1
|
Oñate MK, Oon C, Bhattacharyya S, Low V, Chen C, Zhao X, Arnold F, Yan Z, Pramod S, Hang Y, Ho YJ, Lowe SW, Kim SK, Xia Z, Sherman MH. Stromal KITL/SCF Maintains Pancreas Tissue Homeostasis and Restrains Tumor Progression. Cancer Discov 2025; 15:913-929. [PMID: 39918337 PMCID: PMC12046321 DOI: 10.1158/2159-8290.cd-24-1079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/15/2024] [Accepted: 02/05/2025] [Indexed: 02/12/2025]
Abstract
SIGNIFICANCE By analyzing transcriptional programs in healthy and tumor-associated pancreatic mesenchyme, we find that a subpopulation of mesenchymal cells in healthy pancreas tissue expresses the paracrine signaling factor KITL. The loss of mesenchymal KITL is an accompanying and permissive feature of pancreas tumor evolution, with potential implications for cancer interception. See related article by Dolskii and Cukierman, p. 872.
Collapse
Affiliation(s)
- Maria Kathrina Oñate
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Chet Oon
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Sohinee Bhattacharyya
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Vivien Low
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Canping Chen
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Xiaofan Zhao
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Frank Arnold
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Ziqiao Yan
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
| | - Sneha Pramod
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Graduate School of Medical Science, Weill Cornell Medicine, New York, New York
| | - Yan Hang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, California
| | - Yu-Jui Ho
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Scott W. Lowe
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Seung K. Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, California
- Department of Medicine, Stanford University School of Medicine, Stanford, California
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Zheng Xia
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Mara H. Sherman
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
2
|
Yamamura Y, Sabiu G, Zhao J, Jung S, Seelam AJ, Li X, Song Y, Shirkey MW, Li L, Piao W, Wu L, Zhang T, Ahn S, Kim P, Kasinath V, Azzi JR, Bromberg JS, Abdi R. CXCL12+ fibroblastic reticular cells in lymph nodes facilitate immune tolerance by regulating T cell-mediated alloimmunity. J Clin Invest 2025; 135:e182709. [PMID: 40309773 PMCID: PMC12043101 DOI: 10.1172/jci182709] [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: 05/07/2024] [Accepted: 02/27/2025] [Indexed: 05/02/2025] Open
Abstract
Fibroblastic reticular cells (FRCs) are the master regulators of the lymph node (LN) microenvironment. However, the role of specific FRC subsets in controlling alloimmune responses remains to be studied. Single-cell RNA sequencing (scRNA-Seq) of naive and draining LNs (DLNs) of heart-transplanted mice and human LNs revealed a specific subset of CXCL12hi FRCs that expressed high levels of lymphotoxin-β receptor (LTβR) and are enriched in the expression of immunoregulatory genes. CXCL12hi FRCs had high expression of CCL19, CCL21, indoleamine 2,3-dioxygenase (IDO), IL-10, and TGF-β1. Adoptive transfer of ex vivo-expanded FRCs resulted in their homing to LNs and induced immunosuppressive environments in DLNs to promote heart allograft acceptance. Genetic deletion of LTβR and Cxcl12 in FRCs increased alloreactivity, abrogating the effect of costimulatory blockade in prolonging heart allograft survival. As compared with WT recipients, CXCL12+ FRC-deficient recipients exhibited increased differentiation of CD4+ T cells into Th1 cells. Nano delivery of CXCL12 to DLNs improved allograft survival in heart-transplanted mice. Our study highlights the importance of DLN CXCL12hi FRCs in promoting transplant tolerance.
Collapse
Affiliation(s)
- Yuta Yamamura
- 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
| | - 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
| | - Andy J. Seelam
- 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
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Marina W. Shirkey
- Department of Surgery and
- Center of Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lushen Li
- Department of Surgery and
- Center of Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Wenji Piao
- Department of Surgery and
- Center of Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | - Soyeon Ahn
- R&D Division, IVIM Technology, Seoul, South Korea
| | - Pilhan Kim
- Graduate School of Medical Science and Engineering
- Korea Advanced Institute of Science and Technology Institute for Health Science and Technology, and
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Vivek Kasinath
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jamil R. Azzi
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan S. Bromberg
- Department of Surgery and
- Center of Vascular and Inflammatory Diseases, 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
|
3
|
Yasmin H, Ramesh RPG, Joseph AM, Kishore U. Fibroblast heterogeneity and its role in generating protective immunity in the secondary lymphoid organs. Front Immunol 2025; 16:1519789. [PMID: 40248708 PMCID: PMC12003126 DOI: 10.3389/fimmu.2025.1519789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/04/2025] [Indexed: 04/19/2025] Open
Abstract
Fibroblasts are cells of mesenchymal origin with a range of phenotypic diversity and heterogeneity. One of the major functions of fibroblasts is the formation and turnover of the extracellular matrix and establishing a tissue structure by forming a matrisome from embryonic development to the adult stage. It plays an indispensable role in extracellular matrix remodeling during injury, repair, and infection, providing a scaffold for cell-to-cell interaction. Despite their important pathophysiological roles, molecular markers for tissue-resident fibroblasts are only now being identified. Fibroblasts acquire molecular signatures based on anatomical locations, thus impacting their phenotypic heterogeneity despite their overlapping morphology. Fibroblasts are now recognized as key immune sentinel cells, capable of regulating the inflammatory milieu through their distinct functional subsets that are designed to respond differently with unique immune signatures. Fibroblasts can detect pathogenic and danger signals through their diverse pattern recognition receptors (PRRs) and release soluble mediators that can modulate the immune infiltrates at the site of tissue injury and repair. This review discusses the diversity and heterogeneity of fibroblasts in secondary lymphoid organs such as lymph nodes, spleen, and Peyer's patches, and their contributions to a range of pathological and physiological processes. The role of trans-differentiated effector fibroblast phenotypes that modulate the expression and function of various innate immune components (PRRs, cytokines, chemokines, and complement) in maintaining homeostasis has also been discussed.
Collapse
Affiliation(s)
- Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Remya P. G. Ramesh
- Department of Veterinary Medicine (CAVM), UAE University, Al Ain, United Arab Emirates
| | - Ann Mary Joseph
- Department of Veterinary Medicine (CAVM), UAE University, Al Ain, United Arab Emirates
| | - Uday Kishore
- Department of Veterinary Medicine (CAVM), UAE University, Al Ain, United Arab Emirates
- Zayed Centre for Health Sciences, UAE University, Al Ain, United Arab Emirates
| |
Collapse
|
4
|
Wang J, Xu P, Ji Z, Cheng C, Liu Y, Du G, Zhang S, Miao J, Wang D, Chen R, Li D, Zhang K, Zhao H, Sun Y, Chen X, Jing N, Liu K, He Y, Xi X, Zhang Y, Wang N, Xu L, Yao J, Gao X, Zhou J, Fan S, Wang X, Dong S, Chen F, Hou J, Zhang M, Gao WQ, Shen L, Zhang P, Zhu HH. A Gremlin 1-expressing splenic niche cell population restrains chronic myeloid leukemia by antagonizing the BMP pathway. NATURE CANCER 2025; 6:666-681. [PMID: 40097655 DOI: 10.1038/s43018-025-00933-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025]
Abstract
The spleen plays a critical role in the pathogenesis of leukemia. However, our understanding of the splenic niche is very limited. Herein, we report that induced expression of the secreted protein Gremlin 1 in a mouse model restrains chronic myeloid leukemia (CML) progression and synergizes with tyrosine kinase inhibitor treatment, whereas blockade of Gremlin 1 promotes CML development. Intriguingly, the effect of Gremlin 1 is most evident in the spleen but not in the bone marrow. Gremlin 1 induces apoptosis of leukemic stem cells via antagonizing the BMP pathway. Single-cell RNA sequencing and experimental validation together show that Gremlin 1 marks a unique stromal cell population in the spleens of both mice and humans. Genetic ablation of Gremlin 1+ cells leads to accelerated CML progression. Collectively, Gremlin 1 and Gremlin 1+ cells are key defensive niche components in the spleen that limit CML progression, revealing an unprecedented mechanism for the body to fight off leukemia.
Collapse
MESH Headings
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Animals
- Spleen/metabolism
- Spleen/pathology
- Mice
- Humans
- Signal Transduction
- Bone Morphogenetic Proteins/metabolism
- Bone Morphogenetic Proteins/antagonists & inhibitors
- Intercellular Signaling Peptides and Proteins/metabolism
- Intercellular Signaling Peptides and Proteins/genetics
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Disease Models, Animal
- Apoptosis
- Disease Progression
- Mice, Inbred C57BL
- Stem Cell Niche
Collapse
Affiliation(s)
- Jinming Wang
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Penghui Xu
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhongzhong Ji
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chaping Cheng
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yiyun Liu
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Genyu Du
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Shilei Zhang
- Department of Pathology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juju Miao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Deng Wang
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ruoyang Chen
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dawei Li
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Zhang
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Huifang Zhao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yujiao Sun
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyu Chen
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Na Jing
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Kaiyuan Liu
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yuman He
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xialian Xi
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yingchao Zhang
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Nan Wang
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Longmei Xu
- Shanghai Cancer Institute, Shanghai, China
| | - Jufang Yao
- Animal Facility of Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomei Gao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Jianhua Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Songqing Fan
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaorui Wang
- Clinical Laboratory Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuxian Dong
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangli Chen
- Department of Hematology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Hou
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Zhang
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Qiang Gao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Lijing Shen
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengcheng Zhang
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China
| | - Helen He Zhu
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, Shanghai Cancer Institute, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
5
|
Onder L, Papadopoulou C, Lütge A, Cheng HW, Lütge M, Perez-Shibayama C, Gil-Cruz C, De Martin A, Kurz L, Cadosch N, Pikor NB, Rodriguez R, Born D, Jochum W, Leskow P, Dutly A, Robinson MD, Ludewig B. Fibroblastic reticular cells generate protective intratumoral T cell environments in lung cancer. Cell 2025; 188:430-446.e20. [PMID: 39566495 DOI: 10.1016/j.cell.2024.10.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/28/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024]
Abstract
Stringent control of T cell activity in the tumor microenvironment is essential for the generation of protective antitumor immunity. However, the identity, differentiation, and functions of the cells that create critical fibroblastic niches promoting tumor-infiltrating T cells remain elusive. Here, we show that CCL19-expressing fibroblastic reticular cells (FRCs) generate interconnected T cell environments (TEs) in human non-small cell lung cancer, including tertiary lymphoid structures and T cell tracks. Analysis of the FRC-T cell interactome in TEs indicated molecular networks regulating niche-specific differentiation of CCL19-expressing fibroblasts and T cell activation pathways. Single-cell transcriptomics and cell fate-mapping analyses in mice confirmed that FRCs in TEs originate from mural and adventitial progenitors. Ablation of intratumoral FRC precursors decreased antitumor T cell activity, resulting in reduced tumor control during coronavirus vector-based immunotherapy. In summary, specialized FRC niches in the tumor microenvironment govern the quality and extent of antitumor T cell immunity.
Collapse
Affiliation(s)
- Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland.
| | - Chrysa Papadopoulou
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Almut Lütge
- Department of Molecular Life Sciences and SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich 8057, Switzerland
| | - Hung-Wei Cheng
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Mechthild Lütge
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | | | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Angelina De Martin
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Lisa Kurz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Nadine Cadosch
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Natalia B Pikor
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland; Institute of Microbiology and Immunology, ETH Zurich, Zurich 8093, Switzerland
| | - Regulo Rodriguez
- Institute of Pathology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Diana Born
- Institute of Pathology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Wolfram Jochum
- Institute of Pathology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Pawel Leskow
- Department of Thoracic Surgery, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Andre Dutly
- Department of Thoracic Surgery, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Mark D Robinson
- Department of Molecular Life Sciences and SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich 8057, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland; University Heart Center, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland; Center for Translational and Experimental Cardiology, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland.
| |
Collapse
|
6
|
Allman A, Gaudette BT, Kelly S, Alouche N, Carrington LJ, Perkey E, Brandstadter JD, Outen R, Vanderbeck A, Lederer K, Zhou Y, Faryabi RB, Robertson TF, Burkhardt JK, Tikhonova A, Aifantis I, Scarpellino L, Koch U, Radtke F, Lütge M, De Martin A, Ludewig B, Tveriakhina L, Gossler A, Mosteiro L, Siebel CW, Gómez Atria D, Luther SA, Allman D, Maillard I. Splenic fibroblasts control marginal zone B cell movement and function via two distinct Notch2-dependent regulatory programs. Immunity 2025; 58:143-161.e8. [PMID: 39731910 PMCID: PMC11735314 DOI: 10.1016/j.immuni.2024.12.003] [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: 01/09/2024] [Revised: 08/21/2024] [Accepted: 12/06/2024] [Indexed: 12/30/2024]
Abstract
Innate-like splenic marginal zone (MZ) B (MZB) cells play unique roles in immunity due to their rapid responsiveness to blood-borne microbes. How MZB cells integrate cell-extrinsic and -intrinsic processes to achieve accelerated responsiveness is unclear. We found that Delta-like1 (Dll1) Notch ligands in splenic fibroblasts regulated MZB cell pool size, migration, and function. Dll1 could not be replaced by the alternative Notch ligand Dll4. Dll1-Notch2 signaling regulated a Myc-dependent gene expression program fostering cell growth and a Myc-independent program controlling cell-movement regulators such as sphingosine-1 phosphate receptor 1 (S1PR1). S1pr1-deficient B cells experienced Notch signaling within B cell follicles without entering the MZ and were retained in the spleen upon Notch deprivation. Key elements of the mouse B cell Notch regulome were preserved in subsets of human memory B cells and B cell lymphomas. Thus, specialized niches program the poised state and patrolling behavior of MZB cells via conserved Myc-dependent and Myc-independent Notch2-regulated mechanisms.
