1
|
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: 0] [Impact Index Per Article: 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
|
2
|
Calvanese AL, Cecconi V, Stäheli S, Schnepf D, Nater M, Pereira P, Gschwend J, Heikenwälder M, Schneider C, Ludewig B, Silina K, van den Broek M. Sustained innate interferon is an essential inducer of tertiary lymphoid structures. Eur J Immunol 2024; 54:e2451207. [PMID: 38980268 DOI: 10.1002/eji.202451207] [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: 04/19/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024]
Abstract
Tertiary lymphoid structures (TLS) resemble follicles of secondary lymphoid organs and develop in nonlymphoid tissues during inflammation and cancer. Which cell types and signals drive the development of TLS is largely unknown. To investigate early events of TLS development in the lungs, we repeatedly instilled p(I:C) plus ovalbumin (Ova) intranasally. This induced TLS ranging from lymphocytic aggregates to organized and functional structures containing germinal centers. We found that TLS development is independent of FAP+ fibroblasts, alveolar macrophages, or CCL19 but crucially depends on type I interferon (IFN-I). Mechanistically, IFN-I initiates two synergistic pathways that culminate in the development of TLS. On the one hand, IFN-I induces lymphotoxin (LT)α in lymphoid cells, which stimulate stromal cells to produce the B-cell-attracting chemokine CXCL13 through LTβR-signaling. On the other hand, IFN-I is sensed by stromal cells that produce the T-cell-attracting chemokines CXCL9, CXCL10 as well as CCL19 and CCL21 independently of LTβR. Consequently, B-cell aggregates develop within a week, whereas follicular dendritic cells and germinal centers appear after 3 weeks. Thus, sustained production of IFN-I together with an antigen is essential for the induction of functional TLS in the lungs.
Collapse
Affiliation(s)
| | - Virginia Cecconi
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Severin Stäheli
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Daniel Schnepf
- Institute of Virology, Medical Center University of Freiburg, Freiburg im Breisgau, Germany
| | - Marc Nater
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Paulo Pereira
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Julia Gschwend
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
- M3 Research Institute, Eberhard Karls University Tübingen, Tübingen, Germany
| | | | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Karina Silina
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
3
|
Todd L, Chin MHW, Coppens MO. Two conjectures on 3D Voronoi structures: a toolkit with biomedical case studies. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2024; 9:912-919. [PMID: 39205672 PMCID: PMC11348831 DOI: 10.1039/d4me00036f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/19/2024] [Indexed: 09/04/2024]
Abstract
3D Voronoi scaffolds are widely applied in the field of additive manufacturing as they are known for their light weight structural resilience and share many topological similarities to various natural (bone, tumours, lymph node) and synthetic environments (foam, functionally gradient porous materials). Unfortunately, the structural design features that promote these topological similarities (such as the number of vertices) are often unpredictable and require the trial and error of varying design features to achieve the desired 3D Voronoi structure. This article provides a toolkit, consisting of equations, based on over 12 000 3D Voronoi structures. These equations allow design features, such as the number of generating points (G), to be efficiently and accurately predicted based on the desired structural parameters (within ±3G). Based on these equations we are proposing, to the best of our knowledge, two new mathematical conjectures that relate the number of vertices or edges, and the average edge length to G in Voronoi structures. These equations have been validated for a wide range of parameter values and Voronoi network sizes. A design code is provided allowing any of over 12 000 structures to be selected, easily adjusted based on user requirements, and 3D printed. Biomedical case studies relevant to T-cell culturing, bone scaffolds and kidney tumours are presented to illustrate the design code.
