within lymph nodes plays a pivotal role in the pathogenesis of follicular lymphoma (FL), a malignancy characterized by the accumulation of neoplastic B cells. Here, we report that human FL lymph node mesenchymal stromal cells (FLSCs) display surface protein expression profiles consistent with the standard phenotypic criteria for human mesenchymal stromal/stem cells (MSCs), yet exhibit reduced mesenchymal differentiation capability. FLSCs did not show the typical immunomodulatory protein expression patterns observed in fibroblastic reticular cells, marginal reticular cells, or follicular dendritic cells, as they expressed chemokine (C-X-C motif) ligand 13 and podoplanin but lacked chemokine (C-C motif) ligand 19 and complement receptor 1/2. Functionally, FLSCs exhibited superior FL cell survival-supportive capability in cocultures compared with bone marrow MSCs. This supportive effect was reduced when the cell culture inserts were used. In addition, this supportive capability was accompanied by reduced levels of B-cell-supportive soluble factors such as interleukin-6, regardless of the presence of cell culture inserts. Thus, both cell-cell contact-dependent and -independent mechanisms are involved in this process. Comprehensive transcriptomic analysis revealed that transcription factor paired-like homeodomain 1 (PITX1) is downregulated in FLSCs. Given that PITX1 regulates human telomerase reverse transcriptase (hTERT) transcription, FLSCs exhibited longer telomeres and a higher population-doubling capacity than MSCs. Furthermore, FLSCs expressed elevated podoplanin, whereas MSCs did not. Notably, hTERT-transfected MSCs also showed increased podoplanin expression, suggesting a positive association between hTERT and podoplanin. In summary, our findings indicate that FLSCs deviate from classical MSCs in their differentiation potential and instead exhibit a protumorigenic phenotype. This phenotype supports FL cell survival and is potentially mediated by an aberrant PITX1-hTERT-podoplanin signaling axis. These results highlight the critical role of FLSCs in the FL lymph node microenvironment, with implications for understanding tumor-supportive niches in FL pathogenesis.
Collapse
Affiliation(s)
- Sumie Fujii
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
- Department of Transfusion Medicine and Cell Therapy, Fujita Health University School of Medicine, Aichi, Japan
| | - Noriko Sugino
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
- Department of Hematology, Osaka Red Cross Hospital, Osaka, Japan
| | - Yasuo Miura
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
- Department of Transfusion Medicine and Cell Therapy, Fujita Health University School of Medicine, Aichi, Japan
| |
Collapse
|
2
|
Gocht A, Watermann C, Melzer M, Schumacher U. Lymph node or lymphoid aggregate? Impact on cancer resection quality, clinical prognosis, and tumor staging. Histol Histopathol 2025; 40:133-146. [PMID: 38813797 DOI: 10.14670/hh-18-760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
The clinical outcome of most cancer patients depends on the stage of the primary tumor, the lymph node status, and if distant metastases are present. According to the Union for International Cancer Control (UICC) and the American Joint Committee on Cancer (AJCC), the Tumor Node Metastasis (TNM) classification of malignant tumors requires the examination of a minimum number of regional lymph nodes for each type of cancer to fulfill the criteria of high-quality surgical oncology. Due to the daily challenge of collecting an appropriate number of lymph nodes and time constraints when processing and assessing tissue samples, pathologists may be tempted to identify every histological lymphoid structure mimicking a lymph node as a "true" lymph node. Faced with this issue, we propose to resolve it by specifying histological characteristics to differentiate lymphoid aggregates from "true" lymph nodes. To find a minimum consensus, we suggest defining as lymph nodes only those lymphoid structures composed of lymphoid cells encapsulated by a complete or incomplete fibrous capsule.
Collapse
Affiliation(s)
- Andreas Gocht
- Institut für Pathologie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.
| | - Christian Watermann
- Institut für Pathologie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Madelaine Melzer
- Institut für Pathologie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Udo Schumacher
- Department Humanmedizin, Medical School Berlin, Berlin, Germany
| |
Collapse
|
3
|
Llorca T, Ruiz-Magaña MJ, Abadía AC, Ruiz-Ruiz C, Olivares EG. Decidual stromal cells: fibroblasts specialized in immunoregulation during pregnancy. Trends Immunol 2025; 46:138-152. [PMID: 39947975 DOI: 10.1016/j.it.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/10/2024] [Accepted: 12/30/2024] [Indexed: 02/20/2025]
Abstract
Decidual stromal cells (DSCs) are involved in immunoregulatory mechanisms that prevent fetal rejection by the mammalian maternal immune system. Recent studies using single-cell RNA sequencing demonstrated the existence of different types of human and mouse DSCs, highlighting corresponding differentiation (decidualization) pathways, and suggesting their involvement in the immune response during normal and pathological pregnancy. DSCs may be considered tissue-specialized fibroblasts because both DSCs and fibroblasts share phenotypic and functional similarities in immunologically challenged tissues, especially in terms of their immune functions. Indeed, fibroblasts can setup, support, and suppress immune responses and these functions are also performed by DSCs. Moreover, fibroblasts and DSCs can induce ectopic foci as tertiary lymphoid structures (TLSs), and endometriosis, respectively. Thus, understanding DSC immunoregulatory functions is of timely relevance.
Collapse
Affiliation(s)
- Tatiana Llorca
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain
| | - María José Ruiz-Magaña
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain; Departamento de Biología Celular, Universidad de Granada, Granada, Spain.
| | - Ana C Abadía
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain; Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
| | - Carmen Ruiz-Ruiz
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain; Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
| | - Enrique G Olivares
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain; Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain.
| |
Collapse
|
4
|
Nayar S, Turner JD, Asam S, Fennell E, Pugh M, Colafrancesco S, Berardicurti O, Smith CG, Flint J, Teodosio A, Iannizzotto V, Gardner DH, van Roon J, Korsunsky I, Howdle D, Frei AP, Lassen KG, Bowman SJ, Ng WF, Croft AP, Filer A, Fisher BA, Buckley CD, Barone F. Molecular and spatial analysis of tertiary lymphoid structures in Sjogren's syndrome. Nat Commun 2025; 16:5. [PMID: 39747819 PMCID: PMC11697438 DOI: 10.1038/s41467-024-54686-0] [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: 10/20/2022] [Accepted: 11/18/2024] [Indexed: 01/04/2025] Open
Abstract
Tertiary lymphoid structures play important roles in autoimmune and non-autoimmune conditions. While many of the molecular mechanisms involved in tertiary lymphoid structure formation have been identified, the cellular sources and temporal and spatial relationship remain unknown. Here we use combine single-cell RNA-sequencing, spatial transcriptomics and proteomics of minor salivary glands of patients with Sjogren's disease and Sicca Syndrome, with ex-vivo functional studies to construct a cellular and spatial map of key components involved in the formation and function of tertiary lymphoid structures. We confirm the presence of a fibroblast cell state and identify a pericyte/mural cell state with potential immunological functions. The identification of cellular properties associated with these structures and the molecular and functional interactions identified by this analysis may provide key therapeutic cues for tertiary lymphoid structures associated conditions in autoimmunity and cancer.
Collapse
Affiliation(s)
- Saba Nayar
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Birmingham Tissue Analytics, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Jason D Turner
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Saba Asam
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
- UCL Genomics, Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | - Eanna Fennell
- School of Medicine & HRI & Bernal Institute, University of Limerick, Limerick, Ireland
| | - Matthew Pugh
- Department of Immunology and Immunotherapy, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | | | - Onorina Berardicurti
- Rheumatology, Immunology and Clinical Medicine Unit, Department of Medicine, Università Campus Bio-Medico, Rome, and Immunorheumatology Unit, Fondazione Policlinico Universitario Campus Bio Medico, Rome, Italy
| | - Charlotte G Smith
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Joe Flint
- Birmingham Tissue Analytics, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Ana Teodosio
- Birmingham Tissue Analytics, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Valentina Iannizzotto
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - David H Gardner
- Birmingham Tissue Analytics, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Joel van Roon
- Department of Rheumatology & Clinical Immunology/Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ilya Korsunsky
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
| | - Dawn Howdle
- Birmingham Tissue Analytics, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Andreas P Frei
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Kara G Lassen
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Simon J Bowman
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Wan-Fai Ng
- HRB Clinical Research Facility, University College Cork, Cork, Ireland
| | - Adam P Croft
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Andrew Filer
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Birmingham Tissue Analytics, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Benjamin A Fisher
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Christopher D Buckley
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Francesca Barone
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK.
- Candel Therapeutics, Needham, MA, USA.
| |
Collapse
|
5
|
Le Voyer T, Maglorius Renkilaraj MRL, Moriya K, Pérez Lorenzo M, Nguyen T, Gao L, Rubin T, Cederholm A, Ogishi M, Arango-Franco CA, Béziat V, Lévy R, Migaud M, Rapaport F, Itan Y, Deenick EK, Cortese I, Lisco A, Boztug K, Abel L, Boisson-Dupuis S, Boisson B, Frosk P, Ma CS, Landegren N, Celmeli F, Casanova JL, Tangye SG, Puel A. Inherited human RelB deficiency impairs innate and adaptive immunity to infection. Proc Natl Acad Sci U S A 2024; 121:e2321794121. [PMID: 39231201 PMCID: PMC11406260 DOI: 10.1073/pnas.2321794121] [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: 12/19/2023] [Accepted: 06/24/2024] [Indexed: 09/06/2024] Open
Abstract
We report two unrelated adults with homozygous (P1) or compound heterozygous (P2) private loss-of-function variants of V-Rel Reticuloendotheliosis Viral Oncogene Homolog B (RELB). The resulting deficiency of functional RelB impairs the induction of NFKB2 mRNA and NF-κB2 (p100/p52) protein by lymphotoxin in the fibroblasts of the patients. These defects are rescued by transduction with wild-type RELB complementary DNA (cDNA). By contrast, the response of RelB-deficient fibroblasts to Tumor Necrosis Factor (TNF) or IL-1β via the canonical NF-κB pathway remains intact. P1 and P2 have low proportions of naïve CD4+ and CD8+ T cells and of memory B cells. Moreover, their naïve B cells cannot differentiate into immunoglobulin G (IgG)- or immunoglobulin A (IgA)-secreting cells in response to CD40L/IL-21, and the development of IL-17A/F-producing T cells is strongly impaired in vitro. Finally, the patients produce neutralizing autoantibodies against type I interferons (IFNs), even after hematopoietic stem cell transplantation, attesting to a persistent dysfunction of thymic epithelial cells in T cell selection and central tolerance to some autoantigens. Thus, inherited human RelB deficiency disrupts the alternative NF-κB pathway, underlying a T- and B cell immunodeficiency, which, together with neutralizing autoantibodies against type I IFNs, confers a predisposition to viral, bacterial, and fungal infections.