Collapse
Affiliation(s)
- Anneka Allman
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian T Gaudette
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samantha Kelly
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nagham Alouche
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Léolène J Carrington
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric Perkey
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA
| | - Joshua D Brandstadter
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Riley Outen
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley Vanderbeck
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Katlyn Lederer
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Yeqiao Zhou
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert B Faryabi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tanner F Robertson
- Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA; Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Anastasia Tikhonova
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Iannis Aifantis
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | | | | | | | - Mechthild Lütge
- Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Angelina De Martin
- Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Burkhard Ludewig
- Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Lena Tveriakhina
- Institute for Molecular Biology, Medizinische Hochschule, Hannover, Germany
| | - Achim Gossler
- Institute for Molecular Biology, Medizinische Hochschule, Hannover, Germany
| | | | | | - Daniela Gómez Atria
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sanjiv A Luther
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - David Allman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA; Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA.
| | - Ivan Maillard
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA; Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
7
|
Dos Santos Peixoto R, Miller BF, Brusko MA, Aihara G, Atta L, Anant M, Atkinson MA, Brusko TM, Wasserfall CH, Fan J. Characterizing cell-type spatial relationships across length scales in spatially resolved omics data. Nat Commun 2025; 16:350. [PMID: 39753600 PMCID: PMC11699133 DOI: 10.1038/s41467-024-55700-1] [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: 10/10/2023] [Accepted: 12/18/2024] [Indexed: 01/06/2025] Open
Abstract
Spatially resolved omics (SRO) technologies enable the identification of cell types while preserving their organization within tissues. Application of such technologies offers the opportunity to delineate cell-type spatial relationships, particularly across different length scales, and enhance our understanding of tissue organization and function. To quantify such multi-scale cell-type spatial relationships, we present CRAWDAD, Cell-type Relationship Analysis Workflow Done Across Distances, as an open-source R package. To demonstrate the utility of such multi-scale characterization, recapitulate expected cell-type spatial relationships, and evaluate against other cell-type spatial analyses, we apply CRAWDAD to various simulated and real SRO datasets of diverse tissues assayed by diverse SRO technologies. We further demonstrate how such multi-scale characterization enabled by CRAWDAD can be used to compare cell-type spatial relationships across multiple samples. Finally, we apply CRAWDAD to SRO datasets of the human spleen to identify consistent as well as patient and sample-specific cell-type spatial relationships. In general, we anticipate such multi-scale analysis of SRO data enabled by CRAWDAD will provide useful quantitative metrics to facilitate the identification, characterization, and comparison of cell-type spatial relationships across axes of interest.
Collapse
Affiliation(s)
- Rafael Dos Santos Peixoto
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Brendan F Miller
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Maigan A Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Gohta Aihara
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Lyla Atta
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Manjari Anant
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Todd M Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Clive H Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Jean Fan
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
8
|
Dervas E, Michalopoulou E, Hepojoki J, Thiele T, Baggio F, Hetzel U, Kipar A. Haemolymphatic tissues of captive boa constrictor (Boa constrictor): Morphological features in healthy individuals and with boid inclusion body disease. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105302. [PMID: 39694348 DOI: 10.1016/j.dci.2024.105302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
Knowledge on the structure and composition of the haematopoietic tissue (HT) is essential to understand the basic immune functions of the immune system in any species. For reptiles, it is extremely limited, hence we undertook an in-depth in situ investigation of the HT (bone marrow, thymus, spleen, lymphatic tissue of the alimentary tract) in the common boa (Boa constrictor). We also assessed age- and disease-related changes, with a special focus on Boid Inclusion Body Disease, a highly relevant reptarenavirus-associated disease in boid snakes. The HT was subjected to gross, histological and ultrastructural examination, including special stains to highlight collagen and reticulin fibers and iron, immunohistochemistry, in situ hybridization and morphometric analyses. In general, the HT was dominated by T cells and lacked a clear structural organization, such as follicle formation. BIBD was associated with significantly higher cellularity and a granulomatous response in the spleen, and the presence of virus-infected haematopoietic cells in the bone marrow, suggesting the latter as a persistent source of viremia.
Collapse
Affiliation(s)
- E Dervas
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Switzerland.
| | - E Michalopoulou
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Switzerland
| | - J Hepojoki
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Switzerland; University of Helsinki, Faculty of Medicine, Medicum, Department of Virology, Helsinki, Finland
| | - T Thiele
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Switzerland
| | - F Baggio
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Switzerland
| | - U Hetzel
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Switzerland
| | - A Kipar
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Switzerland
| |
Collapse
|
9
|
Zubkova E, Kalinin A, Beloglazova I, Kurilina E, Menshikov M, Parfyonova Y, Tsokolaeva Z. Artificial Tertiary Lymphoid Structures: Exploring Mesenchymal Stromal Cells as a Platform for Immune Niche Formation. Int J Mol Sci 2024; 25:13286. [PMID: 39769051 PMCID: PMC11676966 DOI: 10.3390/ijms252413286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
Constructing artificial tertiary lymphoid structures (TLSs) opens new avenues for advancing cancer immunotherapy and personalized medicine by creating controllable immune niches. Mesenchymal stromal cells (MSCs) offer an ideal stromal source for such constructs, given their potent immunomodulatory abilities and accessibility. In this study, we explored the potential of adipose-derived MSCs to adopt TLS-supportive phenotypes and facilitate lymphocyte organization. Single-cell RNA sequencing revealed a distinct subpopulation of MSCs expressing key fibroblastic reticular cell (FRC)-associated markers, including IL-7, PDPN, and IL-15, though lacking follicular dendritic cell (FDC) markers. TNF-α stimulation, but not LTα2β1, further enhanced FRC marker expression (IL-7, PDPN, and ICAM1). Notably, in 3D spheroid co-culture with lymphocytes, MSCs upregulated additional FRC markers, specifically CCL21. Upon implantation into adipose tissue, MSC-lymphocyte organoids maintained structural integrity and showed extensive T-cell infiltration and partial vascularization after 15 days in vivo, although organized B-cell follicles and FDC markers were still lacking. These findings highlight MSCs' intrinsic ability to adopt an FRC-like phenotype that supports T-cell and HEV organization, suggesting that further optimization, including genetic modification, may be needed to achieve an FDC phenotype and replicate the full architectural and functional complexity of TLSs.
Collapse
Affiliation(s)
- Ekaterina Zubkova
- National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Moscow 121552, Russia
| | - Alexander Kalinin
- National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Moscow 121552, Russia
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Irina Beloglazova
- National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Moscow 121552, Russia
| | - Ella Kurilina
- National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Moscow 121552, Russia
| | - Mikhail Menshikov
- National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Moscow 121552, Russia
| | - Yelena Parfyonova
- National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Moscow 121552, Russia
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Zoya Tsokolaeva
- National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Moscow 121552, Russia
- Negovsky Research Institute of General Reanimatology, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| |
Collapse
|
10
|
dos Santos Peixoto R, Miller BF, Brusko MA, Aihara G, Atta L, Anant M, Atkinson MA, Brusko TM, Wasserfall CH, Fan J. Characterizing cell-type spatial relationships across length scales in spatially resolved omics data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.05.560733. [PMID: 39314450 PMCID: PMC11418938 DOI: 10.1101/2023.10.05.560733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Spatially resolved omics (SRO) technologies enable the identification of cell types while preserving their organization within tissues. Application of such technologies offers the opportunity to delineate cell-type spatial relationships, particularly across different length scales, and enhance our understanding of tissue organization and function. To quantify such multi-scale cell-type spatial relationships, we developed CRAWDAD, Cell-type Relationship Analysis Workflow Done Across Distances, as an open-source R package with source code and additional documentation at https://jef.works/CRAWDAD/. To demonstrate the utility of such multi-scale characterization, recapitulate expected cell-type spatial relationships, and evaluate against other cell-type spatial analyses, we applied CRAWDAD to various simulated and real SRO datasets of diverse tissues assayed by diverse SRO technologies. We further demonstrate how such multi-scale characterization enabled by CRAWDAD can be used to compare cell-type spatial relationships across multiple samples. Finally, we applied CRAWDAD to SRO datasets of the human spleen to identify consistent as well as patient and sample-specific cell-type spatial relationships. In general, we anticipate such multi-scale analysis of SRO data enabled by CRAWDAD will provide useful quantitative metrics to facilitate the identification, characterization, and comparison of cell-type spatial relationships across axes of interest.
Collapse
Affiliation(s)
- Rafael dos Santos Peixoto
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21211
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Brendan F. Miller
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21211
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Maigan A. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Gohta Aihara
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21211
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Lyla Atta
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21211
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Manjari Anant
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21211
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205
| | - Mark A. Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Todd M. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Clive H. Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Jean Fan
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21211
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218
| |
Collapse
|
11
|
Cheng MF, Abdullah FS, Buechler MB. Essential growth factor receptors for fibroblast homeostasis and activation: Fibroblast Growth Factor Receptor (FGFR), Platelet Derived Growth Factor Receptor (PDGFR), and Transforming Growth Factor β Receptor (TGFβR). F1000Res 2024; 13:120. [PMID: 38988879 PMCID: PMC11234085 DOI: 10.12688/f1000research.143514.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 07/12/2024] Open
Abstract
Fibroblasts are cells of mesenchymal origin that are found throughout the body. While these cells have several functions, their integral roles include maintaining tissue architecture through the production of key extracellular matrix components, and participation in wound healing after injury. Fibroblasts are also key mediators in disease progression during fibrosis, cancer, and other inflammatory diseases. Under these perturbed states, fibroblasts can activate into inflammatory fibroblasts or contractile myofibroblasts. Fibroblasts require various growth factors and mitogenic molecules for survival, proliferation, and differentiation. While the activity of mitogenic growth factors on fibroblasts in vitro was characterized as early as the 1970s, the proliferation and differentiation effects of growth factors on these cells in vivo are unclear. Recent work exploring the heterogeneity of fibroblasts raises questions as to whether all fibroblast cell states exhibit the same growth factor requirements. Here, we will examine and review existing studies on the influence of fibroblast growth factor receptors (FGFRs), platelet-derived growth factor receptors (PDGFRs), and transforming growth factor β receptor (TGFβR) on fibroblast cell states.