Collapse
Affiliation(s)
- Lucy Todd
- Centre for Nature Inspired Engineering & Department of Chemical Engineering, University College London Torrington Place London WC1E 7JE United Kingdom
| | - Matthew H W Chin
- Centre for Nature Inspired Engineering & Department of Chemical Engineering, University College London Torrington Place London WC1E 7JE United Kingdom
| | - Marc-Olivier Coppens
- Centre for Nature Inspired Engineering & Department of Chemical Engineering, University College London Torrington Place London WC1E 7JE United Kingdom
| |
Collapse
|
4
|
Barrett A, Horkeby K, Corciulo C, Carlsten H, Lagerquist MK, Scheffler JM, Islander U. Role of estrogen signaling in fibroblastic reticular cells for innate and adaptive immune responses in antigen-induced arthritis. Immunol Cell Biol 2024; 102:578-592. [PMID: 38726582 DOI: 10.1111/imcb.12773] [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: 04/03/2024] [Accepted: 04/26/2024] [Indexed: 08/03/2024]
Abstract
Women are more prone to develop rheumatoid arthritis, with peak incidence occurring around menopause. Estrogen has major effects on the immune system and is protective against arthritis. We have previously shown that treatment with estrogen inhibits inflammation and joint destruction in murine models of arthritis, although the mechanisms involved remain unclear. Fibroblastic reticular cells (FRCs) are specialized stromal cells that generate the three-dimensional structure of lymph nodes (LNs). FRCs are vital for coordinating immune responses from within LNs and are characterized by the expression of the chemokine CCL19, which attracts immune cells. The aim of this study was to determine whether the influence of estrogen on innate and adaptive immune cells in arthritis is mediated by estrogen signaling in FRCs. Conditional knockout mice lacking estrogen receptor α (ERα) in CCL19-expressing cells (Ccl19-CreERαfl/fl) were generated and tested. Ccl19-CreERαfl/fl mice and littermate controls were ovariectomized, treated with vehicle or estradiol and subjected to the 28-day-long antigen-induced arthritis model to enable analyses of differentiated T- and B-cell populations and innate cells in LNs by flow cytometry. The results reveal that while the response to estradiol treatment in numbers of FRCs per LN is significantly reduced in mice lacking ERα in FRCs, estrogen does not inhibit joint inflammation or markedly affect immune responses in this arthritis model. Thus, this study validates the Ccl19-CreERαfl/fl strain for studying estrogen signaling in FRCs within inflammatory diseases, although the chosen arthritis model is deemed unsuitable for addressing this question.
Collapse
Affiliation(s)
- Aidan Barrett
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Horkeby
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Center, Centre for Bone and Arthritis Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carmen Corciulo
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hans Carlsten
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marie K Lagerquist
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Center, Centre for Bone and Arthritis Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Julia M Scheffler
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulrika Islander
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
5
|
Castranova D, Kenton MI, Kraus A, Dell CW, Park JS, Galanternik MV, Park G, Lumbantobing DN, Dye L, Marvel M, Iben J, Taimatsu K, Pham V, Willms RJ, Blevens L, Robertson TF, Hou Y, Huttenlocher A, Foley E, Parenti LR, Frazer JK, Narayan K, Weinstein BM. The axillary lymphoid organ - an external, experimentally accessible immune organ in the zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.25.605139. [PMID: 39091802 PMCID: PMC11291151 DOI: 10.1101/2024.07.25.605139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Lymph nodes and other secondary lymphoid organs play critical roles in immune surveillance and immune activation in mammals, but the deep internal locations of these organs make it challenging to image and study them in living animals. Here, we describe a previously uncharacterized external immune organ in the zebrafish ideally suited for studying immune cell dynamics in vivo, the axillary lymphoid organ (ALO). This small, translucent organ has an outer cortex teeming with immune cells, an inner medulla with a mesh-like network of fibroblastic reticular cells along which immune cells migrate, and a network of lymphatic vessels draining to a large adjacent lymph sac. Noninvasive high-resolution imaging of transgenically marked immune cells can be carried out in the lobes of living animals, and the ALO is readily accessible to external treatment. This newly discovered tissue provides a superb model for dynamic live imaging of immune cells and their interaction with pathogens and surrounding tissues, including blood and lymphatic vessels.