Collapse
Affiliation(s)
- Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
- Clinical Immunology Department, Assistance Publique Hôpitaux de Paris, Saint-Louis Hospital, Paris75010, France
| | - Majistor Raj Luxman Maglorius Renkilaraj
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
| | - Kunihiko Moriya
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
| | - Malena Pérez Lorenzo
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
| | - Tina Nguyen
- Garvan Institute of Medical Research, Darlinghurst, NSW2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Sydney, NSW2052, Australia
| | - Liwei Gao
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
| | - Tamar Rubin
- Division of Pediatric Clinical Immunology and Allergy, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MBR3A 1S1, Canada
| | - Axel Cederholm
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, UppsalaSE-751 05, Sweden
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY10065
| | - Carlos A. Arango-Franco
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
- Group of Inborn Errors of Immunity, Department of Microbiology and Parasitology, School of Medicine, University of Antioquia, Medellín050010, Colombia
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY10065
| | - Romain Lévy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY10065
| | - Mélanie Migaud
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
| | - Franck Rapaport
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY10065
| | - Yuval Itan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - Elissa K. Deenick
- Garvan Institute of Medical Research, Darlinghurst, NSW2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Sydney, NSW2052, Australia
| | - Irene Cortese
- Experimental Immunotherapeutics Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD20892
| | - Andrea Lisco
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD20892
| | - Kaan Boztug
- St. Anna Children’s Cancer Research Institute, Vienna1090, Austria
- Medical University of Vienna, Department of Pediatrics and Adolescent Medicine, Vienna1090, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna1090, Austria
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY10065
| | - Stéphanie Boisson-Dupuis
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY10065
| | - Bertrand Boisson
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY10065
| | - Patrick Frosk
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MBR3E 0W2, Canada
| | - Cindy S. Ma
- Garvan Institute of Medical Research, Darlinghurst, NSW2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Sydney, NSW2052, Australia
| | - Nils Landegren
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, UppsalaSE-751 05, Sweden
| | - Fatih Celmeli
- Department of Allergy and Immunology, University of Medical Science, Antalya Education and Research Hospital, Antalya07100, Türkiye
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY10065
- Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children, Paris75015, France
- HHMI, New York, NY10065
| | - Stuart G. Tangye
- Garvan Institute of Medical Research, Darlinghurst, NSW2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Sydney, NSW2052, Australia
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Paris75015, France
- Imagine Institute, Paris Cité University, Paris75015, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY10065
| |
Collapse
|
6
|
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
|
7
|
Cardani-Boulton A, Sung SSJ, Petri WA, Hahn YS, Braciale TJ. Leptin Receptor Deficiency Impairs Lymph Node Development and Adaptive Immune Response. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:974-981. [PMID: 38251917 PMCID: PMC10932924 DOI: 10.4049/jimmunol.2100985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/03/2024] [Indexed: 01/23/2024]
Abstract
Activation and clonal expansion of the Ag-specific adaptive immune response in the draining lymph node is essential to clearing influenza A virus infections. Activation sufficient for virus clearance is dependent on the lymph node's architectural organization that is maintained by stromal cells, chiefly fibroblastic reticular cells. During an analysis of influenza A virus clearance in leptin receptor knockout (DB/DB) mice, we observed that the DB/DB mice have markedly reduced numbers of lymph node fibroblastic reticular cells at the steady state. The reduction in lymph node fibroblastic reticular cells resulted in abnormal lymph node organization and diminished numbers of adaptive immune cells in the lymph nodes under homeostatic conditions. As a consequence, the DB/DB mice were impaired in their ability to generate an effective influenza-specific adaptive immune response, which prevented virus clearance. Using leptin receptor mutant mice with point mutations at distinct signaling sites in the leptin receptor, we were able to link the leptin receptor's signaling domain tyrosine 985, which does not contribute to obesity, to lymph node fibroblastic reticular cell development and function. These results demonstrate a novel role for leptin receptor signaling in regulating lymph node development in a manner that is crucial to the generation of Ag-specific adaptive immune responses.
Collapse
Affiliation(s)
- Amber Cardani-Boulton
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, USA
| | - Sun-Sang J Sung
- Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - William A Petri
- Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Young S. Hahn
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Thomas J Braciale
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
8
|
Oranger A, Colaianni G, Ingravallo G, Scarcella VS, Faienza MF, Grano M, Colucci S, Brunetti G. LIGHT/TNFSF14 Affects Adipose Tissue Phenotype. Int J Mol Sci 2024; 25:716. [PMID: 38255789 PMCID: PMC10815871 DOI: 10.3390/ijms25020716] [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: 11/09/2023] [Revised: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
LIGHT/TNFSF14 is linked to several signaling pathways as a crucial member of a larger immunoregulatory network. It is primarily expressed in inflammatory effector cells, and high levels of LIGHT have been reported in obesity. Thus, with the aim of deepening the knowledge of the role of LIGHT on adipose tissue phenotype, we studied wild-type (WT), Tnfsf14-/-, Rag-/- and Rag-/Tnfsf14- (DKO) mice fed a normal diet (ND) or high-fat diet (HFD). Our results show that, although there is no significant weight gain between the mice with different genotypes, it is significant within each of them. We also detected an increase in visceral White Adipose Tissue (vWAT) weight in all mice fed HFD, together with the lowest levels of vWAT weight in Tnfsf14-/- and DKO mice fed ND with respect to the other strain. Inguinal WAT (iWAT) weight is significantly affected by genotype and HFD. The least amount of iWAT was detected in DKO mice fed ND. Histological analysis of vWAT showed that both the genotype and the diet significantly affect the adipocyte area, whereas the number is affected only by the genotype. In iWAT, the genotype and the diet significantly affect mean adipocyte area and number; interestingly, the area with the least adipocyte was detected in DKO mice fed ND, suggesting a potential browning effect due to the simultaneous lack of mature lymphocytes and LIGHT. Consistently, Uncoupling Protein 1 (UCP1) staining of iWAT demonstrated that few positive brown adipocytes appeared in DKO mice. Furthermore, LIGHT deficiency is associated with greater levels of UCP1, highlighting the lack of its expression in Rag-/- mice. Liver examination showed that all mice fed HFD had a steatotic liver, but it was particularly evident for DKO mice. In conclusion, our study demonstrates that the adipose tissue phenotype is affected by LIGHT levels but also much more by mature lymphocytes.
Collapse
Affiliation(s)
- Angela Oranger
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (A.O.); (G.C.); (M.G.)
| | - Graziana Colaianni
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (A.O.); (G.C.); (M.G.)
| | - Giuseppe Ingravallo
- Section of Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy; (G.I.); (V.S.S.)
| | - Vincenza Sara Scarcella
- Section of Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari, 70124 Bari, Italy; (G.I.); (V.S.S.)
| | - Maria Felicia Faienza
- Pediatric Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (A.O.); (G.C.); (M.G.)
| | - Silvia Colucci
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70124 Bari, Italy;
| | - Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy
| |
Collapse
|
9
|
Sylvestre M, Barbier N, Sibut V, Nayar S, Monvoisin C, Leonard S, Saint-Vanne J, Martin A, Guirriec M, Latour M, Jouan F, Baulande S, Bohec M, Verdière L, Mechta-Grigoriou F, Mourcin F, Bertheuil N, Barone F, Tarte K, Roulois D. KDM6B drives epigenetic reprogramming associated with lymphoid stromal cell early commitment and immune properties. SCIENCE ADVANCES 2023; 9:eadh2708. [PMID: 38019914 PMCID: PMC10686565 DOI: 10.1126/sciadv.adh2708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
Mature lymphoid stromal cells (LSCs) are key organizers of immune responses within secondary lymphoid organs. Similarly, inflammation-driven tertiary lymphoid structures depend on immunofibroblasts producing lymphoid cytokines and chemokines. Recent studies have explored the origin and heterogeneity of LSC/immunofibroblasts, yet the molecular and epigenetic mechanisms involved in their commitment are still unknown. This study explored the transcriptomic and epigenetic reprogramming underlying LSC/immunofibroblast commitment. We identified the induction of lysine demethylase 6B (KDM6B) as the primary epigenetic driver of early immunofibroblast differentiation. In addition, we observed an enrichment for KDM6B gene signature in murine inflammatory fibroblasts and pathogenic stroma of patients with autoimmune diseases. Last, KDM6B was required for the acquisition of LSC/immunofibroblast functional properties, including the up-regulation of CCL2 and the resulting recruitment of monocytes. Overall, our results reveal epigenetic mechanisms that participate in the early commitment and immune properties of immunofibroblasts and support the use of epigenetic modifiers as fibroblast-targeting strategies in chronic inflammation.
Collapse
Affiliation(s)
- Marvin Sylvestre
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Nicolas Barbier
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Vonick Sibut
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Saba Nayar
- Centre for Translational inflammation Research, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham Research Laboratories, Queen Elizabeth Hospital, Birmingham, UK
| | - Céline Monvoisin
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Simon Leonard
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, F-35043 Nantes, France
| | - Julien Saint-Vanne
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
- SITI, Pôle Biologie, CHU Rennes, F-35033 Rennes, France
| | - Ansie Martin
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Marion Guirriec
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Maëlle Latour
- SITI, Pôle Biologie, CHU Rennes, F-35033 Rennes, France
| | - Florence Jouan
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Sylvain Baulande
- Institut Curie Genomics of Excellence (ICGex) Platform, Institut Curie Research Center, PSL Research University, F-75005 Paris, France
| | - Mylène Bohec
- Institut Curie Genomics of Excellence (ICGex) Platform, Institut Curie Research Center, PSL Research University, F-75005 Paris, France
| | - Léa Verdière
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Fatima Mechta-Grigoriou
- Stress and Cancer Laboratory, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, INSERM, U830, PSL Research University, 26, rue d’Ulm, F-75005 Paris, France
| | - Frédéric Mourcin
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| | - Nicolas Bertheuil
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
- Department of Plastic Surgery, CHU Rennes, F-35033 Rennes, France
| | | | - Karin Tarte
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
- SITI, Pôle Biologie, CHU Rennes, F-35033 Rennes, France
| | - David Roulois
- Honeycomb team, Equipe Labellisée par la Ligue Nationale Contre le Cancer, Univ Rennes, INSERM, EFS, UMR S1236, Rennes, France
| |
Collapse
|
10
|
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: 5] [Impact Index Per Article: 2.5] [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
|
11
|
Arroz-Madeira S, Bekkhus T, Ulvmar MH, Petrova TV. Lessons of Vascular Specialization From Secondary Lymphoid Organ Lymphatic Endothelial Cells. Circ Res 2023; 132:1203-1225. [PMID: 37104555 PMCID: PMC10144364 DOI: 10.1161/circresaha.123.322136] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023]
Abstract
Secondary lymphoid organs, such as lymph nodes, harbor highly specialized and compartmentalized niches. These niches are optimized to facilitate the encounter of naive lymphocytes with antigens and antigen-presenting cells, enabling optimal generation of adaptive immune responses. Lymphatic vessels of lymphoid organs are uniquely specialized to perform a staggering variety of tasks. These include antigen presentation, directing the trafficking of immune cells but also modulating immune cell activation and providing factors for their survival. Recent studies have provided insights into the molecular basis of such specialization, opening avenues for better understanding the mechanisms of immune-vascular interactions and their applications. Such knowledge is essential for designing better treatments for human diseases given the central role of the immune system in infection, aging, tissue regeneration and repair. In addition, principles established in studies of lymphoid organ lymphatic vessel functions and organization may be applied to guide our understanding of specialization of vascular beds in other organs.