Collapse
Affiliation(s)
- Maye F. Cheng
- Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | | | | |
Collapse
|
12
|
Jiang T, Li Y, Huang X, Jayakumar P, Billiar TR, Deng M. Activation of TLR9 signaling suppresses the immunomodulating functions of CD55 lo fibroblastic reticular cells during bacterial peritonitis. Front Immunol 2024; 15:1337384. [PMID: 38827745 PMCID: PMC11140099 DOI: 10.3389/fimmu.2024.1337384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 04/18/2024] [Indexed: 06/04/2024] Open
Abstract
Fibroblastic reticular cells (FRCs) are a subpopulation of stromal cells modulating the immune environments in health and disease. We have previously shown that activation of TLR9 signaling in FRC in fat-associated lymphoid clusters (FALC) regulate peritoneal immunity via suppressing immune cell recruitment and peritoneal resident macrophage (PRM) retention. However, FRCs are heterogeneous across tissues and organs. The functions of each FRC subset and the regulation of TLR9 in distinct FRC subsets are unknown. Here, we confirmed that specific deletion of TLR9 in FRC improved bacterial clearance and survival during peritoneal infection. Furthermore, using single-cell RNA sequencing, we found two subsets of FRCs (CD55hi and CD55lo) in the mesenteric FALC. The CD55hi FRCs were enriched in gene expression related to extracellular matrix formation. The CD55lo FRCs were enriched in gene expression related to immune response. Interestingly, we found that TLR9 is dominantly expressed in the CD55lo subset. Activation of TLR9 signaling suppressed proliferation, cytokine production, and retinoid metabolism in the CD55lo FRC, but not CD55hi FRC. Notably, we found that adoptive transfer of Tlr9 -/-CD55lo FRC from mesenteric FALC more effectively improved the survival during peritonitis compared with WT-FRC or Tlr9 -/-CD55hi FRC. Furthermore, we identified CD55hi and CD55lo subsets in human adipose tissue-derived FRC and confirmed the suppressive effect of TLR9 on the proliferation and cytokine production in the CD55lo subset. Therefore, inhibition of TLR9 in the CD55lo FRCs from adipose tissue could be a useful strategy to improve the therapeutic efficacy of FRC-based therapy for peritonitis.
Collapse
Affiliation(s)
- Ting Jiang
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yiming Li
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Xingping Huang
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, New York, NY, United States
| | - Preethi Jayakumar
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, New York, NY, United States
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Meihong Deng
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, New York, NY, United States
- Departments of Molecular Medicine and Surgery, Zucker School of Medicine at Hofstra University/Northwell, New York, NY, United States
| |
Collapse
|
13
|
Perrotta S, Carnevale D. Brain-Splenic Immune System Interactions in Hypertension: Cellular and Molecular Mechanisms. Arterioscler Thromb Vasc Biol 2024; 44:65-75. [PMID: 37942610 DOI: 10.1161/atvbaha.123.318230] [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: 04/07/2023] [Accepted: 10/20/2023] [Indexed: 11/10/2023]
Abstract
Hypertension represents a major worldwide cause of death and disability, and it is becoming increasingly clear that available therapies are not sufficient to reduce the risk of major cardiovascular events. Various mechanisms contribute to blood pressure increase: neurohormonal activation, autonomic nervous system imbalance, and immune activation. Of note, the brain is an important regulator of blood pressure levels; it recognizes the peripheral perturbation and organizes a reflex response by modulating immune system and hormonal release to attempt at restoring the homeostasis. The connection between the brain and peripheral organs is mediated by the autonomic nervous system, which also modulates immune and inflammatory responses. Interestingly, an increased autonomic nervous system activity has been correlated with an altered immune response in cardiovascular diseases. The spleen is the largest immune organ exerting a potent influence on the cardiovascular system during disease and is characterized by a dense noradrenergic innervation. Taken together, these aspects led to hypothesize a key role of neuroimmune mechanisms in the onset and progression of hypertension. This review discusses how the nervous and splenic immune systems interact and how the mechanisms underlying the neuroimmune cross talk influence the disease progression.
Collapse
Affiliation(s)
- Sara Perrotta
- Department of Angiocardioneurology and Translational Medicine, Unit of Neuro and Cardiovascular Pathophysiology, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Neuromed, Pozzilli, Italy (S.P., D.C.)
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, Unit of Neuro and Cardiovascular Pathophysiology, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Neuromed, Pozzilli, Italy (S.P., D.C.)
- Department of Molecular Medicine, "Sapienza" University of Rome, Italy (D.C.)
| |
Collapse
|
14
|
De Martin A, Stanossek Y, Pikor NB, Ludewig B. Protective fibroblastic niches in secondary lymphoid organs. J Exp Med 2024; 221:e20221220. [PMID: 38038708 PMCID: PMC10691961 DOI: 10.1084/jem.20221220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023] Open
Abstract
Fibroblastic reticular cells (FRCs) are specialized fibroblasts of secondary lymphoid organs that provide the structural foundation of the tissue. Moreover, FRCs guide immune cells to dedicated microenvironmental niches where they provide lymphocytes and myeloid cells with homeostatic growth and differentiation factors. Inflammatory processes, including infection with pathogens, induce rapid morphological and functional adaptations that are critical for the priming and regulation of protective immune responses. However, adverse FRC reprogramming can promote immunopathological tissue damage during infection and autoimmune conditions and subvert antitumor immune responses. Here, we review recent findings on molecular pathways that regulate FRC-immune cell crosstalk in specialized niches during the generation of protective immune responses in the course of pathogen encounters. In addition, we discuss how FRCs integrate immune cell-derived signals to ensure protective immunity during infection and how therapies for inflammatory diseases and cancer can be developed through improved understanding of FRC-immune cell interactions.
Collapse
Affiliation(s)
- Angelina De Martin
- Institute of Immunobiology, Medical Research Center, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Yves Stanossek
- Institute of Immunobiology, Medical Research Center, Kantonsspital St.Gallen, St.Gallen, Switzerland
- Department of Otorhinolaryngology, Head and Neck Surgery, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Natalia Barbara Pikor
- Institute of Immunobiology, Medical Research Center, Kantonsspital St.Gallen, St.Gallen, Switzerland
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St.Gallen, St.Gallen, Switzerland
| |
Collapse
|
15
|
Di X, Chen J, Li Y, Wang M, Wei J, Li T, Liao B, Luo D. Crosstalk between fibroblasts and immunocytes in fibrosis: From molecular mechanisms to clinical trials. Clin Transl Med 2024; 14:e1545. [PMID: 38264932 PMCID: PMC10807359 DOI: 10.1002/ctm2.1545] [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: 07/17/2023] [Revised: 12/25/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND The impact of fibroblasts on the immune system provides insight into the function of fibroblasts. In various tissue microenvironments, multiple fibroblast subtypes interact with immunocytes by secreting growth factors, cytokines, and chemokines, leading to wound healing, fibrosis, and escape of cancer immune surveillance. However, the specific mechanisms involved in the fibroblast-immunocyte interaction network have not yet been fully elucidated. MAIN BODY AND CONCLUSION Therefore, we systematically reviewed the molecular mechanisms of fibroblast-immunocyte interactions in fibrosis, from the history of cellular evolution and cell subtype divisions to the regulatory networks between fibroblasts and immunocytes. We also discuss how these communications function in different tissue and organ statuses, as well as potential therapies targeting the reciprocal fibroblast-immunocyte interplay in fibrosis. A comprehensive understanding of these functional cells under pathophysiological conditions and the mechanisms by which they communicate may lead to the development of effective and specific therapies targeting fibrosis.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jiawei Chen
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Ya Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Menghua Wang
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jingwen Wei
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Tianyue Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Banghua Liao
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Deyi Luo
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| |
Collapse
|
16
|
Alexandre YO, Mueller SN. Splenic stromal niches in homeostasis and immunity. Nat Rev Immunol 2023; 23:705-719. [PMID: 36973361 DOI: 10.1038/s41577-023-00857-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 03/29/2023]
Abstract
The spleen is a gatekeeper of systemic immunity where immune responses against blood-borne pathogens are initiated and sustained. Non-haematopoietic stromal cells construct microanatomical niches in the spleen that make diverse contributions to physiological spleen functions and regulate the homeostasis of immune cells. Additional signals from spleen autonomic nerves also modify immune responses. Recent insight into the diversity of the splenic fibroblastic stromal cells has revised our understanding of how these cells help to orchestrate splenic responses to infection and contribute to immune responses. In this Review, we examine our current understanding of how stromal niches and neuroimmune circuits direct the immunological functions of the spleen, with a focus on T cell immunity.
Collapse
Affiliation(s)
- Yannick O Alexandre
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| |
Collapse
|
17
|
Lancaster JN. Aging of lymphoid stromal architecture impacts immune responses. Semin Immunol 2023; 70:101817. [PMID: 37572552 PMCID: PMC10929705 DOI: 10.1016/j.smim.2023.101817] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
The secondary lymphoid organs (SLOs) undergo structural changes with age, which correlates with diminishing immune responses against infectious disease. A growing body of research suggests that the aged tissue microenvironment can contribute to decreased immune function, independent of intrinsic changes to hematopoietic cells with age. Stromal cells impart structural integrity, facilitate fluid transport, and provide chemokine and cytokine signals that are essential for immune homeostasis. Mechanisms that drive SLO development have been described, but their roles in SLO maintenance with advanced age are unknown. Disorganization of the fibroblasts of the T cell and B cell zones may reduce the maintenance of naïve lymphocytes and delay immune activation. Reduced lymphatic transport efficiency with age can also delay the onset of the adaptive immune response. This review focuses on recent studies that describe age-associated changes to the stroma of the lymph nodes and spleen. We also review recent investigations into stromal cell biology, which include high-dimensional analysis of the stromal cell transcriptome and viscoelastic testing of lymph node mechanical properties, as they constitute an important framework for understanding aging of the lymphoid tissues.
Collapse
Affiliation(s)
- Jessica N Lancaster
- Department of Immunology, Mayo Clinic, 13400 E. Shea Blvd., Scottsdale, AZ, USA; Department of Cancer Biology, Mayo Clinic, 13400 E. Shea Blvd., Scottsdale, AZ, USA.
| |
Collapse
|
18
|
Yaglova NV, Gagulaeva BB, Timokhina EP, Obernikhin SS, Nazimova SV, Yaglov VV. Features of Spleen Development in Newborn Rats Exposed to Endocrine Disrupter Dichlorodiphenyltrichloroethane during the Prenatal Period. Bull Exp Biol Med 2023; 176:111-114. [PMID: 38085400 DOI: 10.1007/s10517-023-05977-x] [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: 03/14/2023] [Indexed: 12/19/2023]
Abstract
The development of the spleen in newborn male Wistar rats exposed to low-dose endocrine disruptor dichlorodiphenyltrichloroethane during the prenatal period was studied. Histological examination of the spleen revealed a more active development of periarterial lymphoid sheaths and lower granulocyte content in the organ. Cytofluorimetry showed a significantly lower content of B cells. Thus, low-dose exposure of the endocrine disruptor dichlorodiphenyltrichloroethane on the developing organism during the prenatal period induced changes in spleen morphogenesis resulting in extensive development of lymphoid formations and a lower intensity of lymphocyte and granulocyte differentiation.
Collapse
Affiliation(s)
- N V Yaglova
- Laboratory of Endocrine System Development, A. P. Avtsyn Research Institute of Human Morphology, A. P. Petrovsky National Research Center of Surgery, Moscow, Russia.
| | - B B Gagulaeva
- Laboratory of Endocrine System Development, A. P. Avtsyn Research Institute of Human Morphology, A. P. Petrovsky National Research Center of Surgery, Moscow, Russia
| | - E P Timokhina
- Laboratory of Endocrine System Development, A. P. Avtsyn Research Institute of Human Morphology, A. P. Petrovsky National Research Center of Surgery, Moscow, Russia
| | - S S Obernikhin
- Laboratory of Endocrine System Development, A. P. Avtsyn Research Institute of Human Morphology, A. P. Petrovsky National Research Center of Surgery, Moscow, Russia
| | - S V Nazimova
- Laboratory of Endocrine System Development, A. P. Avtsyn Research Institute of Human Morphology, A. P. Petrovsky National Research Center of Surgery, Moscow, Russia
| | - V V Yaglov
- Laboratory of Endocrine System Development, A. P. Avtsyn Research Institute of Human Morphology, A. P. Petrovsky National Research Center of Surgery, Moscow, Russia
| |
Collapse
|
19
|
Sonar SA, Watanabe M, Nikolich JŽ. Disorganization of secondary lymphoid organs and dyscoordination of chemokine secretion as key contributors to immune aging. Semin Immunol 2023; 70:101835. [PMID: 37651849 PMCID: PMC10840697 DOI: 10.1016/j.smim.2023.101835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023]
Abstract
Aging is characterized by progressive loss of organ and tissue function, and the immune system is no exception to that inevitable principle. Of all the age-related changes in the body, reduction of the size of, and naïve T (Tn) cell output from, the thymus occurs earliest, being prominent already before or by the time of puberty. Therefore, to preserve immunity against new infections, over much of their lives, vertebrates dominantly rely on peripheral maintenance of the Tn cell pool in the secondary lymphoid organs (SLO). However, SLO structure and function subsequently also deteriorate with aging. Several recent studies have made a convincing case that this deterioration is of major importance to the erosion of protective immunity in the last third of life. Specifically, the SLO were found to accumulate multiple degenerative changes with aging. Importantly, the results from adoptive transfer and parabiosis studies teach us that the old microenvironment is the limiting factor for protective immunity in old mice. In this review, we discuss the extent, mechanisms, and potential role of stromal cell aging in the age-related alteration of T cell homeostatic maintenance and immune function decline. We use that discussion to frame the potential strategies to correct the SLO stromal aging defects - in the context of other immune rejuvenation approaches, - to improve functional immune responses and protective immunity in older adults.