Collapse
Affiliation(s)
- Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Madeleine I. Kenton
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Aurora Kraus
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Christopher W. Dell
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA and Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jong S. Park
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Marina Venero Galanternik
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Gilseung Park
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Daniel N. Lumbantobing
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC 20560, USA
| | - Louis Dye
- Microscopy and Imaging Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Miranda Marvel
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - James Iben
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Kiyohito Taimatsu
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Van Pham
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Reegan J. Willms
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lucas Blevens
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Tanner F. Robertson
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706
| | - Yiran Hou
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Lynne R. Parenti
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC 20560, USA
| | - J. Kimble Frazer
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA and Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Brant M. Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| |
Collapse
|
6
|
Velankar K, Liu W, Hartmeier PR, Veleke SR, Reddy GA, Clegg B, Gawalt ES, Fan Y, Meng WS. Fibril-Guided Three-Dimensional Assembly of Human Fibroblastic Reticular Cells. ACS APPLIED BIO MATERIALS 2024; 7:3953-3963. [PMID: 38805413 PMCID: PMC11190984 DOI: 10.1021/acsabm.4c00331] [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: 03/07/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024]
Abstract
Fibroblastic reticular cells (FRCs) are stromal cells (SCs) that can be isolated from lymph node (LN) biopsies. Studies have shown that these nonhematopoietic cells have the capacity to shape and regulate adaptive immunity and can become a form of personalized cell therapy. Successful translational efforts, however, require the cells to be formulated as injectable units, with their native architecture preserved. The intrinsic reticular organization of FRCs, however, is lost in the monolayer cultures. Organizing FRCs into three-dimensional (3D) clusters would recapitulate their structural and functional attributes. Herein, we report a scaffolding method based on the self-assembling peptide (SAP) EAKII biotinylated at the N-terminus (EAKbt). Cross-linking with avidin transformed the EAKbt fibrils into a dense network of coacervates. The combined forces of fibrillization and bioaffinity interactions in the cross-linked EAKbt likely drove the cells into a cohesive 3D reticula. This facile method of generating clustered FRCs (clFRCs) can be completed within 10 days. In vitro clFRCs attracted the infiltration of T cells and rendered an immunosuppressive milieu in the cocultures. These results demonstrate the potential of clFRCs as a method for stromal cell delivery.
Collapse
Affiliation(s)
- Ketki
Y. Velankar
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, Pittsburgh Pennsylvania 15282, United States
| | - Wen Liu
- Allegheny
Health Network Cancer Institute, Allegheny Health Network, Pittsburgh Pennsylvania 15212, United States
| | - Paul R. Hartmeier
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, Pittsburgh Pennsylvania 15282, United States
| | - Samuel R. Veleke
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, Pittsburgh Pennsylvania 15282, United States
| | - Gayathri Aparnasai Reddy
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, Pittsburgh Pennsylvania 15282, United States
| | - Benjamin Clegg
- Department
of Chemistry and Biochemistry, Duquesne
University, Pittsburgh, Pennsylvania 15282, United States
| | - Ellen S. Gawalt
- Department
of Chemistry and Biochemistry, Duquesne
University, Pittsburgh, Pennsylvania 15282, United States
- McGowan
Institute for Regenerative Medicine, University
of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Yong Fan
- Allegheny
Health Network Cancer Institute, Allegheny Health Network, Pittsburgh Pennsylvania 15212, United States
- Department
of Biomedical Engineering, Carnegie Mellon
University, Pittsburgh,Pennsylvania 15213, United States
| | - Wilson S. Meng
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, Pittsburgh Pennsylvania 15282, United States
- McGowan
Institute for Regenerative Medicine, University
of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
7
|
Horsnell HL, Cao WH, Belz GT, Mueller SN, Alexandre YO. The transcription factor SpiB regulates the fibroblastic reticular cell network and CD8 + T-cell responses in lymph nodes. Immunol Cell Biol 2024; 102:269-279. [PMID: 38441326 DOI: 10.1111/imcb.12740] [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: 10/13/2023] [Revised: 02/02/2024] [Accepted: 02/14/2024] [Indexed: 04/04/2024]
Abstract
Fibroblastic reticular cells (FRCs) construct microanatomical niches that support lymph node (LN) homeostasis and coordination of immune responses. Transcription factors regulating the functionality of FRCs remain poorly understood. Here, we investigated the role of the transcription factor SpiB that is expressed in LN FRCs. Conditional ablation of SpiB in FRCs impaired the FRC network in the T-cell zone of LNs, leading to reduced numbers of FRCs and altered homeostatic functions including reduced CCL21 and interleukin-7 expression. The size and cellularity of LNs remained intact in the absence of SpiB but the space between the reticular network increased, indicating that although FRCs were reduced in number they stretched to maintain network integrity. Following virus infection, antiviral CD8+ T-cell responses were impaired, suggesting a role for SpiB expression in FRCs in orchestrating immune responses. Together, our findings reveal a new role for SpiB as an important regulator of FRC functions and immunity in LNs.