Collapse
Affiliation(s)
- Silvia Arroz-Madeira
- Department of Oncology, University of Lausanne, Switzerland (S.A.M., T.V.P.)
- Ludwig Institute for Cancer Research Lausanne, Switzerland (S.A.M., T.V.P.)
| | - Tove Bekkhus
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden (T.B., M.H.U.)
| | - Maria H. Ulvmar
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden (T.B., M.H.U.)
| | - Tatiana V. Petrova
- Department of Oncology, University of Lausanne, Switzerland (S.A.M., T.V.P.)
- Ludwig Institute for Cancer Research Lausanne, Switzerland (S.A.M., T.V.P.)
| |
Collapse
|
12
|
Bekkhus T, Olofsson A, Sun Y, Magnusson PU, Ulvmar MH. Stromal transdifferentiation drives lipomatosis and induces extensive vascular remodeling in the aging human lymph node. J Pathol 2023; 259:236-253. [PMID: 36367235 PMCID: PMC10108032 DOI: 10.1002/path.6030] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/18/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Lymph node (LN) lipomatosis is a common but rarely discussed phenomenon associated with aging that involves a gradual exchange of the LN parenchyma into adipose tissue. The mechanisms behind these changes and the effects on the LN are unknown. We show that LN lipomatosis starts in the medullary regions of the human LN and link the initiation of lipomatosis to transdifferentiation of LN fibroblasts into adipocytes. The latter is associated with a downregulation of lymphotoxin beta expression. We also show that isolated medullary and CD34+ fibroblasts, in contrast to the reticular cells of the T-cell zone, display an inherently higher sensitivity for adipogenesis. Progression of lipomatosis leads to a gradual loss of the medullary lymphatic network, but at later stages, collecting-like lymphatic vessels are found inside the adipose tissue. The stromal dysregulation includes a dramatic remodeling and dilation of the high endothelial venules associated with reduced density of naïve T-cells. Abnormal clustering of plasma cells is also observed. Thus, LN lipomatosis causes widespread stromal dysfunction with consequences for the immune contexture of the human LN. Our data warrant an increased awareness of LN lipomatosis as a factor contributing to decreased immune functions in the elderly and in disease. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Tove Bekkhus
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Anna Olofsson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Ying Sun
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Peetra U Magnusson
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Maria H Ulvmar
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
13
|
Zhao J, Jung S, Li X, Li L, Kasinath V, Zhang H, Movahedi SN, Mardini A, Sabiu G, Hwang Y, Saxena V, Song Y, Ma B, Acton SE, Kim P, Madsen JC, Sage PT, Tullius SG, Tsokos GC, Bromberg JS, Abdi R. Delivery of costimulatory blockade to lymph nodes promotes transplant acceptance in mice. J Clin Invest 2022; 132:e159672. [PMID: 36519543 PMCID: PMC9754003 DOI: 10.1172/jci159672] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 10/11/2022] [Indexed: 12/15/2022] Open
Abstract
The lymph node (LN) is the primary site of alloimmunity activation and regulation during transplantation. Here, we investigated how fibroblastic reticular cells (FRCs) facilitate the tolerance induced by anti-CD40L in a murine model of heart transplantation. We found that both the absence of LNs and FRC depletion abrogated the effect of anti-CD40L in prolonging murine heart allograft survival. Depletion of FRCs impaired homing of T cells across the high endothelial venules (HEVs) and promoted formation of alloreactive T cells in the LNs in heart-transplanted mice treated with anti-CD40L. Single-cell RNA sequencing of the LNs showed that anti-CD40L promotes a Madcam1+ FRC subset. FRCs also promoted the formation of regulatory T cells (Tregs) in vitro. Nanoparticles (NPs) containing anti-CD40L were selectively delivered to the LNs by coating them with MECA-79, which binds to peripheral node addressin (PNAd) glycoproteins expressed exclusively by HEVs. Treatment with these MECA-79-anti-CD40L-NPs markedly delayed the onset of heart allograft rejection and increased the presence of Tregs. Finally, combined MECA-79-anti-CD40L-NPs and rapamycin treatment resulted in markedly longer allograft survival than soluble anti-CD40L and rapamycin. These data demonstrate that FRCs are critical to facilitating costimulatory blockade. LN-targeted nanodelivery of anti-CD40L could effectively promote heart allograft acceptance.
Collapse
Affiliation(s)
- Jing Zhao
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sungwook Jung
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaofei Li
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lushen Li
- Department of Surgery and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Vivek Kasinath
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hengcheng Zhang
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Said N. Movahedi
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ahmad Mardini
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gianmarco Sabiu
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yoonha Hwang
- IVIM Technology, Daejeon, South Korea
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Vikas Saxena
- Department of Surgery and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Bing Ma
- Institute for Genome Sciences and
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sophie E. Acton
- Stromal Immunology Group, Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Pilhan Kim
- IVIM Technology, Daejeon, South Korea
- Graduate School of Nanoscience and Technology and
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Joren C. Madsen
- Center for Transplantation Sciences, Department of Surgery
- Division of Cardiac Surgery, Department of Surgery, and
| | - Peter T. Sage
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefan G. Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - George C. Tsokos
- Division of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan S. Bromberg
- Department of Surgery and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Reza Abdi
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Lee J, Park BC, Jang NY, Lee S, Cho YK, Sharma P, Byun SW, Jeon K, Jeon Y, Park U, Ro HJ, Park HR, Kim Y, Lee D, Chung S, Kim YK, Cho N. Inducing Ectopic T Cell Clusters Using Stromal Vascular Fraction Spheroid-Based Immunotherapy to Enhance Anti-Tumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203842. [PMID: 36058002 PMCID: PMC9534947 DOI: 10.1002/advs.202203842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/21/2022] [Indexed: 05/09/2023]
Abstract
Tertiary lymphoid structures (TLSs) provide specialized niches for immune cells, resulting in improved prognoses for patients undergoing cancer immunotherapy. Shaping TLS-like niches may improve anti-cancer immunity and overcome the current limitations of immune cell-based immunotherapy. Here, it is shown that stromal vascular fraction (SVF) from adipose tissues can enhance dendritic cell (DC)-mediated T cell immunity by inducing ectopic T lymphocyte clusters. SVF cells expanded ex vivo have phenotypes and functions similar to those of fibroblastic reticular cells in a secondary lymphoid organ, and their properties can be modulated using three-dimensional spheroid culture and coculture with DCs spiked with antigen-loaded iron oxide-zinc oxide core-shell nanoparticles. Thereby, the combination of SVF spheroids and mature DCs significantly augments T cell recruitment and retention at the injection site. This strategy elicits enhanced antigen-specific immune response and anti-tumoral immunity in mice, illustrating the potential for a novel immunotherapeutic design using SVF as a structural scaffold for TLS.
Collapse
Affiliation(s)
- Jae‐Won Lee
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
- Institute of Endemic DiseasesCollege of MedicineSeoul National UniversitySeoul03080Korea
| | - Bum Chul Park
- Department of Materials Science and EngineeringKorea UniversitySeoul02481Korea
- Brain Korea Center for Smart Materials and DevicesKorea UniversitySeoul02841Korea
| | - Na Yoon Jang
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Sihyeon Lee
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Young Kyu Cho
- School of Mechanical EngineeringKorea UniversitySeoul02841Republic of Korea
| | - Prashant Sharma
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Sang Won Byun
- Department of Materials Science and EngineeringKorea UniversitySeoul02481Korea
| | - Kyeongseok Jeon
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Yun‐Hui Jeon
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
| | - Uni Park
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Hyo Jin Ro
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Hyo Ree Park
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
| | - Yuri Kim
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
- Institute of Endemic DiseasesCollege of MedicineSeoul National UniversitySeoul03080Korea
| | - Dong‐Sup Lee
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
| | - Seok Chung
- School of Mechanical EngineeringKorea UniversitySeoul02841Republic of Korea
| | - Young Keun Kim
- Department of Materials Science and EngineeringKorea UniversitySeoul02481Korea
- Brain Korea Center for Smart Materials and DevicesKorea UniversitySeoul02841Korea
| | - Nam‐Hyuk Cho
- Department of Biomedical SciencesSeoul National University College of MedicineSeoul03080Korea
- Department of Microbiology and ImmunologySeoul National University College of MedicineSeoul03080Korea
- Institute of Endemic DiseasesCollege of MedicineSeoul National UniversitySeoul03080Korea
- Seoul National University Bundang HospitalSeongnam‐siGyeonggi‐do13620Republic of Korea
| |
Collapse
|
15
|
Ware CF, Croft M, Neil GA. Realigning the LIGHT signaling network to control dysregulated inflammation. J Exp Med 2022; 219:213236. [PMID: 35604387 PMCID: PMC9130030 DOI: 10.1084/jem.20220236] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 11/10/2022] Open
Abstract
Advances in understanding the physiologic functions of the tumor necrosis factor superfamily (TNFSF) of ligands, receptors, and signaling networks are providing deeper insight into pathogenesis of infectious and autoimmune diseases and cancer. LIGHT (TNFSF14) has emerged as an important modulator of critical innate and adaptive immune responses. LIGHT and its signaling receptors, herpesvirus entry mediator (TNFRSF14), and lymphotoxin β receptor, form an immune regulatory network with two co-receptors of herpesvirus entry mediator, checkpoint inhibitor B and T lymphocyte attenuator, and CD160. Deciphering the fundamental features of this network reveals new understanding to guide therapeutic development. Accumulating evidence from infectious diseases points to the dysregulation of the LIGHT network as a disease-driving mechanism in autoimmune and inflammatory reactions in barrier organs, including coronavirus disease 2019 pneumonia and inflammatory bowel diseases. Recent clinical results warrant further investigation of the LIGHT regulatory network and application of target-modifying therapeutics for disease intervention.