Collapse
Affiliation(s)
- Sandip Ashok Sonar
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Makiko Watanabe
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Janko Ž Nikolich
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; the Aegis Consortium for Pandemic-free Future, University of Arizona Health Sciences, USA; BIO5 Institute, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
20
|
Yoshikawa T, Oguchi A, Toriu N, Sato Y, Kobayashi T, Ogawa O, Haga H, Sakurai S, Yamamoto T, Murakawa Y, Yanagita M. Tertiary Lymphoid Tissues Are Microenvironments with Intensive Interactions between Immune Cells and Proinflammatory Parenchymal Cells in Aged Kidneys. J Am Soc Nephrol 2023; 34:1687-1708. [PMID: 37548710 PMCID: PMC10561819 DOI: 10.1681/asn.0000000000000202] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/10/2023] [Indexed: 08/08/2023] Open
Abstract
SIGNIFICANCE STATEMENT Ectopic lymphoid structures called tertiary lymphoid tissues (TLTs) develop in several kidney diseases and are associated with poor renal prognosis. However, the mechanisms underlying TLT expansion and their effect on renal regeneration remain unclear. The authors report that single-nucleus RNA sequencing and validation experiments demonstrate that TLTs potentially amplify inflammation in aged injured kidneys. Lymphocytes within TLTs promote proinflammatory phenotypes of the surrounding proximal tubules and fibroblasts within the TLTs via proinflammatory cytokine production. These proinflammatory parenchymal cells then interact with immune cells by chemokine or cytokine production. Such cell-cell interactions potentially increase inflammation, expand TLTs, and exacerbate kidney injury. These findings help illuminate renal TLT pathology and suggest potential therapeutic targets. BACKGROUND Ectopic lymphoid structures called tertiary lymphoid tissues (TLTs) develop in several kidney diseases and are associated with poor renal prognosis. However, the mechanisms that expand TLTs and underlie exacerbation of kidney injury remain unclear. METHODS We performed single-nucleus RNA sequencing (snRNA-seq) on aged mouse kidneys with TLTs after ischemia-reperfusion injury. The results were validated using immunostaining, in situ hybridization of murine and human kidneys, and in vitro experiments. RESULTS Using snRNA-seq, we identified proinflammatory and profibrotic Vcam1+ injured proximal tubules (PTs) with NF κ B and IFN-inducible transcription factor activation. VCAM1 + PTs were preferentially localized around TLTs and drove inflammation and fibrosis via the production of multiple chemokines or cytokines. Lymphocytes within TLTs expressed Tnf and Ifng at high levels, which synergistically upregulated VCAM1 and chemokine expression in cultured PT cells. In addition, snRNA-seq also identified proinflammatory and profibrotic fibroblasts, which resided within and outside TLTs, respectively. Proinflammatory fibroblasts exhibited STAT1 activation and various chemokine or cytokine production, including CXCL9/CXCL10 and B cell-activating factor, contributing to lymphocyte recruitment and survival. IFN γ upregulated the expression of these molecules in cultured fibroblasts in a STAT1-dependent manner, indicating potential bidirectional interactions between IFN γ -producing CXCR3 + T cells and proinflammatory fibroblasts within TLTs. The cellular and molecular components described in this study were confirmed in human kidneys with TLTs. CONCLUSIONS These findings suggest that TLTs potentially amplify inflammation by providing a microenvironment that allows intense interactions between renal parenchymal and immune cells. These interactions may serve as novel therapeutic targets in kidney diseases involving TLT formation.
Collapse
Affiliation(s)
- Takahisa Yoshikawa
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akiko Oguchi
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Naoya Toriu
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Yuki Sato
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Kobayashi
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Osamu Ogawa
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hironori Haga
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoko Sakurai
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Takuya Yamamoto
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Yasuhiro Murakawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- IFOM-ETS, Milan, Italy
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| |
Collapse
|
21
|
Brandstadter JD, De Martin A, Lϋtge M, Ferreira A, Gaudette BT, Stanossek Y, Wang S, Gonzalez MV, Camiolo E, Wertheim G, Austin B, Allman D, Bagg A, Lim MS, Fajgenbaum DC, Aster JC, Ludewig B, Maillard I. A novel cryopreservation and biobanking strategy to study lymphoid tissue stromal cells in human disease. Eur J Immunol 2023; 53:e2250362. [PMID: 37366295 PMCID: PMC10529925 DOI: 10.1002/eji.202250362] [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: 12/29/2022] [Revised: 05/03/2023] [Accepted: 05/19/2023] [Indexed: 06/28/2023]
Abstract
Nonhematopoietic lymph node stromal cells (LNSCs) regulate lymphocyte trafficking, survival, and function for key roles in host defense, autoimmunity, alloimmunity, and lymphoproliferative disorders. However, the study of LNSCs in human diseases is complicated by a dependence on viable lymphoid tissues, which are most often excised prior to establishment of a specific diagnosis. Here, we demonstrate that cryopreservation can be used to bank lymphoid tissue for the study of LNSCs in human disease. Using human tonsils and lymph nodes (LN), lymphoid tissue fragments were cryopreserved for subsequent enzymatic digestion and recovery of viable nonhematopoietic cells. Flow cytometry and single-cell transcriptomics identified comparable proportions of LN stromal cell types in fresh and cryopreserved tissue. Moreover, cryopreservation had little effect on transcriptional profiles, which showed significant overlap between tonsils and LN. The presence and spatial distribution of transcriptionally defined cell types were confirmed by in situ analyses. Our broadly applicable approach promises to greatly enable research into the roles of LNSCs in human disease.
Collapse
Affiliation(s)
- Joshua D Brandstadter
- Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angelina De Martin
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Mechthild Lϋtge
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Antonio Ferreira
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian T Gaudette
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yves Stanossek
- Department of Otorhinolaryngology, Head and Neck Surgery, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Shumei Wang
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael V Gonzalez
- Center for Cytokine Storm Treatment and Laboratory, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Camiolo
- Children’s Hospital of Philadelphia, Department of Pathology and Laboratory Medicine, Philadelphia, PA, USA
| | - Gerald Wertheim
- Children’s Hospital of Philadelphia, Department of Pathology and Laboratory Medicine, Philadelphia, PA, USA
| | - Bridget Austin
- Center for Cytokine Storm Treatment and Laboratory, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Allman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Megan S Lim
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - David C Fajgenbaum
- Center for Cytokine Storm Treatment and Laboratory, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jon C Aster
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Ivan Maillard
- Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
22
|
Ghonim MA, Boyd DF, Flerlage T, Thomas PG. Pulmonary inflammation and fibroblast immunoregulation: from bench to bedside. J Clin Invest 2023; 133:e170499. [PMID: 37655660 PMCID: PMC10471178 DOI: 10.1172/jci170499] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
In recent years, there has been an explosion of interest in how fibroblasts initiate, sustain, and resolve inflammation across disease states. Fibroblasts contain heterogeneous subsets with diverse functionality. The phenotypes of these populations vary depending on their spatial distribution within the tissue and the immunopathologic cues contributing to disease progression. In addition to their roles in structurally supporting organs and remodeling tissue, fibroblasts mediate critical interactions with diverse immune cells. These interactions have important implications for defining mechanisms of disease and identifying potential therapeutic targets. Fibroblasts in the respiratory tract, in particular, determine the severity and outcome of numerous acute and chronic lung diseases, including asthma, chronic obstructive pulmonary disease, acute respiratory distress syndrome, and idiopathic pulmonary fibrosis. Here, we review recent studies defining the spatiotemporal identity of the lung-derived fibroblasts and the mechanisms by which these subsets regulate immune responses to insult exposures and highlight past, current, and future therapeutic targets with relevance to fibroblast biology in the context of acute and chronic human respiratory diseases. This perspective highlights the importance of tissue context in defining fibroblast-immune crosstalk and paves the way for identifying therapeutic approaches to benefit patients with acute and chronic pulmonary disorders.
Collapse
Affiliation(s)
- Mohamed A. Ghonim
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al Azhar University, Cairo, Egypt
| | - David F. Boyd
- Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, USA
| | - Tim Flerlage
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
23
|
Lütge M, De Martin A, Gil-Cruz C, Perez-Shibayama C, Stanossek Y, Onder L, Cheng HW, Kurz L, Cadosch N, Soneson C, Robinson MD, Stoeckli SJ, Ludewig B, Pikor NB. Conserved stromal-immune cell circuits secure B cell homeostasis and function. Nat Immunol 2023; 24:1149-1160. [PMID: 37202489 PMCID: PMC10307622 DOI: 10.1038/s41590-023-01503-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 04/03/2023] [Indexed: 05/20/2023]
Abstract
B cell zone reticular cells (BRCs) form stable microenvironments that direct efficient humoral immunity with B cell priming and memory maintenance being orchestrated across lymphoid organs. However, a comprehensive understanding of systemic humoral immunity is hampered by the lack of knowledge of global BRC sustenance, function and major pathways controlling BRC-immune cell interactions. Here we dissected the BRC landscape and immune cell interactome in human and murine lymphoid organs. In addition to the major BRC subsets underpinning the follicle, including follicular dendritic cells, PI16+ RCs were present across organs and species. As well as BRC-produced niche factors, immune cell-driven BRC differentiation and activation programs governed the convergence of shared BRC subsets, overwriting tissue-specific gene signatures. Our data reveal that a canonical set of immune cell-provided cues enforce bidirectional signaling programs that sustain functional BRC niches across lymphoid organs and species, thereby securing efficient humoral immunity.
Collapse
Affiliation(s)
- Mechthild Lütge
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Angelina De Martin
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | | | - Yves Stanossek
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
- Department of Otorhinolaryngology Head and Neck Surgery, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Hung-Wei Cheng
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Lisa Kurz
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Nadine Cadosch
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Charlotte Soneson
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Mark D Robinson
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Sandro J Stoeckli
- Department of Otorhinolaryngology Head and Neck Surgery, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland.
| | - Natalia B Pikor
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland.
| |
Collapse
|
24
|
Ben-Chetrit N, Niu X, Swett AD, Sotelo J, Jiao MS, Stewart CM, Potenski C, Mielinis P, Roelli P, Stoeckius M, Landau DA. Integration of whole transcriptome spatial profiling with protein markers. Nat Biotechnol 2023; 41:788-793. [PMID: 36593397 PMCID: PMC10272089 DOI: 10.1038/s41587-022-01536-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 09/29/2022] [Indexed: 01/03/2023]
Abstract
Spatial transcriptomics and proteomics provide complementary information that independently transformed our understanding of complex biological processes. However, experimental integration of these modalities is limited. To overcome this, we developed Spatial PrOtein and Transcriptome Sequencing (SPOTS) for high-throughput simultaneous spatial transcriptomics and protein profiling. Compared with unimodal measurements, SPOTS substantially improves signal resolution and cell clustering and enhances the discovery power in differential gene expression analysis across tissue regions.
Collapse
Affiliation(s)
- Nir Ben-Chetrit
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Xiang Niu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Ariel D Swett
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Jesus Sotelo
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Maria S Jiao
- Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Caitlin M Stewart
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Catherine Potenski
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | | | | | | | - Dan A Landau
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
- New York Genome Center, New York, NY, USA.
| |
Collapse
|
25
|
De Martin A, Stanossek Y, Lütge M, Cadosch N, Onder L, Cheng HW, Brandstadter JD, Maillard I, Stoeckli SJ, Pikor NB, Ludewig B. PI16 + reticular cells in human palatine tonsils govern T cell activity in distinct subepithelial niches. Nat Immunol 2023:10.1038/s41590-023-01502-4. [PMID: 37202490 DOI: 10.1038/s41590-023-01502-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 04/03/2023] [Indexed: 05/20/2023]
Abstract
Fibroblastic reticular cells (FRCs) direct the interaction and activation of immune cells in discrete microenvironments of lymphoid organs. Despite their important role in steering innate and adaptive immunity, the age- and inflammation-associated changes in the molecular identity and functional properties of human FRCs have remained largely unknown. Here, we show that human tonsillar FRCs undergo dynamic reprogramming during life and respond vigorously to inflammatory perturbation in comparison to other stromal cell types. The peptidase inhibitor 16 (PI16)-expressing reticular cell (PI16+ RC) subset of adult tonsils exhibited the strongest inflammation-associated structural remodeling. Interactome analysis combined with ex vivo and in vitro validation revealed that T cell activity within subepithelial niches is controlled by distinct molecular pathways during PI16+ RC-lymphocyte interaction. In sum, the topological and molecular definition of the human tonsillar stromal cell landscape reveals PI16+ RCs as a specialized FRC niche at the core of mucosal immune responses in the oropharynx.