Collapse
Affiliation(s)
- Harry L Horsnell
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Wang Hj Cao
- 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 Frazer Institute, University of Queensland, Brisbane, QLD, Australia
| | - 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 Frazer 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
| | - Yannick O Alexandre
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
8
|
Morrison AI, Mikula AM, Spiekstra SW, de Kok M, Affandi AJ, Roest HP, van der Laan LJW, de Winde CM, Koning JJ, Gibbs S, Mebius RE. An Organotypic Human Lymph Node Model Reveals the Importance of Fibroblastic Reticular Cells for Dendritic Cell Function. Tissue Eng Regen Med 2024; 21:455-471. [PMID: 38114886 PMCID: PMC10987465 DOI: 10.1007/s13770-023-00609-x] [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: 07/14/2023] [Revised: 10/19/2023] [Accepted: 10/22/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Human lymph node (HuLN) models have emerged with invaluable potential for immunological research and therapeutic application given their fundamental role in human health and disease. While fibroblastic reticular cells (FRCs) are instrumental to HuLN functioning, their inclusion and recognition of importance for organotypic in vitro lymphoid models remain limited. METHODS Here, we established an in vitro three-dimensional (3D) model in a collagen-fibrin hydrogel with primary FRCs and a dendritic cell (DC) cell line (MUTZ-3 DC). To study and characterise the cellular interactions seen in this 3D FRC-DC organotypic model compared to the native HuLN; flow cytometry, immunohistochemistry, immunofluorescence and cytokine/chemokine analysis were performed. RESULTS FRCs were pivotal for survival, proliferation and localisation of MUTZ-3 DCs. Additionally, we found that CD1a expression was absent on MUTZ-3 DCs that developed in the presence of FRCs during cytokine-induced MUTZ-3 DC differentiation, which was also seen with primary monocyte-derived DCs (moDCs). This phenotype resembled HuLN-resident DCs, which we detected in primary HuLNs, and these CD1a- MUTZ-3 DCs induced T cell proliferation within a mixed leukocyte reaction (MLR), indicating a functional DC status. FRCs expressed podoplanin (PDPN), CD90 (Thy-1), CD146 (MCAM) and Gremlin-1, thereby resembling the DC supporting stromal cell subset identified in HuLNs. CONCLUSION This 3D FRC-DC organotypic model highlights the influence and importance of FRCs for DC functioning in a more realistic HuLN microenvironment. As such, this work provides a starting point for the development of an in vitro HuLN.
Collapse
Affiliation(s)
- Andrew I Morrison
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Aleksandra M Mikula
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Sander W Spiekstra
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Michael de Kok
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Alsya J Affandi
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Henk P Roest
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Charlotte M de Winde
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jasper J Koning
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Susan Gibbs
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Department Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands
| | - Reina E Mebius
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Chin MHW, Reid B, Lachina V, Acton SE, Coppens MO. Bioinspired 3D microprinted cell scaffolds: Integration of graph theory to recapitulate complex network wiring in lymph nodes. Biotechnol J 2024; 19:e2300359. [PMID: 37986209 DOI: 10.1002/biot.202300359] [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: 07/28/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/22/2023]
Abstract
Physical networks are ubiquitous in nature, but many of them possess a complex organizational structure that is difficult to recapitulate in artificial systems. This is especially the case in biomedical and tissue engineering, where the microstructural details of 3D cell scaffolds are important. Studies of biological networks-such as fibroblastic reticular cell (FRC) networks-have revealed the crucial role of network topology in a range of biological functions. However, cell scaffolds are rarely analyzed, or designed, using graph theory. To understand how networks affect adhered cells, 3D culture platforms capturing the complex topological properties of biologically relevant networks would be needed. In this work, we took inspiration from the small-world organization (high clustering and low path length) of FRC networks to design cell scaffolds. An algorithmic toolset was created to generate the networks and process them to improve their 3D printability. We employed tools from graph theory to show that the networks were small-world (omega factor, ω = -0.10 ± 0.02; small-world propensity, SWP = 0.74 ± 0.01). 3D microprinting was employed to physicalize networks as scaffolds, which supported the survival of FRCs. This work, therefore, represents a bioinspired, graph theory-driven approach to control the networks of microscale cell niches.