Collapse
Affiliation(s)
- Carl F Ware
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Michael Croft
- Division of Immune Regulation, La Jolla Institute for Immunology, La Jolla, CA
| | | |
Collapse
|
16
|
Denton AE, Dooley J, Cinti I, Silva-Cayetano A, Fra-Bido S, Innocentin S, Hill DL, Carr EJ, McKenzie ANJ, Liston A, Linterman MA. Targeting TLR4 during vaccination boosts MAdCAM-1 + lymphoid stromal cell activation and promotes the aged germinal center response. Sci Immunol 2022; 7:eabk0018. [PMID: 35522725 PMCID: PMC7612953 DOI: 10.1126/sciimmunol.abk0018] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The failure to generate enduring humoral immunity after vaccination is a hallmark of advancing age. This can be attributed to a reduction in the germinal center (GC) response, which generates long-lived antibody-secreting cells that protect against (re)infection. Despite intensive investigation, the primary cellular defect underlying impaired GCs in aging has not been identified. Here, we used heterochronic parabiosis to demonstrate that GC formation was dictated by the age of the lymph node (LN) microenvironment rather than the age of the immune cells. Lymphoid stromal cells are a key determinant of the LN microenvironment and are also an essential component underpinning GC structure and function. Using mouse models, we demonstrated that mucosal adressin cell adhesion molecule-1 (MAdCAM-1)-expressing lymphoid stromal cells were among the first cells to respond to NP-KLH + Alum immunization, proliferating and up-regulating cell surface proteins such as podoplanin and cell adhesion molecules. This response was essentially abrogated in aged mice. By targeting TLR4 using adjuvants, we improved the MAdCAM-1+ stromal cell response to immunization. This correlated with improved GC responses in both younger adult and aged mice, suggesting a link between stromal cell responses to immunization and GC initiation. Using bone marrow chimeras, we also found that MAdCAM-1+ stromal cells could respond directly to TLR4 ligands. Thus, the age-associated defect in GC and stromal cell responses to immunization can be targeted to improve vaccines in older people.
Collapse
Affiliation(s)
- Alice E Denton
- Immunology Programme, Babraham Institute, Cambridge UK
- Department of Immunology and Inflammation, Imperial College London, London UK
| | - James Dooley
- Immunology Programme, Babraham Institute, Cambridge UK
- Adaptive Immunology Laboratory, VIB and University of Leuven, Leuven Belgium
| | - Isabella Cinti
- Department of Immunology and Inflammation, Imperial College London, London UK
| | | | | | | | - Danika L Hill
- Immunology Programme, Babraham Institute, Cambridge UK
- Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Victoria, Australia
| | - Edward J Carr
- Immunology Programme, Babraham Institute, Cambridge UK
- Department of Medicine, University of Cambridge, Cambridge UK
- The Francis Crick Institute, London UK
| | - Andrew NJ McKenzie
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Adrian Liston
- Immunology Programme, Babraham Institute, Cambridge UK
- Adaptive Immunology Laboratory, VIB and University of Leuven, Leuven Belgium
| | | |
Collapse
|
17
|
Jacob JM, Di Carlo SE, Stzepourginski I, Lepelletier A, Ndiaye PD, Varet H, Legendre R, Kornobis E, Benabid A, Nigro G, Peduto L. PDGFRα-induced stromal maturation is required to restrain postnatal intestinal epithelial stemness and promote defense mechanisms. Cell Stem Cell 2022; 29:856-868.e5. [PMID: 35523143 DOI: 10.1016/j.stem.2022.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 01/18/2022] [Accepted: 04/07/2022] [Indexed: 11/03/2022]
Abstract
After birth, the intestine undergoes major changes to shift from an immature proliferative state to a functional intestinal barrier. By combining inducible lineage tracing and transcriptomics in mouse models, we identify a prodifferentiation PDGFRαHigh intestinal stromal lineage originating from postnatal LTβR+ perivascular stromal progenitors. The genetic blockage of this lineage increased the intestinal stem cell pool while decreasing epithelial and immune maturation at weaning age, leading to reduced postnatal growth and dysregulated repair responses. Ablating PDGFRα in the LTBR stromal lineage demonstrates that PDGFRα has a major impact on the lineage fate and function, inducing a transcriptomic switch from prostemness genes, such as Rspo3 and Grem1, to prodifferentiation factors, including BMPs, retinoic acid, and laminins, and on spatial organization within the crypt-villus and repair responses. Our results show that the PDGFRα-induced transcriptomic switch in intestinal stromal cells is required in the first weeks after birth to coordinate postnatal intestinal maturation and function.
Collapse
Affiliation(s)
- Jean-Marie Jacob
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Selene E Di Carlo
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Igor Stzepourginski
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Anthony Lepelletier
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Papa Diogop Ndiaye
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Hugo Varet
- Transcriptome and Epigenome Platform-Biomics Pole, Institut Pasteur, Université Paris Cité, Paris, France; Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Rachel Legendre
- Transcriptome and Epigenome Platform-Biomics Pole, Institut Pasteur, Université Paris Cité, Paris, France; Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Etienne Kornobis
- Transcriptome and Epigenome Platform-Biomics Pole, Institut Pasteur, Université Paris Cité, Paris, France; Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Adam Benabid
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Giulia Nigro
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Lucie Peduto
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France.
| |
Collapse
|
18
|
Lenti E, Genovese L, Bianchessi S, Maurizio A, Sain SB, di Lillo A, Mattavelli G, Harel I, Bernassola F, Hehlgans T, Pfeffer K, Crosti M, Abrignani S, Evans SM, Sitia G, Guimarães-Camboa N, Russo V, van de Pavert SA, Garcia-Manteiga JM, Brendolan A. Fate mapping and scRNA sequencing reveal origin and diversity of lymph node stromal precursors. Immunity 2022; 55:606-622.e6. [PMID: 35358427 DOI: 10.1016/j.immuni.2022.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/30/2021] [Accepted: 03/03/2022] [Indexed: 11/25/2022]
Abstract
Lymph node (LN) stromal cells play a crucial role in LN development and in supporting adaptive immune responses. However, their origin, differentiation pathways, and transcriptional programs are still elusive. Here, we used lineage-tracing approaches and single-cell transcriptome analyses to determine origin, transcriptional profile, and composition of LN stromal and endothelial progenitors. Our results showed that all major stromal cell subsets and a large proportion of blood endothelial cells originate from embryonic Hoxb6+ progenitors of the lateral plate mesoderm (LPM), whereas lymphatic endothelial cells arise from Pax3+ progenitors of the paraxial mesoderm (PXM). Single-cell RNA sequencing revealed the existence of different Cd34+ and Cxcl13+ stromal cell subsets and showed that embryonic LNs contain proliferating progenitors possibly representing the amplifying populations for terminally differentiated cells. Taken together, our work identifies the earliest embryonic sources of LN stromal and endothelial cells and demonstrates that stromal diversity begins already during LN development.
Collapse
Affiliation(s)
- Elisa Lenti
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Genovese
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Bianchessi
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Aurora Maurizio
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Simona Baghai Sain
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessia di Lillo
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Greta Mattavelli
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Itamar Harel
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 9190401, Israel
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Thomas Hehlgans
- Leibniz Institute of Immunotherapy (LIT), Chair for Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Klaus Pfeffer
- Institute of Medical, Microbiology and Hospital Hygiene, University Hospital Düsseldorf, 40225 Düsseldorf, Germany
| | - Mariacristina Crosti
- INGM, Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy
| | - Sergio Abrignani
- INGM, Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy; Department of Clinical Science and Community Health (DISCCO), University of Milan, Milan, Italy
| | - Sylvia M Evans
- Skaggs School of Pharmacy, University of California at San Diego, La Jolla, CA 92093, USA
| | - Giovanni Sitia
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nuno Guimarães-Camboa
- Institute of Cardiovascular Regeneration, Goethe-University, Frankfurt 60590, Germany; German Center for Cardiovascular Research, Berlin (partner site Frankfurt Rhine-Main), Germany
| | - Vincenzo Russo
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serge A van de Pavert
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université, INSERM, CNRS, Marseille, France
| | | | - Andrea Brendolan
- Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
19
|
Davis JL, Pokhrel NK, Cox L, Rohatgi N, Faccio R, Veis DJ. Conditional loss of IKKα in Osterix + cells has no effect on bone but leads to age-related loss of peripheral fat. Sci Rep 2022; 12:4915. [PMID: 35318397 PMCID: PMC8940989 DOI: 10.1038/s41598-022-08914-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/10/2022] [Indexed: 11/09/2022] Open
Abstract
NF-κB has been reported to both promote and inhibit bone formation. To explore its role in osteolineage cells, we conditionally deleted IKKα, an upstream kinase required for non-canonical NF-κB activation, using Osterix (Osx)-Cre. Surprisingly, we found no effect on either cancellous or cortical bone, even following mechanical loading. However, we noted that IKKα conditional knockout (cKO) mice began to lose body weight after 6 months of age with severe reductions in fat mass and lower adipocyte size in geriatric animals. qPCR analysis of adipogenic markers in fat pads of cKO mice indicated no difference in early differentiation, but instead markedly lower leptin with age. We challenged young mice with a high fat diet finding that cKO mice gained less weight and showed improved glucose metabolism. Low levels of recombination at the IKKα locus were detected in fat pads isolated from old cKO mice. To determine whether recombination occurs in adipocytes, we examined fat pads in Osx-Cre;TdT reporter mice; these showed increasing Osx-Cre-mediated expression in peripheral adipocytes from 6 weeks to 18 months. Since Osx-Cre drives recombination in peripheral adipocytes with age, we conclude that fat loss in cKO mice is most likely caused by progressive deficits of IKKα in adipocytes.