Collapse
Affiliation(s)
- Angelina De Martin
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Yves Stanossek
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Otorhinolaryngology, Head and Neck Surgery, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Mechthild Lütge
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Nadine Cadosch
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Hung-Wei Cheng
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Joshua D Brandstadter
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ivan Maillard
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sandro J Stoeckli
- Department of Otorhinolaryngology, Head and Neck Surgery, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Natalia B Pikor
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.
| |
Collapse
|
26
|
Gábris F, Kiss G, Szirmay B, Szomor Á, Berta G, Jakus Z, Kellermayer Z, Balogh P. Absence of Nkx2-3 induces ectopic lymphatic endothelial differentiation associated with impaired extramedullary stress hematopoiesis in the spleen. Front Cell Dev Biol 2023; 11:1170389. [PMID: 37091975 PMCID: PMC10113473 DOI: 10.3389/fcell.2023.1170389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/27/2023] [Indexed: 04/09/2023] Open
Abstract
The red and white pulps as two main parts of the spleen are arranged around distinct types of vasculature, and perform significantly different functions in both humans and mice. Previous observations indicated a profound alteration of the local vessel specialization in mice lacking Nkx2-3 homeodomain transcription factor, including contradictory results suggesting presence of an ectopic lymphatic vascular structure. Furthermore, how the absence of Nkx2-3 and the consequential changes in endothelial components affect the extramedullary hematopoietic activity restricted to the splenic red pulp is unknown. In this work, we investigated the role of Nkx2-3 homeodomain transcription factor as a major morphogenic determinant for vascular specification, and its effect in the extramedullary hematopoiesis following acute blood loss and pharmacological stimulation of megakaryocyte differentiation after treatment with thrombopoietin-receptor mimetic Romiplostim. We found that, in mice lacking Nkx2-3, Prox1-positive lymphatic capillaries containing gp38/CD31 double positive lymphatic endothelial cells develop, arranged into an extensive meshwork, while the Clever1-positive venous segments of red pulp blood vasculature are absent. This lymphatic endothelial shift is coupled with a severely compromised splenic erythropoiesis and a significantly reduced splenic megakaryocyte colony formation following Romiplostim treatment in mice lacking Nkx2-3. These findings indicate that the shift of microvascular patterning in the absence of Nkx2-3 includes the emergence of ectopic Prox1-positive lymphatic vessels, and that this pivoting towards lymph node-like vascular patterning is associated with an impaired reserve hematopoietic capacity of the splenic red pulp.
Collapse
Affiliation(s)
- Fanni Gábris
- Department of Immunology and Biotechnology, University of Pécs Medical School, Pécs, Hungary
- Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Gabriella Kiss
- Department of Laboratory Medicine, University of Pécs Medical School, Pécs, Hungary
| | - Balázs Szirmay
- Department of Laboratory Medicine, University of Pécs Medical School, Pécs, Hungary
| | - Árpád Szomor
- Department of Internal Medicine, University of Pécs Medical School, Pécs, Hungary
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Pécs, Hungary
| | - Zoltán Jakus
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Zoltán Kellermayer
- Department of Immunology and Biotechnology, University of Pécs Medical School, Pécs, Hungary
- Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Péter Balogh
- Department of Immunology and Biotechnology, University of Pécs Medical School, Pécs, Hungary
- Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs, Pécs, Hungary
- *Correspondence: Péter Balogh,
| |
Collapse
|
27
|
Lim VY, Feng X, Miao R, Zehentmeier S, Ewing-Crystal N, Lee M, Tumanov AV, Oh JE, Iwasaki A, Wang A, Choi J, Pereira JP. Mature B cells and mesenchymal stem cells control emergency myelopoiesis. Life Sci Alliance 2023; 6:e202301924. [PMID: 36717247 PMCID: PMC9889502 DOI: 10.26508/lsa.202301924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
Systemic inflammation halts lymphopoiesis and prioritizes myeloid cell production. How blood cell production switches from homeostasis to emergency myelopoiesis is incompletely understood. Here, we show that lymphotoxin-β receptor (LTβR) signaling in combination with TNF and IL-1 receptor signaling in bone marrow mesenchymal stem cells (MSCs) down-regulates Il7 expression to shut down lymphopoiesis during systemic inflammation. LTβR signaling in MSCs also promoted CCL2 production during systemic inflammation. Pharmacological or genetic blocking of LTβR signaling in MSCs partially enabled lymphopoiesis and reduced monocyte numbers in the spleen during systemic inflammation, which correlated with reduced survival during systemic bacterial and viral infections. Interestingly, lymphotoxin-α1β2 delivered by B-lineage cells, and specifically by mature B cells, contributed to promote Il7 down-regulation and reduce MSC lymphopoietic activity. Our studies revealed an unexpected role of LTβR signaling in MSCs and identified recirculating mature B cells as an important regulator of emergency myelopoiesis.
Collapse
Affiliation(s)
- Vivian Y Lim
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Xing Feng
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Runfeng Miao
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Sandra Zehentmeier
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Nathan Ewing-Crystal
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Moonyoung Lee
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
| | - Alexei V Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ji Eun Oh
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Andrew Wang
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Medicine (Rheumatology), School of Medicine, Yale University, New Haven, CT, USA
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA
| | - João P Pereira
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
28
|
Brandstadter JD, De Martin A, Lütge M, Ferreira A, Gaudette BT, Stanossek Y, Wang S, Gonzalez MV, Camiolo E, Wertheim G, Austin B, Allman D, Lim MS, Fajgenbaum DC, Aster JC, Ludewig B, Maillard I. A novel cryopreservation and biobanking strategy to study lymphoid tissue stromal cells in human disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.525604. [PMID: 36798373 PMCID: PMC9934566 DOI: 10.1101/2023.02.06.525604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Non-hematopoietic lymph node stromal cells (LNSCs) regulate lymphocyte trafficking, survival, and function for key roles in host defense, autoimmunity, alloimmunity, and lymphoproliferative disorders. However, study of LNSCs in human diseases is complicated by a dependence on viable lymphoid tissues, which are most often excised prior to establishment of a specific diagnosis. Here, we demonstrate that cryopreservation can be used to bank lymphoid tissue for the study of LNSCs in human disease. Using human tonsils, lymphoid tissue fragments were cryopreserved for subsequent enzymatic digestion and recovery of viable non-hematopoietic cells. Flow cytometry and single-cell transcriptomics identified comparable proportions of LNSC cell types in fresh and cryopreserved tissue. Moreover, cryopreservation had little effect on transcriptional profiles, which showed significant overlap between tonsils and lymph nodes. The presence and spatial distribution of transcriptionally defined cell types was confirmed by in situ analyses. Our broadly applicable approach promises to greatly enable research into the roles of LNSC in human disease.
Collapse
|
29
|
Gauthier V, Kyriazi M, Nefla M, Pucino V, Raza K, Buckley CD, Alsaleh G. Fibroblast heterogeneity: Keystone of tissue homeostasis and pathology in inflammation and ageing. Front Immunol 2023; 14:1137659. [PMID: 36926329 PMCID: PMC10011104 DOI: 10.3389/fimmu.2023.1137659] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Fibroblasts, derived from the embryonic mesenchyme, are a diverse array of cells with roles in development, homeostasis, repair, and disease across tissues. In doing so, fibroblasts maintain micro-environmental homeostasis and create tissue niches by producing a complex extracellular matrix (ECM) including various structural proteins. Although long considered phenotypically homogenous and functionally identical, the emergence of novel technologies such as single cell transcriptomics has allowed the identification of different phenotypic and cellular states to be attributed to fibroblasts, highlighting their role in tissue regulation and inflammation. Therefore, fibroblasts are now recognised as central actors in many diseases, increasing the need to discover new therapies targeting those cells. Herein, we review the phenotypic heterogeneity and functionality of these cells and their roles in health and disease.
Collapse
Affiliation(s)
- Vincent Gauthier
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom.,The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom.,Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Maria Kyriazi
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom.,Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Meriam Nefla
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom.,Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Valentina Pucino
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom.,Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Karim Raza
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.,Department of Rheumatology, Sandwell and West, Birmingham Hospitals NHS Trust, Birmingham, United Kingdom
| | - Christopher D Buckley
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom.,Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Ghada Alsaleh
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom.,The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
| |
Collapse
|
30
|
Pezoldt J, Wiechers C, Zou M, Litovchenko M, Biocanin M, Beckstette M, Sitnik K, Palatella M, van Mierlo G, Chen W, Gardeux V, Floess S, Ebel M, Russeil J, Arampatzi P, Vafardanejad E, Saliba AE, Deplancke B, Huehn J. Postnatal expansion of mesenteric lymph node stromal cells towards reticular and CD34 + stromal cell subsets. Nat Commun 2022; 13:7227. [PMID: 36433946 PMCID: PMC9700677 DOI: 10.1038/s41467-022-34868-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
Gut-draining mesenteric lymph nodes (LN) provide the framework to shape intestinal adaptive immune responses. Based on the transcriptional signatures established by our previous work, the composition and immunomodulatory function of LN stromal cells (SC) vary according to location. Here, we describe the single-cell composition and development of the SC compartment within mesenteric LNs derived from postnatal to aged mice. We identify CD34+ SC and fibroblastic reticular stromal cell (FRC) progenitors as putative progenitors, both supplying the typical rapid postnatal mesenteric LN expansion. We further establish the location-specific chromatin accessibility and DNA methylation landscape of non-endothelial SCs and identify a microbiota-independent core epigenomic signature, showing characteristic differences between SCs from mesenteric and skin-draining peripheral LNs. The epigenomic landscape of SCs points to dynamic expression of Irf3 along the differentiation trajectories of FRCs. Accordingly, a mesenchymal stem cell line acquires a Cxcl9+ FRC molecular phenotype upon lentiviral overexpression of Irf3, and the relevance of Irf3 for SC biology is further underscored by the diminished proportion of Ccl19+ and Cxcl9+ FRCs in LNs of Irf3-/- mice. Together, our data constitute a comprehensive transcriptional and epigenomic map of mesenteric LNSC development in early life and dissect location-specific, microbiota-independent properties of non-endothelial SCs.
Collapse
Affiliation(s)
- Joern Pezoldt
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany ,grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Carolin Wiechers
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Mangge Zou
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Maria Litovchenko
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Marjan Biocanin
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Michael Beckstette
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany ,grid.512472.7Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine, Helmholtz Centre for Infection Research and Hannover Medical School, 30625 Hannover, Germany ,grid.7491.b0000 0001 0944 9128Genome Informatics Group, Bielefeld Institute for Bioinformatics Infrastructure, Department of Technology, Bielefeld University, 33615 Bielefeld, Germany
| | - Katarzyna Sitnik
- grid.6583.80000 0000 9686 6466Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Martina Palatella
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Guido van Mierlo
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Wanze Chen
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Vincent Gardeux
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Stefan Floess
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Maria Ebel
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Julie Russeil
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Panagiota Arampatzi
- grid.8379.50000 0001 1958 8658Core Unit Systems Medicine, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Ehsan Vafardanejad
- grid.498164.6Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), 97080 Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- grid.498164.6Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), 97080 Würzburg, Germany
| | - Bart Deplancke
- grid.5333.60000000121839049Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jochen Huehn
- grid.7490.a0000 0001 2238 295XDepartment Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany ,grid.10423.340000 0000 9529 9877Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
31
|
Huang J, Deng R, Li W, Jiang M, Xiang AP, Zhang X. Nestin+ Mesenchymal Precursors Generate Distinct Spleen Stromal Cell Subsets and Have Immunomodulatory Function. Int J Mol Sci 2022; 23:ijms231911819. [PMID: 36233119 PMCID: PMC9569994 DOI: 10.3390/ijms231911819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are known to be widespread in many tissues and possess a broad spectrum of immunoregulatory properties. They have been used in the treatment of a variety of inflammatory diseases; however, the therapeutic effects are still inconsistent owing to their heterogeneity. Spleen stromal cells have evolved to regulate the immune response at many levels as they are bathed in a complex inflammatory milieu during infection. Therefore, it is unknown whether they have stronger immunomodulatory effects than their counterparts derived from other tissues. Here, using a transgenic mouse model expressing GFP driven by the Nestin (Nes) promoter, Nes-GFP+ cells from bone marrow and spleen were collected. Artificial lymphoid reconstruction in vivo was performed. Cell phenotype, inhibition of T cell inflammatory cytokines, and in vivo therapeutic effects were assessed. We observed Nes-GFP+ cells colocalized with splenic stromal cells and further demonstrated that these Nes-GFP+ cells had the ability to establish ectopic lymphoid-like structures in vivo. Moreover, we showed that the Nes-GFP+ cells possessed the characteristics of MSCs. Spleen-derived Nes-GFP+ cells exhibited greater immunomodulatory ability in vitro and more remarkable therapeutic efficacy in inflammatory diseases, especially inflammatory bowel disease (IBD) than bone marrow-derived Nes-GFP+ cells. Overall, our data showed that Nes-GFP+ cells contributed to subsets of spleen stromal populations and possessed the biological characteristics of MSCs with a stronger immunoregulatory function and therapeutic potential than bone marrow-derived Nes-GFP+ cells.
Collapse
Affiliation(s)
- Jing Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Ronghai Deng
- Department of Organ Transplantation, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Weiqiang Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Meihua Jiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510080, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
- Correspondence: ; Tel.: +86-20-87335982
| |
Collapse
|
32
|
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
|
33
|
Zehentmeier S, Lim VY, Ma Y, Fossati J, Ito T, Jiang Y, Tumanov AV, Lee HJ, Dillinger L, Kim J, Csomos K, Walter JE, Choi J, Pereira JP. Dysregulated stem cell niches and altered lymphocyte recirculation cause B and T cell lymphopenia in WHIM syndrome. Sci Immunol 2022; 7:eabo3170. [PMID: 36149943 PMCID: PMC9614684 DOI: 10.1126/sciimmunol.abo3170] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Gain-of-function (GOF) mutations in CXCR4 cause WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) syndrome, characterized by infections, leukocyte retention in bone marrow (BM), and blood leukopenias. B lymphopenia is evident at early progenitor stages, yet why do CXCR4 GOF mutations that cause B (and T) lymphopenia remain obscure? Using a CXCR4 R334X GOF mouse model of WHIM syndrome, we showed that lymphopoiesis is reduced because of a dysregulated mesenchymal stem cell (MSC) transcriptome characterized by a switch from an adipogenic to an osteolineage-prone program with limited lymphopoietic activity. We identify lymphotoxin beta receptor (LTβR) as a critical pathway promoting interleukin-7 (IL-7) down-regulation in MSCs. Blocking LTβR or CXCR4 signaling restored IL-7 production and B cell development in WHIM mice. LTβR blocking also increased production of IL-7 and B cell activating factor (BAFF) in secondary lymphoid organs (SLOs), increasing B and T cell numbers in the periphery. These studies revealed that LTβR signaling in BM MSCs and SLO stromal cells limits the lymphocyte compartment size.
Collapse
Affiliation(s)
- Sandra Zehentmeier
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Vivian Y Lim
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Yifan Ma
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Julia Fossati
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Takeshi Ito
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Yawen Jiang
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| | - Alexei V Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ho-Joon Lee
- Department of Genetics and Yale Center for Genome Analysis, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA
| | - Lukas Dillinger
- X4 Pharmaceuticals Inc., Cambridge, MA, USA
- X4 Pharmaceuticals Inc., Vienna, Austria
| | - Jihyun Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Krisztian Csomos
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jolan E Walter
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Division Allergy and Immunology, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Jungmin Choi
- Department of Genetics and Yale Center for Genome Analysis, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - João P Pereira
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, USA
| |
Collapse
|
34
|
Kwok T, Medovich SC, Silva-Junior IA, Brown EM, Haug JC, Barrios MR, Morris KA, Lancaster JN. Age-Associated Changes to Lymph Node Fibroblastic Reticular Cells. FRONTIERS IN AGING 2022; 3:838943. [PMID: 35821826 PMCID: PMC9261404 DOI: 10.3389/fragi.2022.838943] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/07/2022] [Indexed: 12/26/2022]
Abstract
The decreased proportion of antigen-inexperienced, naïve T cells is a hallmark of aging in both humans and mice, and contributes to reduced immune responses, particularly against novel and re-emerging pathogens. Naïve T cells depend on survival signals received during their circulation among the lymph nodes by direct contacts with stroma, in particular fibroblastic reticular cells. Macroscopic changes to the architecture of the lymph nodes have been described, but it is unclear how lymph node stroma are altered with age, and whether these changes contribute to reduced naïve T cell maintenance. Here, using 2-photon microscopy, we determined that the aged lymph node displayed increased fibrosis and correspondingly, that naïve T-cell motility was impaired in the aged lymph node, especially in proximity to fibrotic deposition. Functionally, adoptively transferred young naïve T-cells exhibited reduced homeostatic turnover in aged hosts, supporting the role of T cell-extrinsic mechanisms that regulate their survival. Further, we determined that early development of resident fibroblastic reticular cells was impaired, which may correlate to the declining levels of naïve T-cell homeostatic factors observed in aged lymph nodes. Thus, our study addresses the controversy as to whether aging impacts the composition lymph node stroma and supports a model in which impaired differentiation of lymph node fibroblasts and increased fibrosis inhibits the interactions necessary for naïve T cell homeostasis.
Collapse
Affiliation(s)
- Tina Kwok
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | | | | | - Elise M Brown
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | - Joel C Haug
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | | | - Karina A Morris
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | | |
Collapse
|
35
|
Assen FP, Abe J, Hons M, Hauschild R, Shamipour S, Kaufmann WA, Costanzo T, Krens G, Brown M, Ludewig B, Hippenmeyer S, Heisenberg CP, Weninger W, Hannezo E, Luther SA, Stein JV, Sixt M. Multitier mechanics control stromal adaptations in the swelling lymph node. Nat Immunol 2022; 23:1246-1255. [PMID: 35817845 PMCID: PMC9355878 DOI: 10.1038/s41590-022-01257-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 06/07/2022] [Indexed: 11/09/2022]
Abstract
Lymph nodes (LNs) comprise two main structural elements: fibroblastic reticular cells that form dedicated niches for immune cell interaction and capsular fibroblasts that build a shell around the organ. Immunological challenge causes LNs to increase more than tenfold in size within a few days. Here, we characterized the biomechanics of LN swelling on the cellular and organ scale. We identified lymphocyte trapping by influx and proliferation as drivers of an outward pressure force, causing fibroblastic reticular cells of the T-zone (TRCs) and their associated conduits to stretch. After an initial phase of relaxation, TRCs sensed the resulting strain through cell matrix adhesions, which coordinated local growth and remodeling of the stromal network. While the expanded TRC network readopted its typical configuration, a massive fibrotic reaction of the organ capsule set in and countered further organ expansion. Thus, different fibroblast populations mechanically control LN swelling in a multitier fashion. Sixt and colleagues show that different fibroblast populations in the lymph node mechanically control its swelling in a multitier fashion.
Collapse
Affiliation(s)
- Frank P Assen
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria. .,Department of Dermatology, Medical University Vienna, Vienna, Austria.
| | - Jun Abe
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Miroslav Hons
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria.,BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Robert Hauschild
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Shayan Shamipour
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Walter A Kaufmann
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Tommaso Costanzo
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Gabriel Krens
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Markus Brown
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | | | - Wolfgang Weninger
- Department of Dermatology, Medical University Vienna, Vienna, Austria
| | - Edouard Hannezo
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Sanjiv A Luther
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Michael Sixt
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria.
| |
Collapse
|
36
|
Dwyer GK, Mathews LR, Villegas JA, Lucas A, Gonzalez de Peredo A, Blazar BR, Girard JP, Poholek AC, Luther SA, Shlomchik W, Turnquist HR. IL-33 acts as a costimulatory signal to generate alloreactive Th1 cells in graft-versus-host disease. J Clin Invest 2022; 132:e150927. [PMID: 35503257 PMCID: PMC9197517 DOI: 10.1172/jci150927] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 04/28/2022] [Indexed: 12/02/2022] Open
Abstract
Antigen-presenting cells (APCs) integrate signals emanating from local pathology and program appropriate T cell responses. In allogeneic hematopoietic stem cell transplantation (alloHCT), recipient conditioning releases damage-associated molecular patterns (DAMPs) that generate proinflammatory APCs that secrete IL-12, which is a driver of donor Th1 responses, causing graft-versus-host disease (GVHD). Nevertheless, other mechanisms exist to initiate alloreactive T cell responses, as recipients with disrupted DAMP signaling or lacking IL-12 develop GVHD. We established that tissue damage signals are perceived directly by donor CD4+ T cells and promoted T cell expansion and differentiation. Specifically, the fibroblastic reticular cell-derived DAMP IL-33 is increased by recipient conditioning and is critical for the initial activation, proliferation, and differentiation of alloreactive Th1 cells. IL-33 stimulation of CD4+ T cells was not required for lymphopenia-induced expansion, however. IL-33 promoted IL-12-independent expression of Tbet and generation of Th1 cells that infiltrated GVHD target tissues. Mechanistically, IL-33 augmented CD4+ T cell TCR-associated signaling pathways in response to alloantigen. This enhanced T cell expansion and Th1 polarization, but inhibited the expression of regulatory molecules such as IL-10 and Foxp3. These data establish an unappreciated role for IL-33 as a costimulatory signal for donor Th1 generation after alloHCT.
Collapse
Affiliation(s)
- Gaelen K. Dwyer
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute and
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lisa R. Mathews
- Thomas E. Starzl Transplantation Institute and
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - José A. Villegas
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Anna Lucas
- Thomas E. Starzl Transplantation Institute and
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anne Gonzalez de Peredo
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique, Université Paul Sabatier, Toulouse, France
| | - Bruce R. Blazar
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Centre National de la Recherche Scientifique, Université Paul Sabatier, Toulouse, France
| | - Amanda C. Poholek
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pediatric Rheumatology, and
| | - Sanjiv A. Luther
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Warren Shlomchik
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute and
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Hēth R. Turnquist
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Thomas E. Starzl Transplantation Institute and
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
37
|
Scheffler JM, Gustafsson KL, Barrett A, Corciulo C, Drevinge C, Del Carpio Pons AM, Humeniuk P, Engdahl C, Gustafsson J, Ohlsson C, Carlsten H, Lagerquist MK, Islander U. ERα signaling in a subset of CXCL12‐abundant reticular cells regulates trabecular bone in mice. JBMR Plus 2022; 6:e10657. [PMID: 35991530 PMCID: PMC9382863 DOI: 10.1002/jbm4.10657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/22/2022] [Accepted: 05/22/2022] [Indexed: 12/02/2022] Open
Abstract
Estrogen has pronounced effects on the immune system, which also influences bone homeostasis. In recent years, stromal cells in lymphoid organs have gained increasing attention as they not only support the regulation of immune responses but also affect bone remodeling. A conditional knockout mouse model where estrogen receptor alpha (ERα) is deleted in CCL19‐expressing stromal cells (Ccl19‐Cre ERαfl/fl mice) was generated and bone densitometry was performed to analyze the importance of stromal cell–specific ERα signaling on the skeleton. Results showed that female Ccl19‐Cre ERαfl/fl mice display reduced total bone mineral density and detailed X‐ray analyses revealed that ERα expression in CCL19‐expressing stromal cells is important for trabecular but not cortical bone homeostasis. Further analysis showed that the trabecular bone loss is caused by increased osteoclastogenesis. Additionally, the bone formation rate was reduced; however, the expression of osteoprogenitor genes was not altered. Analysis of the bone marrow stromal cell compartment revealed a deletion of ERα in a subgroup of CXCL12‐abundant reticular (CAR) cells resulting in increased secretion of the pro‐osteoclastogenic chemokine CXCL12. In conclusion, this study reveals the importance of ERα signaling in CAR cells for bone health. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Julia M. Scheffler
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Sweden
| | - Karin L. Gustafsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Sweden
| | - Aidan Barrett
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Sweden
| | - Carmen Corciulo
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Sweden
| | - Christina Drevinge
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Sweden
| | - Alicia M. Del Carpio Pons
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Sweden
| | - Piotr Humeniuk
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Sweden
| | - Cecilia Engdahl
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Sweden
| | - Jan‐Åke Gustafsson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry University of Houston Houston Texas USA
- Department of Biosciences and Nutrition Karolinska Institute Huddinge Sweden
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Sweden
- Department of Drug Treatment Sahlgrenska University Hospital, Region Västra Götaland Gothenburg Sweden
| | - Hans Carlsten
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Sweden
| | - Marie Kristina Lagerquist
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Sweden
| | - Ulrika Islander
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy University of Gothenburg Sweden
| |
Collapse
|
38
|
Intestinal fibroblastic reticular cell niches control innate lymphoid cell homeostasis and function. Nat Commun 2022; 13:2027. [PMID: 35440118 PMCID: PMC9018819 DOI: 10.1038/s41467-022-29734-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 03/21/2022] [Indexed: 11/30/2022] Open
Abstract
Innate lymphoid cells (ILCs) govern immune cell homeostasis in the intestine and protect the host against microbial pathogens. Various cell-intrinsic pathways have been identified that determine ILC development and differentiation. However, the cellular components that regulate ILC sustenance and function in the intestinal lamina propria are less known. Using single-cell transcriptomic analysis of lamina propria fibroblasts, we identify fibroblastic reticular cells (FRCs) that underpin cryptopatches (CPs) and isolated lymphoid follicles (ILFs). Genetic ablation of lymphotoxin-β receptor expression in Ccl19-expressing FRCs blocks the maturation of CPs into mature ILFs. Interactome analysis shows the major niche factors and processes underlying FRC-ILC crosstalk. In vivo validation confirms that a sustained lymphotoxin-driven feedforward loop of FRC activation including IL-7 generation is critical for the maintenance of functional ILC populations. In sum, our study indicates critical fibroblastic niches within the intestinal lamina propria that control ILC homeostasis and functionality and thereby secure protective gut immunity. Fibroblastic reticular cells (FRCs) support localisation of immune cells in secondary lymphoid tissues but less is known about the lamina propria. Here the authors use scRNA-seq and intestinal infection to characterise FRCs in the intestinal lamina propria and show specialised niches that foster innate lymphoid cells during homeostasis and infection.