Collapse
Affiliation(s)
- Jennifer L Davis
- Musculoskeletal Research Center, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Nitin Kumar Pokhrel
- Musculoskeletal Research Center, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Linda Cox
- Musculoskeletal Research Center, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Nidhi Rohatgi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Roberta Faccio
- Musculoskeletal Research Center, Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Shriners Hospitals for Children, St. Louis, MO, 63110, USA
| | - Deborah J Veis
- Musculoskeletal Research Center, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Shriners Hospitals for Children, St. Louis, MO, 63110, USA.
| |
Collapse
|
20
|
Nahmgoong H, Jeon YG, Park ES, Choi YH, Han SM, Park J, Ji Y, Sohn JH, Han JS, Kim YY, Hwang I, Lee YK, Huh JY, Choe SS, Oh TJ, Choi SH, Kim JK, Kim JB. Distinct properties of adipose stem cell subpopulations determine fat depot-specific characteristics. Cell Metab 2022; 34:458-472.e6. [PMID: 35021043 DOI: 10.1016/j.cmet.2021.11.014] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/16/2021] [Accepted: 11/22/2021] [Indexed: 12/22/2022]
Abstract
In mammals, white adipose tissues are largely divided into visceral epididymal adipose tissue (EAT) and subcutaneous inguinal adipose tissue (IAT) with distinct metabolic properties. Although emerging evidence suggests that subpopulations of adipose stem cells (ASCs) would be important to explain fat depot differences, ASCs of two fat depots have not been comparatively investigated. Here, we characterized heterogeneous ASCs and examined the effects of intrinsic and tissue micro-environmental factors on distinct ASC features. We demonstrated that ASC subpopulations in EAT and IAT exhibited different molecular features with three adipogenic stages. ASC transplantation experiments revealed that intrinsic ASC features primarily determined their adipogenic potential. Upon obesogenic stimuli, EAT-specific SDC1+ ASCs promoted fibrotic remodeling, whereas IAT-specific CXCL14+ ASCs suppressed macrophage infiltration. Moreover, IAT-specific BST2high ASCs exhibited a high potential to become beige adipocytes. Collectively, our data broaden the understanding of ASCs with new insights into the origin of white fat depot differences.
Collapse
Affiliation(s)
- Hahn Nahmgoong
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yong Geun Jeon
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun Seo Park
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea
| | - Yoon Ha Choi
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea
| | - Sang Mun Han
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeu Park
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yul Ji
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jee Hyung Sohn
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji Seul Han
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Ye Young Kim
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Injae Hwang
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yun Kyung Lee
- Internal Medicine, Seoul National University College of Medicine & Seoul National University Bundang Hospital, Seoul 03080, Republic of Korea
| | - Jin Young Huh
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung Sik Choe
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Tae Jung Oh
- Internal Medicine, Seoul National University College of Medicine & Seoul National University Bundang Hospital, Seoul 03080, Republic of Korea
| | - Sung Hee Choi
- Internal Medicine, Seoul National University College of Medicine & Seoul National University Bundang Hospital, Seoul 03080, Republic of Korea
| | - Jong Kyoung Kim
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea.
| | - Jae Bum Kim
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
21
|
CD40L-expressing CD4+ T cells prime adipose-derived stromal cells to produce inflammatory chemokines. Cytotherapy 2022; 24:500-507. [DOI: 10.1016/j.jcyt.2022.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/21/2021] [Accepted: 01/20/2022] [Indexed: 11/18/2022]
|
22
|
Makris S, de Winde CM, Horsnell HL, Cantoral-Rebordinos JA, Finlay RE, Acton SE. Immune function and dysfunction are determined by lymphoid tissue efficacy. Dis Model Mech 2022; 15:dmm049256. [PMID: 35072206 PMCID: PMC8807573 DOI: 10.1242/dmm.049256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Lymphoid tissue returns to a steady state once each immune response is resolved, and although this occurs multiple times throughout life, its structural integrity and functionality remain unaffected. Stromal cells orchestrate cellular interactions within lymphoid tissue, and any changes to the microenvironment can have detrimental outcomes and drive disease. A breakdown in lymphoid tissue homeostasis can lead to a loss of tissue structure and function that can cause aberrant immune responses. This Review highlights recent advances in our understanding of lymphoid tissue function and remodelling in adaptive immunity and in disease states. We discuss the functional role of lymphoid tissue in disease progression and explore the changes to lymphoid tissue structure and function driven by infection, chronic inflammatory conditions and cancer. Understanding the role of lymphoid tissues in immune responses to a wide range of pathologies allows us to take a fuller systemic view of disease progression.
Collapse
Affiliation(s)
- Spyridon Makris
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Charlotte M. de Winde
- Department for Molecular Cell Biology and Immunology, Amsterdam UMC, location VUmc, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands
| | - Harry L. Horsnell
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Jesús A. Cantoral-Rebordinos
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Rachel E. Finlay
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester M13 9PL, UK
| | - Sophie E. Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
23
|
Nakagawa M, Hayashi S, Matsuo S, Yamazaki M, Kato A. Lipomatosis of axillary lymph nodes in a cynomolgus monkey ( Macaca fascicularis). J Toxicol Pathol 2022; 35:113-116. [PMID: 35221504 PMCID: PMC8828608 DOI: 10.1293/tox.2021-0054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/28/2021] [Indexed: 12/01/2022] Open
Abstract
Lipomatosis of lymph nodes is defined as the replacement of the lymphatic parenchyma by
adipose tissue which grows in the node from the hilus toward the cortical zone. In humans,
it is considered as part of the normal aging process and is common in obese patients, but
there are no reports in non-human primates. In this report, we describe the first case of
lymph node lipomatosis in the bilateral axillary lymph nodes of a young adult cynomolgus
monkey. Macroscopically, there were no apparent abnormalities in the axillary lymph nodes
on either side, and their volumes were unchanged. At the cut surface, pale yellow fat-like
tissue was observed in the medullary area. Histopathologically, well differentiated
adipocytes replaced a large part of the lymphatic parenchyma in the area from the hilus to
the medulla without any malignant findings. Based on these findings, the patient was
diagnosed with lipomatosis of the lymph nodes.
Collapse
Affiliation(s)
- Minto Nakagawa
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Shuji Hayashi
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Saori Matsuo
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Masaki Yamazaki
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Atsuhiko Kato
- Translational Research Division, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| |
Collapse
|
24
|
Soliman H, Theret M, Scott W, Hill L, Underhill TM, Hinz B, Rossi FMV. Multipotent stromal cells: One name, multiple identities. Cell Stem Cell 2021; 28:1690-1707. [PMID: 34624231 DOI: 10.1016/j.stem.2021.09.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Multipotent stromal cells (MSCs) are vital for development, maintenance, function, and regeneration of most tissues. They can differentiate along multiple connective lineages, but unlike most other stem/progenitor cells, they carry out various other functions while maintaining their developmental potential. MSCs function as damage sensors, respond to injury by fostering regeneration through secretion of trophic factors as well as extracellular matrix (ECM) molecules, and contribute to fibrotic reparative processes when regeneration fails. Tissue-specific MSC identity, fate(s), and function(s) are being resolved through fate mapping coupled with single cell "omics," providing unparalleled insights into the secret lives of tissue-resident MSCs.
Collapse
Affiliation(s)
- Hesham Soliman
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Aspect Biosystems, Vancouver, BC V6P 6P2, Canada
| | - Marine Theret
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Wilder Scott
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Lesley Hill
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Tully Michael Underhill
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Fabio M V Rossi
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
25
|
Poirot J, Medvedovic J, Trichot C, Soumelis V. Compartmentalized multicellular crosstalk in lymph nodes coordinates the generation of potent cellular and humoral immune responses. Eur J Immunol 2021; 51:3146-3160. [PMID: 34606627 PMCID: PMC9298410 DOI: 10.1002/eji.202048977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/13/2021] [Accepted: 09/22/2021] [Indexed: 12/24/2022]
Abstract
Distributed throughout the body, lymph nodes (LNs) constitute an important crossroad where resident and migratory immune cells interact to initiate antigen‐specific immune responses supported by a dynamic 3‐dimensional network of stromal cells, that is, endothelial cells and fibroblastic reticular cells (FRCs). LNs are organized into four major subanatomically separated compartments: the subcapsular sinus (SSC), the paracortex, the cortex, and the medulla. Each compartment is underpinned by particular FRC subsets that physically support LN architecture and delineate functional immune niches by appropriately providing environmental cues, nutrients, and survival factors to the immune cell subsets they interact with. In this review, we discuss how FRCs drive the structural and functional organization of each compartment to give rise to prosperous interactions and coordinate immune cell activities. We also discuss how reciprocal communication makes FRCs and immune cells perfect compatible partners for the generation of potent cellular and humoral immune responses.
Collapse
Affiliation(s)
- Justine Poirot
- Université de Paris, INSERM U976, Paris, France.,Université Paris-Saclay, Saint Aubin, France
| | | | | | - Vassili Soumelis
- Université de Paris, INSERM U976, Paris, France.,AP-HP, Hôpital Saint-Louis, Laboratoire d'Immunologie-Histocompatibilité, Paris, France
| |
Collapse
|
26
|
Lamaison C, Tarte K. B cell/stromal cell crosstalk in health, disease, and treatment: Follicular lymphoma as a paradigm. Immunol Rev 2021; 302:273-285. [PMID: 34060097 DOI: 10.1111/imr.12983] [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: 04/16/2021] [Revised: 05/09/2021] [Accepted: 05/10/2021] [Indexed: 12/16/2022]
Abstract
Stromal cells organize specific anatomic compartments within bone marrow (BM) and secondary lymphoid organs where they finely regulate the behavior of mature normal B cells. In particular, lymphoid stromal cells (LSCs) form a phenotypically heterogeneous compartment including various cell subsets variably supporting B-cell survival, activation, proliferation, and differentiation. In turn, activated B cells trigger in-depth remodeling of LSC networks within lymph nodes (LN) and BM. Follicular lymphoma (FL) is one of the best paradigms of a B-cell neoplasia depending on a specific tumor microenvironment (TME), including cancer-associated fibroblasts (CAFs) emerging from the reprogramming of LN LSCs or poorly characterized local BM precursors. FL-CAFs support directly malignant B-cell growth and orchestrate FL permissive cell niche by contributing, through a bidirectional crosstalk, to the recruitment and polarization of immune TME subsets. Recent studies have highlighted a previously unexpected level of heterogeneity of both FL B cells and FL TME, underlined by FL-CAF plasticity. A better understanding of the signaling pathways, molecular mechanisms, and kinetic of stromal cell remodeling in FL would be useful to delineate new predictive markers and new therapeutic approaches in this still fatal malignancy.