Collapse
|
39
|
A single-cell atlas of non-haematopoietic cells in human lymph nodes and lymphoma reveals a landscape of stromal remodelling. Nat Cell Biol 2022; 24:565-578. [PMID: 35332263 PMCID: PMC9033586 DOI: 10.1038/s41556-022-00866-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 02/10/2022] [Indexed: 12/14/2022]
Abstract
The activities of non-haematopoietic cells (NHCs), including mesenchymal stromal cells and endothelial cells, in lymphomas are reported to underlie lymphomagenesis. However, our understanding of lymphoma NHCs has been hampered by unexplained NHC heterogeneity, even in normal human lymph nodes (LNs). Here we constructed a single-cell transcriptome atlas of more than 100,000 NHCs collected from 27 human samples, including LNs and various nodal lymphomas, and it revealed 30 distinct subclusters, including some that were previously unrecognized. Notably, this atlas was useful for comparative analyses with lymphoma NHCs, which revealed an unanticipated landscape of subcluster-specific changes in gene expression and interaction with malignant cells in follicular lymphoma NHCs. This facilitates our understanding of stromal remodelling in lymphoma and highlights potential clinical biomarkers. Our study largely updates NHC taxonomy in human LNs and analysis of disease status, and provides a rich resource and deeper insights into LN and lymphoma biology to advance lymphoma management and therapy. Abe et al. profile, characterize and compare non-haematopoietic cells in normal human lymph nodes versus nodal lymphomas from patients, providing insights into stromal modelling in health and disease.
Collapse
|
40
|
Liu D, Duan L, Cyster JG. Chemo- and mechanosensing by dendritic cells facilitate antigen surveillance in the spleen. Immunol Rev 2022; 306:25-42. [PMID: 35147233 PMCID: PMC8852366 DOI: 10.1111/imr.13055] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/05/2021] [Indexed: 12/30/2022]
Abstract
Spleen dendritic cells (DC) are critical for initiation of adaptive immune responses against blood-borne invaders. Key to DC function is their positioning at sites of pathogen entry, and their abilities to selectively capture foreign antigens and promptly engage T cells. Focusing on conventional DC2 (cDC2), we discuss the contribution of chemoattractant receptors (EBI2 or GPR183, S1PR1, and CCR7) and integrins to cDC2 positioning and function. We give particular attention to a newly identified role in cDC2 for adhesion G-protein coupled receptor E5 (Adgre5 or CD97) and its ligand CD55, detailing how this mechanosensing system contributes to splenic cDC2 positioning and homeostasis. Additional roles of CD97 in the immune system are reviewed. The ability of cDC2 to be activated by circulating missing self-CD47 cells and to integrate multiple red blood cell (RBC)-derived inputs is discussed. Finally, we describe the process of activated cDC2 migration to engage and prime helper T cells. Throughout the review, we consider the insights into cDC function in the spleen that have emerged from imaging studies.
Collapse
Affiliation(s)
- Dan Liu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| | - Lihui Duan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| |
Collapse
|
41
|
CD169 + macrophages in lymph node and spleen critically depend on dual RANK and LTbetaR signaling. Proc Natl Acad Sci U S A 2022; 119:2108540119. [PMID: 35031565 PMCID: PMC8784161 DOI: 10.1073/pnas.2108540119] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2021] [Indexed: 12/16/2022] Open
Abstract
The CD169+ macrophages that play an important role in the fight against infections and cancer are receptive to environmental signals for their differentiation. We show that lymph node and splenic CD169+ macrophages require both LTβR and RANK signaling since the conditional deficiency of either receptor results in their disappearance. Using a reporter mouse, we observe RANKL expression by a splenic mesenchymal cell subset and show that it participates in CD169+ macrophage differentiation. Their absence leads to a reduced viral capture and a greatly attenuated virus-specific CD8+ T cell expansion. Thus, tight control mechanisms operate for the precise positioning of these macrophages at sites where numerous immune-stimulatory forces converge. CD169+ macrophages reside in lymph node (LN) and spleen and play an important role in the immune defense against pathogens. As resident macrophages, they are responsive to environmental cues to shape their tissue-specific identity. We have previously shown that LN CD169+ macrophages require RANKL for formation of their niche and their differentiation. Here, we demonstrate that they are also dependent on direct lymphotoxin beta (LTβ) receptor (R) signaling. In the absence or the reduced expression of either RANK or LTβR, their differentiation is perturbed, generating myeloid cells expressing SIGN-R1 in LNs. Conditions of combined haploinsufficiencies of RANK and LTβR revealed that both receptors contribute equally to LN CD169+ macrophage differentiation. In the spleen, the Cd169-directed ablation of either receptor results in a selective loss of marginal metallophilic macrophages (MMMs). Using a RANKL reporter mouse, we identify splenic marginal zone stromal cells as a source of RANKL and demonstrate that it participates in MMM differentiation. The loss of MMMs had no effect on the splenic B cell compartments but compromised viral capture and the expansion of virus-specific CD8+ T cells. Taken together, the data provide evidence that CD169+ macrophage differentiation in LN and spleen requires dual signals from LTβR and RANK with implications for the immune response.
Collapse
|
42
|
Alexandre YO, Schienstock D, Lee HJ, Gandolfo LC, Williams CG, Devi S, Pal B, Groom JR, Cao W, Christo SN, Gordon CL, Starkey G, D'Costa R, Mackay LK, Haque A, Ludewig B, Belz GT, Mueller SN. A diverse fibroblastic stromal cell landscape in the spleen directs tissue homeostasis and immunity. Sci Immunol 2022; 7:eabj0641. [PMID: 34995096 DOI: 10.1126/sciimmunol.abj0641] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yannick O Alexandre
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Dominik Schienstock
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Hyun Jae Lee
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Luke C Gandolfo
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.,Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia
| | - Cameron G Williams
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Sapna Devi
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Bhupinder Pal
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
| | - Joanna R Groom
- Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Wang Cao
- Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Susan N Christo
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Claire L Gordon
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.,Department of Infectious Diseases, Austin Health, Melbourne, VIC, Australia
| | - Graham Starkey
- Liver and Intestinal Transplant Unit, Austin Health, Melbourne, VIC, Australia.,Department of Surgery, University of Melbourne, Austin Health, Melbourne, VIC, Australia
| | - Rohit D'Costa
- DonateLife Victoria, Carlton, VIC, Australia.,Department of Intensive Care Medicine, Melbourne Health, Melbourne, VIC, Australia
| | - Laura K Mackay
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Ashraful Haque
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Gabrielle T Belz
- Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,University of Queensland Diamantina Institute, University of Queensland, Brisbane, QLD, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
43
|
Onder L, Cheng HW, Ludewig B. Visualization and functional characterization of lymphoid organ fibroblasts. Immunol Rev 2021; 306:108-122. [PMID: 34866192 PMCID: PMC9300201 DOI: 10.1111/imr.13051] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 11/29/2022]
Abstract
Fibroblastic reticular cells (FRCs) are specialized stromal cells of lymphoid organs that generate the structural foundation of the tissue and actively interact with immune cells. Distinct FRC subsets position lymphocytes and myeloid cells in specialized niches where they present processed or native antigen and provide essential growth factors and cytokines for immune cell activation and differentiation. Niche‐specific functions of FRC subpopulations have been defined using genetic targeting, high‐dimensional transcriptomic analyses, and advanced imaging methods. Here, we review recent findings on FRC‐immune cell interaction and the elaboration of FRC development and differentiation. We discuss how imaging approaches have not only shaped our understanding of FRC biology, but have critically advanced the niche concept of immune cell maintenance and control of immune reactivity.
Collapse
Affiliation(s)
- Lucas Onder
- Institute of Immunobiology, Medical Research Center, Kantonsspital St.Gallen, St.Gallen, Switzerland
| | - Hung-Wei Cheng
- 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
| |
Collapse
|
44
|
Single-cell transcriptional profiling of splenic fibroblasts reveals subset-specific innate immune signatures in homeostasis and during viral infection. Commun Biol 2021; 4:1355. [PMID: 34857864 PMCID: PMC8640036 DOI: 10.1038/s42003-021-02882-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 11/11/2021] [Indexed: 01/12/2023] Open
Abstract
Our understanding of the composition and functions of splenic stromal cells remains incomplete. Here, based on analysis of over 20,000 single cell transcriptomes of splenic fibroblasts, we characterized the phenotypic and functional heterogeneity of these cells in healthy state and during virus infection. We describe eleven transcriptionally distinct fibroblastic cell clusters, reassuring known subsets and revealing yet unascertained heterogeneity amongst fibroblasts occupying diverse splenic niches. We further identify striking differences in innate immune signatures of distinct stromal compartments in vivo. Compared to other fibroblasts and to endothelial cells, Ly6C+ fibroblasts of the red pulp were selectively endowed with enhanced interferon-stimulated gene expression in homeostasis, upon systemic interferon stimulation and during virus infection in vivo. Collectively, we provide an updated map of fibroblastic cell diversity in the spleen that suggests a specialized innate immune function for splenic red pulp fibroblasts.
Collapse
|
45
|
Börner K, Teichmann SA, Quardokus EM, Gee JC, Browne K, Osumi-Sutherland D, Herr BW, Bueckle A, Paul H, Haniffa M, Jardine L, Bernard A, Ding SL, Miller JA, Lin S, Halushka MK, Boppana A, Longacre TA, Hickey J, Lin Y, Valerius MT, He Y, Pryhuber G, Sun X, Jorgensen M, Radtke AJ, Wasserfall C, Ginty F, Ho J, Sunshine J, Beuschel RT, Brusko M, Lee S, Malhotra R, Jain S, Weber G. Anatomical structures, cell types and biomarkers of the Human Reference Atlas. Nat Cell Biol 2021; 23:1117-1128. [PMID: 34750582 PMCID: PMC10079270 DOI: 10.1038/s41556-021-00788-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 09/29/2021] [Indexed: 02/05/2023]
Abstract
The Human Reference Atlas (HRA) aims to map all of the cells of the human body to advance biomedical research and clinical practice. This Perspective presents collaborative work by members of 16 international consortia on two essential and interlinked parts of the HRA: (1) three-dimensional representations of anatomy that are linked to (2) tables that name and interlink major anatomical structures, cell types, plus biomarkers (ASCT+B). We discuss four examples that demonstrate the practical utility of the HRA.
Collapse
Affiliation(s)
- Katy Börner
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA.