Collapse
Affiliation(s)
- Claire Lamaison
- UMR_S 1236, Université Rennes 1, INSERM, Etablissement Français du Sang, Rennes, France
| | - Karin Tarte
- UMR_S 1236, Université Rennes 1, INSERM, Etablissement Français du Sang, Rennes, France.,SITI, Pôle de Biologie, CHU Pontchaillou, Rennes, France
| |
Collapse
|
27
|
Lütge M, Pikor NB, Ludewig B. Differentiation and activation of fibroblastic reticular cells. Immunol Rev 2021; 302:32-46. [PMID: 34046914 PMCID: PMC8361914 DOI: 10.1111/imr.12981] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/17/2021] [Accepted: 04/30/2021] [Indexed: 12/29/2022]
Abstract
Secondary lymphoid organs (SLO) are underpinned by fibroblastic reticular cells (FRC) that form dedicated microenvironmental niches to secure induction and regulation of innate and adaptive immunity. Distinct FRC subsets are strategically positioned in SLOs to provide niche factors and govern efficient immune cell interaction. In recent years, the use of specialized mouse models in combination with single-cell transcriptomics has facilitated the elaboration of the molecular FRC landscape at an unprecedented resolution. While single-cell RNA-sequencing has advanced the resolution of FRC subset characterization and function, the high dimensionality of the generated data necessitates careful analysis and validation. Here, we reviewed novel findings from high-resolution transcriptomic analyses that refine our understanding of FRC differentiation and activation processes in the context of infection and inflammation. We further discuss concepts, strategies, and limitations for the analysis of single-cell transcriptome data from FRCs and the wide-ranging implications for our understanding of stromal cell biology.
Collapse
Affiliation(s)
- Mechthild Lütge
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Natalia B Pikor
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
28
|
The Tumor Microenvironment in Follicular Lymphoma: Its Pro-Malignancy Role with Therapeutic Potential. Int J Mol Sci 2021; 22:ijms22105352. [PMID: 34069564 PMCID: PMC8160856 DOI: 10.3390/ijms22105352] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023] Open
Abstract
In the follicular lymphoma (FL) microenvironment, CXCR5+ICOS+PD1+BCL6+ follicular helper T (Tfh) cells, which closely correlate with FL B cells in neoplastic follicles, play a major role in supporting FL. Interleukin-4 secreted by Tfh cells triggers the upregulation of the lymphocyte chemoattractant CXCL12 in stromal cell precursors, in particular by fibroblastic reticular cells (FRCs). In turn, mesenchymal stem cells (MSCs) can be committed to FRC differentiation in the bone marrow and lymph nodes involved by FL. Noteworthy, MSCs can promote the differentiation of Tfh cells into highly immunosuppressive T-follicular regulatory cells. The tumor suppressor HVEM is highly mutated in FL cells, and its deficiency increases Tfh cell frequency. In contrast, PI3Kδ inhibition impedes the recruitment of Tfh/regulatory T cells and impairs the proliferation of follicular dendritic cells (FDCs) and FDC-induced angiogenesis. Since TIGIT ligands are expressed by FDCs, the immune checkpoint receptor TIGIT plays an important role in tumor-infiltrating T cells. Thus, TIGIT blockade might invigorate cytotoxic T cells in the FL microenvironment. Given their potential to simultaneously reduce the neoplastic B cells, Tfh, and TFR cells could also reinforce the effects of the cytotoxic T cells. This combinatory strategy should be explored as a treatment option to tackle FL.
Collapse
|
29
|
Jimenez MT, Michieletto MF, Henao-Mejia J. A new perspective on mesenchymal-immune interactions in adipose tissue. Trends Immunol 2021; 42:375-388. [PMID: 33849777 DOI: 10.1016/j.it.2021.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/11/2022]
Abstract
The mammalian immune system has crucial homeostatic functions in different adipose depots. However, white adipose tissue (WAT) inflammation is a hallmark of obesity and can contribute to type 2 diabetes mellitus (T2DM). Recently, mesenchymal cells were identified as highly heterogenous populations displaying specialized immune functions in immune cell migration, activation, survival, and overall lymphoid tissue organization in several tissues. How they regulate the inflammatory milieu within different adipose depots remains unknown. Using recently published single-cell RNA-sequencing (scRNAseq) data sets, we analyze cytokine and chemokine expression of mouse WAT mesenchymal cell subpopulations to highlight potential immunological heterogeneity and specialization, hypothesizing on their immunological functions. This new perspective on immune-mesenchymal cell interactions in adipose tissue may promote studies that heighten our understanding of immune cell processes within WAT during health and obesity. We hope that these studies redefine our knowledge of the roles of mesenchymal cells in regulating adipose tissue inflammation and physiology.
Collapse
Affiliation(s)
- Monica T Jimenez
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michaël F Michieletto
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jorge Henao-Mejia
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Protective Immunity, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
30
|
Adipose stem cells in obesity: challenges and opportunities. Biosci Rep 2021; 40:225001. [PMID: 32452515 PMCID: PMC7284323 DOI: 10.1042/bsr20194076] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/08/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue, the storage of excessive energy in the body, secretes various proteins called adipokines, which connect the body’s nutritional status to the regulation of energy balance. Obesity triggers alterations of quantity and quality of various types of cells that reside in adipose tissue, including adipose stem cells (ASCs; referred to as adipose-derived stem/stromal cells in vitro). These alterations in the functionalities and properties of ASCs impair adipose tissue remodeling and adipose tissue function, which induces low-grade systemic inflammation, progressive insulin resistance, and other metabolic disorders. In contrast, the ability of ASCs to recruit new adipocytes when faced with caloric excess leads to healthy adipose tissue expansion, associated with lower amounts of inflammation, fibrosis, and insulin resistance. This review focuses on recent advances in our understanding of the identity of ASCs and their roles in adipose tissue development, homeostasis, expansion, and thermogenesis, and how these roles go awry in obesity. A better understanding of the biology of ASCs and their adipogenesis may lead to novel therapeutic targets for obesity and metabolic disease.
Collapse
|
31
|
Kuper CF, Pieters RHH, van Bilsen JHM. Nanomaterials and the Serosal Immune System in the Thoracic and Peritoneal Cavities. Int J Mol Sci 2021; 22:ijms22052610. [PMID: 33807632 PMCID: PMC7961545 DOI: 10.3390/ijms22052610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/23/2021] [Accepted: 02/27/2021] [Indexed: 11/16/2022] Open
Abstract
The thoracic and peritoneal cavities are lined by serous membranes and are home of the serosal immune system. This immune system fuses innate and adaptive immunity, to maintain local homeostasis and repair local tissue damage, and to cooperate closely with the mucosal immune system. Innate lymphoid cells (ILCs) are found abundantly in the thoracic and peritoneal cavities, and they are crucial in first defense against pathogenic viruses and bacteria. Nanomaterials (NMs) can enter the cavities intentionally for medical purposes, or unintentionally following environmental exposure; subsequent serosal inflammation and cancer (mesothelioma) has gained significant interest. However, reports on adverse effects of NM on ILCs and other components of the serosal immune system are scarce or even lacking. As ILCs are crucial in the first defense against pathogenic viruses and bacteria, it is possible that serosal exposure to NM may lead to a reduced resistance against pathogens. Additionally, affected serosal lymphoid tissues and cells may disturb adipose tissue homeostasis. This review aims to provide insight into key effects of NM on the serosal immune system.
Collapse
Affiliation(s)
- C. Frieke Kuper
- Consultant, Haagstraat 13, 3581 SW Utrecht, The Netherlands
- Correspondence: (C.F.K.); (J.H.M.v.B.)
| | - Raymond H. H. Pieters
- Immunotoxicology, Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands;
- Innovative Testing in Life Sciences & Chemistry, Research Centre for Healthy and Sustainable Living, University of Applied Sciences Utrecht, Padualaan 97, 3584 CH Utrecht, The Netherlands
| | - Jolanda H. M. van Bilsen
- Department for Risk Analysis for Products in Development, Netherlands Organization for Applied Scientific Research (TNO), Princetonlaan 6, 3584 CB Utrecht, The Netherlands
- Correspondence: (C.F.K.); (J.H.M.v.B.)
| |
Collapse
|
32
|
Foxall R, Narang P, Glaysher B, Hub E, Teal E, Coles MC, Ashton-Key M, Beers SA, Cragg MS. Developing a 3D B Cell Lymphoma Culture System to Model Antibody Therapy. Front Immunol 2021; 11:605231. [PMID: 33628205 PMCID: PMC7897703 DOI: 10.3389/fimmu.2020.605231] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022] Open
Abstract
Diffuse large cell B cell lymphoma (DLBCL) accounts for approximately 30%-40% of all non-Hodgkin lymphoma (NHL) cases. Current first line DLBCL treatment results in long-term remission in more than 60% of cases. However, those patients with primary refractory disease or early relapse exhibit poor prognosis, highlighting a requirement for alternative therapies. Our aim was to develop a novel model of DLBCL that facilitates in vitro testing of current and novel therapies by replicating key components of the tumor microenvironment (TME) in a three-dimensional (3D) culture system that would enable primary DLBCL cell survival and study ex vivo. The TME is a complex ecosystem, comprising malignant and non-malignant cells, including cancer-associated fibroblasts (CAF) and tumor-associated macrophages (TAM) whose reciprocal crosstalk drives tumor initiation and growth while fostering an immunosuppressive milieu enabling its persistence. The requirement to recapitulate, at least to some degree, this complex, interactive network is exemplified by the rapid cell death of primary DLBCL cells removed from their TME and cultured alone in vitro. Building on previously described methodologies to generate lymphoid-like fibroblasts from adipocyte derived stem cells (ADSC), we confirmed lymphocytes, specifically B cells, interacted with this ADSC-derived stroma, in the presence or absence of monocyte-derived macrophages (MDM), in both two-dimensional (2D) cultures and a 3D collagen-based spheroid system. Furthermore, we demonstrated that DLBCL cells cultured in this system interact with its constituent components, resulting in their improved viability as compared to ex-vivo 2D monocultures. We then assessed the utility of this system as a platform to study therapeutics in the context of antibody-directed phagocytosis, using rituximab as a model immunotherapeutic antibody. Overall, we describe a novel 3D spheroid co-culture system comprising key components of the DLBCL TME with the potential to serve as a testbed for novel therapeutics, targeting key cellular constituents of the TME, such as CAF and/or TAM.
Collapse
Affiliation(s)
- Russell Foxall
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
| | - Priyanka Narang
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
| | - Bridget Glaysher
- Centre for Immunology and Infection, University of York, York, United Kingdom
| | - Elin Hub
- Centre for Immunology and Infection, University of York, York, United Kingdom
| | - Emma Teal
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
| | - Mark C Coles
- Centre for Immunology and Infection, University of York, York, United Kingdom.,Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Margaret Ashton-Key
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom.,Department of Cellular Pathology, Southampton University Hospital Trust, Southampton, United Kingdom
| | - Stephen A Beers
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
| | - Mark S Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
| |
Collapse
|
33
|
Jia X, Berta G, Gábris F, Kellermayer Z, Balogh P. Role of adipose-associated lymphoid tissues in the immunological homeostasis of the serosal surface. Immunol Lett 2020; 228:135-141. [PMID: 33166529 DOI: 10.1016/j.imlet.2020.11.001] [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: 09/15/2020] [Revised: 10/13/2020] [Accepted: 11/03/2020] [Indexed: 11/30/2022]
Abstract
Although not typical lymphoid organs, analysis of the visceral adipose-associated lymphoid tissues has recently substantially expanded our knowledge about the immunological features of these elusive compartments. Recent data have highlighted their considerable complexity in cellular organization and interactions in several biological processes, including adaptive immune responses, tissue plasticity to accommodate mesenchymal stem cells and progenitors, and providing a suitable microenvironment for serosal tumor propagation. This review aims to present a comprehensive view of the adipose-associated lymphoid tissues in local and systemic immune responsiveness, with particular emphasis on the omental and mesenteric lymphoid tissues in the serosal defense of abdominal organs.
Collapse
Affiliation(s)
- Xinkai Jia
- Department of Immunology and Biotechnology, Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary; Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Hungary
| | - Fanni Gábris
- Department of Immunology and Biotechnology, Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary; Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary
| | - Zoltán Kellermayer
- Department of Immunology and Biotechnology, Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary; Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary
| | - Péter Balogh
- Department of Immunology and Biotechnology, Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary; Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary.
| |
Collapse
|
34
|
Li X, Zhao J, Kasinath V, Uehara M, Jiang L, Banouni N, McGrath MM, Ichimura T, Fiorina P, Lemos DR, Shin SR, Ware CF, Bromberg JS, Abdi R. Lymph node fibroblastic reticular cells deposit fibrosis-associated collagen following organ transplantation. J Clin Invest 2020; 130:4182-4194. [PMID: 32597832 PMCID: PMC7410068 DOI: 10.1172/jci136618] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/22/2020] [Indexed: 02/05/2023] Open
Abstract
Although the immune response within draining lymph nodes (DLNs) has been studied for decades, how their stromal compartment contributes to this process remains to be fully explored. Here, we show that donor mast cells were prominent activators of collagen I deposition by fibroblastic reticular cells (FRCs) in DLNs shortly following transplantation. Serial analysis of the DLN indicated that the LN stroma did not return to its baseline microarchitecture following organ rejection and that the DLN contained significant fibrosis following repetitive organ transplants. Using several FRC conditional-knockout mice, we show that induction of senescence in the FRCs of the DLN resulted in massive production of collagen I and a proinflammatory milieu within the DLN. Stimulation of herpes virus entry mediator (HVEM) on FRCs by its ligand LIGHT contributed chiefly to the induction of senescence in FRCs and overproduction of collagen I. Systemic administration of ex vivo-expanded FRCs to mice decreased DLN fibrosis and strengthened the effect of anti-CD40L in prolonging heart allograft survival. These data demonstrate that the transformation of FRCs into proinflammatory myofibroblasts is critically important for the maintenance of a proinflammatory milieu within a fibrotic DLN.
Collapse
Affiliation(s)
- Xiaofei Li
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University School of Pharmaceutical Sciences, Wuhan, Hubei, China
| | - Jing Zhao
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Vivek Kasinath
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mayuko Uehara
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Liwei Jiang
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Naima Banouni
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Martina M. McGrath
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University School of Pharmaceutical Sciences, Wuhan, Hubei, China
| | | | - Paolo Fiorina
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dario R. Lemos
- Renal Division, Brigham and Women’s Hospital
- Harvard Stem Cell Institute, and
| | - Su Ryon Shin
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carl F. Ware
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Jonathan S. Bromberg
- Department of Surgery and Microbiology and Immunobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Najibi AJ, Mooney DJ. Cell and tissue engineering in lymph nodes for cancer immunotherapy. Adv Drug Deliv Rev 2020; 161-162:42-62. [PMID: 32750376 PMCID: PMC7736208 DOI: 10.1016/j.addr.2020.07.023] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/03/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
Abstract
In cancer, lymph nodes (LNs) coordinate tumor antigen presentation necessary for effective antitumor immunity, both at the levels of local cellular interactions and tissue-level organization. In this review, we examine how LNs may be engineered to improve the therapeutic outcomes of cancer immunotherapy. At the cellular scale, targeting the LNs impacts the potency of cancer vaccines, immune checkpoint blockade, and adoptive cell transfer. On a tissue level, macro-scale biomaterials mimicking LN features can function as immune niches for cell reprogramming or delivery in vivo, or be utilized in vitro to enable preclinical testing of drugs and vaccines. We additionally review strategies to induce ectopic lymphoid sites reminiscent of LNs that may improve antitumor T cell priming.
Collapse
Affiliation(s)
- Alexander J Najibi
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138.
| |
Collapse
|
36
|
Eom J, Park SM, Feisst V, Chen CJJ, Mathy JE, McIntosh JD, Angel CE, Bartlett A, Martin R, Mathy JA, Cebon JS, Black MA, Brooks AES, Dunbar PR. Distinctive Subpopulations of Stromal Cells Are Present in Human Lymph Nodes Infiltrated with Melanoma. Cancer Immunol Res 2020; 8:990-1003. [PMID: 32580941 DOI: 10.1158/2326-6066.cir-19-0796] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 03/22/2020] [Accepted: 06/19/2020] [Indexed: 11/16/2022]
Abstract
Metastasis of human tumors to lymph nodes (LN) is a universally negative prognostic factor. LN stromal cells (SC) play a crucial role in enabling T-cell responses, and because tumor metastases modulate their structure and function, this interaction may suppress immune responses to tumor antigens. The SC subpopulations that respond to infiltration of malignant cells into human LNs have not been defined. Here, we identify distinctive subpopulations of CD90+ SCs present in melanoma-infiltrated LNs and compare them with their counterparts in normal LNs. The first population (CD90+ podoplanin+ CD105+ CD146+ CD271+ VCAM-1+ ICAM-1+ α-SMA+) corresponds to fibroblastic reticular cells that express various T-cell modulating cytokines, chemokines, and adhesion molecules. The second (CD90+ CD34+ CD105+ CD271+) represents a novel population of CD34+ SCs embedded in collagenous structures, such as the capsule and trabeculae, that predominantly produce extracellular matrix. We also demonstrated that these two SC subpopulations are distinct from two subsets of human LN pericytes, CD90+ CD146+ CD36+ NG2- pericytes in the walls of high endothelial venules and other small vessels, and CD90+ CD146+ NG2+ CD36- pericytes in the walls of larger vessels. Distinguishing between these CD90+ SC subpopulations in human LNs allows for further study of their respective impact on T-cell responses to tumor antigens and clinical outcomes.
Collapse
Affiliation(s)
- Jennifer Eom
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Saem Mul Park
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Vaughan Feisst
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Chun-Jen J Chen
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Joanna E Mathy
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Julie D McIntosh
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Catherine E Angel
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Adam Bartlett
- Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand.,Department of Surgery, Faculty of Medical Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard Martin
- Department of Surgery, Waitemata District Health Board, Auckland, New Zealand
| | - Jon A Mathy
- Department of Surgery, Faculty of Medical Health Sciences, University of Auckland, Auckland, New Zealand.,Auckland Regional Plastic, Reconstructive & Hand Surgery Unit, Auckland, New Zealand
| | - Jonathan S Cebon
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
| | - Michael A Black
- Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand.,Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Anna E S Brooks
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - P Rod Dunbar
- School of Biological Sciences, University of Auckland, Auckland, New Zealand. .,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| |
Collapse
|
37
|
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: 10] [Impact Index Per Article: 2.0] [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
|
38
|
Jackson-Jones LH, Bénézech C. FALC stromal cells define a unique immunological niche for the surveillance of serous cavities. Curr Opin Immunol 2020; 64:42-49. [PMID: 32353646 DOI: 10.1016/j.coi.2020.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 03/04/2020] [Accepted: 03/10/2020] [Indexed: 01/14/2023]
Abstract
The serous cavities contain specialised adipose tissues which house small clusters of immune cells known as fat-associated lymphoid clusters (FALCs). The continuous flow of fluid from the serous cavities through FALCs makes them unique niches for the clearance of fluid phase contaminants and initiation of locally protective immune responses during infection and inflammation. Development, and activation of FALCs both at homeostasis and following inflammation are co-ordinated by the close interaction of mesothelial and fibroblastic stromal cell populations with immune cells. In this review we discuss recent developments in FALC stromal cell biology and highlight key interactions that occur between FALC stroma and immune cells.
Collapse
Affiliation(s)
- Lucy H Jackson-Jones
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster LA1 4YQ, UK
| | - Cécile Bénézech
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
39
|
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: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/17/2020] [Indexed: 01/03/2023]
Abstract
Lymph nodes (LNs) are strategically positioned at dedicated sites throughout the body to facilitate rapid and efficient immunity. Central to the structural integrity and framework of LNs, and the recruitment and positioning of leukocytes therein, are mesenchymal and endothelial lymph node stromal cells (LNSCs). Advances in the last decade have expanded our understanding and appreciation of LNSC heterogeneity, and the role they play in coordinating immunity has grown rapidly. In this review, we will highlight the functional contributions of distinct stromal cell populations during LN development in maintaining immune homeostasis and tolerance and in the activation and control of immune responses.
Collapse
|
40
|
YAP/TAZ direct commitment and maturation of lymph node fibroblastic reticular cells. Nat Commun 2020; 11:519. [PMID: 31980640 PMCID: PMC6981200 DOI: 10.1038/s41467-020-14293-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 12/31/2019] [Indexed: 02/07/2023] Open
Abstract
Fibroblastic reticular cells (FRCs) are immunologically specialized myofibroblasts of lymphoid organ, and FRC maturation is essential for structural and functional properties of lymph nodes (LNs). Here we show that YAP and TAZ (YAP/TAZ), the final effectors of Hippo signaling, regulate FRC commitment and maturation. Selective depletion of YAP/TAZ in FRCs impairs FRC growth and differentiation and compromises the structural organization of LNs, whereas hyperactivation of YAP/TAZ enhances myofibroblastic characteristics of FRCs and aggravates LN fibrosis. Mechanistically, the interaction between YAP/TAZ and p52 promotes chemokine expression that is required for commitment of FRC lineage prior to lymphotoxin-β receptor (LTβR) engagement, whereas LTβR activation suppresses YAP/TAZ activity for FRC maturation. Our findings thus present YAP/TAZ as critical regulators of commitment and maturation of FRCs, and hold promise for better understanding of FRC-mediated pathophysiologic processes. Fibroblastic reticular cells (FRC) are important for lymph node (LN) structure and function. Here the authors show that the YAP/TAZ complex downstream of Hippo signalling regulates FRC commitment and maturation, with YAP/TAZ deficiency impairing FRC differentiation, while hyperactivation of YAZ/TAZ inducing myofibroblastic FRCs and LN fibrosis.