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Ellen M Quardokus
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - James C Gee
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristen Browne
- Department of Health and Human Services, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David Osumi-Sutherland
- European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Cambridge, UK
| | - Bruce W Herr
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Andreas Bueckle
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Hrishikesh Paul
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Laura Jardine
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | | | | | - Shin Lin
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Marc K Halushka
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Avinash Boppana
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Teri A Longacre
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - John Hickey
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yiing Lin
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
| | - M Todd Valerius
- Harvard Institute of Medicine, Harvard Medical School, Boston, MA, USA
| | - Yongqun He
- Department of Microbiology and Immunology, and Center for Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gloria Pryhuber
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Xin Sun
- Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Marda Jorgensen
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Andrea J Radtke
- Center for Advanced Tissue Imaging, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Clive Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Fiona Ginty
- Biology and Applied Physics, General Electric Research, Niskayuna, NY, USA
| | - Jonhan Ho
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joel Sunshine
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rebecca T Beuschel
- Center for Advanced Tissue Imaging, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Maigan Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Sujin Lee
- Division of Vascular Surgery and Endovascular Therapy, Massachusetts General Hospital, Boston, MA, USA
| | - Rajeev Malhotra
- Harvard Institute of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Vascular Surgery and Endovascular Therapy, Massachusetts General Hospital, Boston, MA, USA
| | - Sanjay Jain
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Griffin Weber
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
Menzel L, Zschummel M, Crowley T, Franke V, Grau M, Ulbricht C, Hauser A, Siffrin V, Bajénoff M, Acton SE, Akalin A, Lenz G, Willimsky G, Höpken UE, Rehm A. Lymphocyte access to lymphoma is impaired by high endothelial venule regression. Cell Rep 2021; 37:109878. [PMID: 34706240 PMCID: PMC8567313 DOI: 10.1016/j.celrep.2021.109878] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/27/2021] [Accepted: 10/01/2021] [Indexed: 12/15/2022] Open
Abstract
Blood endothelial cells display remarkable plasticity depending on the demands of a malignant microenvironment. While studies in solid tumors focus on their role in metabolic adaptations, formation of high endothelial venules (HEVs) in lymph nodes extends their role to the organization of immune cell interactions. As a response to lymphoma growth, blood vessel density increases; however, the fate of HEVs remains elusive. Here, we report that lymphoma causes severe HEV regression in mouse models that phenocopies aggressive human B cell lymphomas. HEV dedifferentiation occurrs as a consequence of a disrupted lymph-carrying conduit system. Mechanosensitive fibroblastic reticular cells then deregulate CCL21 migration paths, followed by deterioration of dendritic cell proximity to HEVs. Loss of this crosstalk deprives HEVs of lymphotoxin-β-receptor (LTβR) signaling, which is indispensable for their differentiation and lymphocyte transmigration. Collectively, this study reveals a remodeling cascade of the lymph node microenvironment that is detrimental for immune cell trafficking in lymphoma.
Collapse
Affiliation(s)
- Lutz Menzel
- Translational Tumorimmunology, Max-Delbrück-Center for Molecular Medicine Berlin, Germany, 13125 Berlin, Germany
| | - Maria Zschummel
- Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine Berlin, 13125 Berlin, Germany
| | - Tadhg Crowley
- Neuroimmunology Laboratory, Max-Delbrück-Center for Molecular Medicine Berlin, Germany, 13125 Berlin, Germany
| | - Vedran Franke
- Bioinformatics & Omics Data Science Platform, BIMSB at Max-Delbrück-Center for Molecular Medicine Berlin, 13125 Berlin, Germany
| | - Michael Grau
- Medical Department A for Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Carolin Ulbricht
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, and Immune Dynamics, Deutsches Rheumaforschungszentrum Berlin, 10117 Berlin, Germany
| | - Anja Hauser
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, and Immune Dynamics, Deutsches Rheumaforschungszentrum Berlin, 10117 Berlin, Germany
| | - Volker Siffrin
- Neuroimmunology Laboratory, Max-Delbrück-Center for Molecular Medicine Berlin, Germany, 13125 Berlin, Germany; Neuroimmunology Laboratory, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Marc Bajénoff
- Aix Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, 13288 Marseille, France
| | - Sophie E Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, WC1E 6BT London, UK
| | - Altuna Akalin
- Bioinformatics & Omics Data Science Platform, BIMSB at Max-Delbrück-Center for Molecular Medicine Berlin, 13125 Berlin, Germany
| | - Georg Lenz
- Medical Department A for Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Gerald Willimsky
- Institute of Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; German Cancer Research Center, 69120 Heidelberg, Germany; German Cancer Consortium, partner site Berlin, Germany
| | - Uta E Höpken
- Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine Berlin, 13125 Berlin, Germany
| | - Armin Rehm
- Translational Tumorimmunology, Max-Delbrück-Center for Molecular Medicine Berlin, Germany, 13125 Berlin, Germany.
| |
Collapse
|
47
|
Wittenberns BM, Thamm DH, Palmer EP, Regan DP. Canine Non-Angiogenic, Non-Myogenic Splenic Stromal Sarcoma: a Retrospective Clinicopathological Analysis and Investigation of Podoplanin as a Marker of Tumour Histogenesis. J Comp Pathol 2021; 188:1-12. [PMID: 34686271 DOI: 10.1016/j.jcpa.2021.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/30/2021] [Accepted: 07/30/2021] [Indexed: 10/20/2022]
Abstract
Splenic stromal sarcomas are rarely reported tumours that were previously grouped as non-angiomatous, non-lymphomatous mesenchymal neoplasms of the canine spleen. Highly variable survival times have been reported probably due to their heterogeneous nature. The purpose of this study was to assess the outcome and prognostic factors in dogs with splenic stromal sarcoma after treatment by splenectomy. Clinical data were collected retrospectively and histopathology was reviewed for 47 patients. Histological classification, based on morphology in haematoxylin and eosin-stained sections, in conjunction with immunolabelling of macrophage scavenger receptor-A (CD204), desmin, factor VIII-related antigen and smooth muscle actin ,yielded diagnoses of undifferentiated stromal sarcoma (n = 22), complex nodular hyperplasia (CNH, n = 9), sarcoma arising from benign complex nodular hyperplasia (n = 3), histiocytic sarcoma (n = 3), haemangiosarcoma (n = 1) and leiomyosarcoma (n = 1). Four samples were excluded from analysis due to extensive necrosis. An anti-podoplanin (PDPN) antibody was validated on canine tissue and used to assess expression of this protein as a potential indicator of the tissue of origin of the neoplasms (28/42 tumours were positive). There was a statistically significant difference in survival time between patients with stromal sarcoma (sarcoma from benign CNH and undifferentiated stromal sarcoma) and CNH (178 d versus 637 d, respectively; P = 0.027). Dogs with stromal sarcomas and high mitotic count (≥9 per 10 high-power fields) had a significantly shorter survival time (67 d versus 439 d; P = 0.01). Clinical diagnosis of splenic tumours should include evaluation for the presence of benign nodular hyperplasia morphology and immunohistochemistry to exclude more aggressive malignancies where adjuvant therapy is recommended. As in humans, PDPN may be an effective marker for stromal sarcomas of the canine spleen and immunopositivity suggests a fibroblastic reticular or follicular dendritic cell origin.
Collapse
Affiliation(s)
- Brittany M Wittenberns
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA; Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Douglas H Thamm
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA; Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Eric P Palmer
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA; Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Daniel P Regan
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA; Flint Animal Cancer Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA.
| |
Collapse
|
48
|
Duan L, Liu D, Chen H, Mintz MA, Chou MY, Kotov DI, Xu Y, An J, Laidlaw BJ, Cyster JG. Follicular dendritic cells restrict interleukin-4 availability in germinal centers and foster memory B cell generation. Immunity 2021; 54:2256-2272.e6. [PMID: 34555336 PMCID: PMC8516727 DOI: 10.1016/j.immuni.2021.08.028] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/02/2021] [Accepted: 08/27/2021] [Indexed: 11/24/2022]
Abstract
B cells within germinal centers (GCs) enter cycles of antibody affinity maturation or exit the GC as memory cells or plasma cells. Here, we examined the contribution of interleukin (IL)-4 on B cell fate decisions in the GC. Single-cell RNA-sequencing identified a subset of light zone GC B cells expressing high IL-4 receptor-a (IL4Ra) and CD23 and lacking a Myc-associated signature. These cells could differentiate into pre-memory cells. B cell-specific deletion of IL4Ra or STAT6 favored the pre-memory cell trajectory, and provision of exogenous IL-4 in a wild-type context reduced pre-memory cell frequencies. IL-4 acted during antigen-specific interactions but also influenced bystander cells. Deletion of IL4Ra from follicular dendritic cells (FDCs) increased the availability of IL-4 in the GC, impaired the selection of affinity-matured B cells, and reduced memory cell generation. We propose that GC FDCs establish a niche that limits bystander IL-4 activity, focusing IL-4 action on B cells undergoing selection and enhancing memory cell differentiation.
Collapse
Affiliation(s)
- Lihui Duan
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dan Liu
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hsin Chen
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michelle A Mintz
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marissa Y Chou
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dmitri I Kotov
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, and Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ying Xu
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinping An
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Brian J Laidlaw
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jason G Cyster
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
49
|
Heesters BA, van Megesen K, Tomris I, de Vries RP, Magri G, Spits H. Characterization of human FDCs reveals regulation of T cells and antigen presentation to B cells. J Exp Med 2021; 218:e20210790. [PMID: 34424268 PMCID: PMC8404474 DOI: 10.1084/jem.20210790] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/02/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
Stromal-derived follicular dendritic cells (FDCs) are essential for germinal centers (GCs), the site where B cells maturate their antibodies. FDCs present native antigen to B cells and maintain a CXCL13 gradient to form the B cell follicle. Yet despite their essential role, the transcriptome of human FDCs remains undefined. Using single-cell RNA sequencing and microarray, we provided the transcriptome of these enigmatic cells as a comprehensive resource. Key genes were validated by flow cytometry and microscopy. Surprisingly, marginal reticular cells (MRCs) rather than FDCs expressed B cell activating factor (BAFF). Furthermore, we found that human FDCs expressed TLR4 and can alter antigen availability in response to pathogen-associated molecular patterns (PAMPs). High expression of PD-L1 and PD-L2 on FDCs activated PD1 on T cells. In addition, we found expression of genes related to T cell regulation, such as HLA-DRA, CD40, and others. These data suggest intimate contact between human FDCs and T cells.
Collapse
Affiliation(s)
- Balthasar A. Heesters
- Amsterdam University Medical Centers, University of Amsterdam, Department of Experimental Immunology, Amsterdam institute for Infection and Immunity, Amsterdam, Netherlands
| | - Kyah van Megesen
- Amsterdam University Medical Centers, University of Amsterdam, Department of Experimental Immunology, Amsterdam institute for Infection and Immunity, Amsterdam, Netherlands
| | - Ilhan Tomris
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Robert P. de Vries
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Giuliana Magri
- Program for Inflammatory and Cardiovascular Disorders, Institut Hospital del Mar d’Investigacions Mèdiques, Barcelona, Spain
| | - Hergen Spits
- Amsterdam University Medical Centers, University of Amsterdam, Department of Experimental Immunology, Amsterdam institute for Infection and Immunity, Amsterdam, Netherlands
| |
Collapse
|
50
|
Grasso C, Pierie C, Mebius RE, van Baarsen LGM. Lymph node stromal cells: subsets and functions in health and disease. Trends Immunol 2021; 42:920-936. [PMID: 34521601 DOI: 10.1016/j.it.2021.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 02/04/2023]
Abstract
Lymph nodes (LNs) aid the interaction between lymphocytes and antigen-presenting cells, resulting in adequate and prolonged adaptive immune responses. LN stromal cells (LNSCs) are crucially involved in steering adaptive immune responses at different levels. Most knowledge on LNSCs has been obtained from mouse studies, and few studies indicate similarities with their human counterparts. Recent advances in single-cell technologies have revealed significant LNSC heterogeneity among different subsets with potential selective functions in immunity. This review provides an overview of current knowledge of LNSCs based on human and murine studies describing the role of these cells in health and disease.
Collapse
Affiliation(s)
- C Grasso
- Department of Rheumatology and Clinical Immunology, Department of Experimental Immunology, Amsterdam UMC, Amsterdam Infection and Immunity Institute, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Rheumatology and Immunology Center (ARC), Academic Medical Center, Amsterdam, The Netherlands
| | - C Pierie
- Department of Rheumatology and Clinical Immunology, Department of Experimental Immunology, Amsterdam UMC, Amsterdam Infection and Immunity Institute, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Rheumatology and Immunology Center (ARC), Academic Medical Center, Amsterdam, The Netherlands
| | - R E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands.
| | - L G M van Baarsen
- Department of Rheumatology and Clinical Immunology, Department of Experimental Immunology, Amsterdam UMC, Amsterdam Infection and Immunity Institute, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Rheumatology and Immunology Center (ARC), Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|