Collapse
|
41
|
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: 2.0] [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
|
42
|
Saxena V, Li L, Paluskievicz C, Kasinath V, Bean A, Abdi R, Jewell CM, Bromberg JS. Role of lymph node stroma and microenvironment in T cell tolerance. Immunol Rev 2019; 292:9-23. [PMID: 31538349 PMCID: PMC6935411 DOI: 10.1111/imr.12799] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/22/2019] [Indexed: 12/12/2022]
Abstract
Lymph nodes (LNs) are at the cross roads of immunity and tolerance. These tissues are compartmentalized into specialized niche areas by lymph node stromal cells (LN SCs). LN SCs shape the LN microenvironment and guide immunological cells into different zones through establishment of a CCL19 and CCL21 gradient. Following local immunological cues, LN SCs modulate activity to support immune cell priming, activation, and fate. This review will present our current understanding of LN SC subsets roles in regulating T cell tolerance. Three major types of LN SC subsets, namely fibroblastic reticular cells, lymphatic endothelial cells, and blood endothelial cells, are discussed. These subsets serve as scaffolds to support and regulate T cell homeostasis. They contribute to tolerance by presenting peripheral tissue antigens to both CD4 and CD8 T cells. The role of LN SCs in regulating T cell migration and tolerance induction is discussed. Looking forward, recent advances in bioengineered materials and approaches to leverage LN SCs to induce T cell tolerance are highlighted, as are current clinical practices that allow for manipulation of the LN microenvironment to induce tolerance. Increased understanding of LN architecture, how different LN SCs integrate immunological cues and shape immune responses, and approaches to induce T cell tolerance will help further combat autoimmune diseases and graft rejection.
Collapse
Affiliation(s)
- Vikas Saxena
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Lushen Li
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Christina Paluskievicz
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Vivek Kasinath
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Asher Bean
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Reza Abdi
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher M. Jewell
- Fischell Department of Bioengineering, Robert E. Fischell Institute for Biomedical Devices University of Maryland, College Park, MD 20742, USA
- United States Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD 21201, USA
| | - Jonathan S. Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
43
|
Lamaison C, Tarte K. Impact of B cell/lymphoid stromal cell crosstalk in B-cell physiology and malignancy. Immunol Lett 2019; 215:12-18. [DOI: 10.1016/j.imlet.2019.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/25/2019] [Accepted: 02/26/2019] [Indexed: 12/17/2022]
|
44
|
Ménard C, Dulong J, Roulois D, Hébraud B, Verdière L, Pangault C, Sibut V, Bezier I, Bescher N, Monvoisin C, Gadelorge M, Bertheuil N, Flécher E, Casteilla L, Collas P, Sensebé L, Bourin P, Espagnolle N, Tarte K. Integrated transcriptomic, phenotypic, and functional study reveals tissue-specific immune properties of mesenchymal stromal cells. Stem Cells 2019; 38:146-159. [PMID: 31502731 DOI: 10.1002/stem.3077] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/08/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022]
Abstract
Clinical-grade mesenchymal stromal cells (MSCs) can be expanded from bone marrow and adipose tissue to treat inflammatory diseases and degenerative disorders. However, the influence of their tissue of origin on their functional properties, including their immunosuppressive activity, remains unsolved. In this study, we produced paired bone marrow-derived mesenchymal stromal cell (BM-MSC) and adipose-derived stromal cell (ASC) batches from 14 healthy donors. We then compared them using transcriptomic, phenotypic, and functional analyses and validated our results on purified native MSCs to infer which differences were really endowed by tissue of origin. Cultured MSCs segregated together owing to their tissue of origin based on their gene expression profile analyzed using differential expression and weighted gene coexpression network analysis. This translated into distinct immune-related gene signatures, phenotypes, and functional cell interactions. Importantly, sorted native BM-MSCs and ASCs essentially displayed the same distinctive patterns than their in vitro-expanded counterparts. As a whole, ASCs exhibited an immune profile consistent with a stronger inhibition of immune response and a lower immunogenicity, supporting the use of adipose tissue as a valuable source for clinical applications.
Collapse
Affiliation(s)
- Cédric Ménard
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France
| | - Joëlle Dulong
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France
| | - David Roulois
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Benjamin Hébraud
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Léa Verdière
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Céline Pangault
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,Pôle Biologie, CHU Rennes, Rennes, France
| | - Vonick Sibut
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France
| | - Isabelle Bezier
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France
| | - Nadège Bescher
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France
| | - Céline Monvoisin
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Mélanie Gadelorge
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Nicolas Bertheuil
- SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France.,Department of Plastic Surgery, CHU Rennes, Rennes, France
| | - Erwan Flécher
- Department of Thoracic and Cardiac Surgery, CHU Rennes, Rennes, France
| | - Louis Casteilla
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Philippe Collas
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Luc Sensebé
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | | | - Nicolas Espagnolle
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Karin Tarte
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France
| |
Collapse
|
45
|
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: 2.5] [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
|
46
|
IL-4Rα-Expressing B Cells Are Required for CXCL13 Production by Fibroblastic Reticular Cells. Cell Rep 2019; 27:2442-2458.e5. [DOI: 10.1016/j.celrep.2019.04.079] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 01/14/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022] Open
|
47
|
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: 10.7] [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
|
48
|
Barron AMS, Mantero JC, Ho JD, Nazari B, Horback KL, Bhawan J, Lafyatis R, Lam C, Browning JL. Perivascular Adventitial Fibroblast Specialization Accompanies T Cell Retention in the Inflamed Human Dermis. THE JOURNAL OF IMMUNOLOGY 2018; 202:56-68. [PMID: 30510068 DOI: 10.4049/jimmunol.1801209] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Perivascular accumulation of lymphocytes can be a prominent histopathologic feature of various human inflammatory skin diseases. Select examples include systemic sclerosis, spongiotic dermatitis, and cutaneous lupus. Although a large body of work has described various aspects of the endothelial and vascular smooth muscle layers in these diseases, the outer adventitial compartment is poorly explored. The goal of the current study was to characterize perivascular adventitial fibroblast states in inflammatory human skin diseases and relate these states to perivascular lymphocyte accumulation. In normal skin, adventitial fibroblasts are distinguished by CD90 expression, and dense perivascular lymphocytic infiltrates are uncommon. In systemic sclerosis, this compartment expands, but lymphocyte infiltrates remain sparse. In contrast, perivascular adventitial fibroblast expression of VCAM1 is upregulated in spongiotic dermatitis and lupus and is associated with a dense perivascular T cell infiltrate. VCAM1 expression marks transitioned fibroblasts that show some resemblance to the reticular stromal cells in secondary lymphoid organs. Expanded adventitial compartments with perivascular infiltrates similar to the human settings were not seen in the inflamed murine dermis. This species difference may hinder the dissection of aspects of perivascular adventitial pathology. The altered perivascular adventitial compartment and its associated reticular network form a niche for lymphocytes and appear to be fundamental in the development of an inflammatory pattern.
Collapse
Affiliation(s)
- Alexander M S Barron
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| | - Julio C Mantero
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| | - Jonathan D Ho
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Banafsheh Nazari
- Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118
| | - Katharine L Horback
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| | - Jag Bhawan
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Robert Lafyatis
- Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118.,Division of Rheumatology and Clinical Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213
| | - Christina Lam
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Jeffrey L Browning
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118; .,Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118
| |
Collapse
|
49
|
Onder L, Ludewig B. A Fresh View on Lymph Node Organogenesis. Trends Immunol 2018; 39:775-787. [DOI: 10.1016/j.it.2018.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/07/2018] [Accepted: 08/07/2018] [Indexed: 01/18/2023]
|
50
|
Kou Y, Liu Q, Liu W, Sun H, Liang M, Kong F, Zhang B, Wei Y, Liu Z, Wang Y. LIGHT/TNFSF14 signaling attenuates beige fat biogenesis. FASEB J 2018; 33:1595-1604. [DOI: 10.1096/fj.201800792r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Yanbo Kou
- Jiangsu Key Laboratory of Immunity and MetabolismXuzhou Medical UniversityXuzhouChina
| | - Qingya Liu
- Jiangsu Key Laboratory of Immunity and MetabolismXuzhou Medical UniversityXuzhouChina
- Laboratory of Infection and ImmunityDepartment of Pathogenic Biology and ImmunologyXuzhou Medical UniversityXuzhouChina
| | - Wenli Liu
- Jiangsu Key Laboratory of Immunity and MetabolismXuzhou Medical UniversityXuzhouChina
- Laboratory of Infection and ImmunityDepartment of Pathogenic Biology and ImmunologyXuzhou Medical UniversityXuzhouChina
| | - Hongxiang Sun
- Jiangsu Key Laboratory of Immunity and MetabolismXuzhou Medical UniversityXuzhouChina
- Laboratory of Infection and ImmunityDepartment of Pathogenic Biology and ImmunologyXuzhou Medical UniversityXuzhouChina
| | - Ming Liang
- Jiangsu Key Laboratory of Immunity and MetabolismXuzhou Medical UniversityXuzhouChina
- Laboratory of Infection and ImmunityDepartment of Pathogenic Biology and ImmunologyXuzhou Medical UniversityXuzhouChina
| | - Fanyun Kong
- Jiangsu Key Laboratory of Immunity and MetabolismXuzhou Medical UniversityXuzhouChina
- Laboratory of Infection and ImmunityDepartment of Pathogenic Biology and ImmunologyXuzhou Medical UniversityXuzhouChina
| | - Bo Zhang
- Jiangsu Key Laboratory of Immunity and MetabolismXuzhou Medical UniversityXuzhouChina
- Laboratory of Infection and ImmunityDepartment of Pathogenic Biology and ImmunologyXuzhou Medical UniversityXuzhouChina
| | - Yanxia Wei
- Jiangsu Key Laboratory of Immunity and MetabolismXuzhou Medical UniversityXuzhouChina
- Laboratory of Infection and ImmunityDepartment of Pathogenic Biology and ImmunologyXuzhou Medical UniversityXuzhouChina
| | - Zhuanzhuan Liu
- Jiangsu Key Laboratory of Immunity and MetabolismXuzhou Medical UniversityXuzhouChina
- Laboratory of Infection and ImmunityDepartment of Pathogenic Biology and ImmunologyXuzhou Medical UniversityXuzhouChina
| | - Yugang Wang
- Jiangsu Key Laboratory of Immunity and MetabolismXuzhou Medical UniversityXuzhouChina
- Laboratory of Infection and ImmunityDepartment of Pathogenic Biology and ImmunologyXuzhou Medical UniversityXuzhouChina
| |
Collapse
|