Collapse
Affiliation(s)
- Matthew H W Chin
- EPSRC "Frontier Engineering" Centre for Nature-Inspired Engineering (CNIE) and Department of Chemical Engineering, University College London, Torrington Place, London, UK
| | - Barry Reid
- EPSRC "Frontier Engineering" Centre for Nature-Inspired Engineering (CNIE) and Department of Chemical Engineering, University College London, Torrington Place, London, UK
| | - Veronika Lachina
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Sophie E Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Marc-Olivier Coppens
- EPSRC "Frontier Engineering" Centre for Nature-Inspired Engineering (CNIE) and Department of Chemical Engineering, University College London, Torrington Place, London, UK
| |
Collapse
|
10
|
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
|
11
|
D'Rozario J, Knoblich K, Lütge M, Shibayama CP, Cheng HW, Alexandre YO, Roberts D, Campos J, Dutton EE, Suliman M, Denton AE, Turley SJ, Boyd RL, Mueller SN, Ludewig B, Heng TSP, Fletcher AL. Fibroblastic reticular cells provide a supportive niche for lymph node-resident macrophages. Eur J Immunol 2023; 53:e2250355. [PMID: 36991561 PMCID: PMC10947543 DOI: 10.1002/eji.202250355] [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: 12/23/2022] [Revised: 03/13/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023]
Abstract
The lymph node (LN) is home to resident macrophage populations that are essential for immune function and homeostasis, but key factors controlling this niche are undefined. Here, we show that fibroblastic reticular cells (FRCs) are an essential component of the LN macrophage niche. Genetic ablation of FRCs caused rapid loss of macrophages and monocytes from LNs across two in vivo models. Macrophages co-localized with FRCs in human LNs, and murine single-cell RNA-sequencing revealed that FRC subsets broadly expressed master macrophage regulator CSF1. Functional assays containing purified FRCs and monocytes showed that CSF1R signaling was sufficient to support macrophage development. These effects were conserved between mouse and human systems. These data indicate an important role for FRCs in maintaining the LN parenchymal macrophage niche.
Collapse
Affiliation(s)
- Joshua D'Rozario
- Department of Biochemistry and Molecular Biology, and Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Konstantin Knoblich
- Department of Biochemistry and Molecular Biology, and Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Mechthild Lütge
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | | | - Hung-Wei Cheng
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Yannick O Alexandre
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, VIC, Melbourne, Australia
| | - David Roberts
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Joana Campos
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Emma E Dutton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Muath Suliman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Alice E Denton
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Shannon J Turley
- Department of Cancer Immunology, Genentech Inc., South San Francisco, CA, USA
| | - Richard L Boyd
- Cartherics Pty Ltd, Hudson Institute for Medical Research, Clayton, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, VIC, Melbourne, Australia
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Tracy S P Heng
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, Australia
| | - Anne L Fletcher
- Department of Biochemistry and Molecular Biology, and Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| |
Collapse
|
12
|
Song S, Senoussi M, Escande P, Villoutreix P. Random walk informed heterogeneity detection reveals how the lymph node conduit network influences T cells collective exploration behavior. PLoS Comput Biol 2023; 19:e1011168. [PMID: 37224180 DOI: 10.1371/journal.pcbi.1011168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 05/09/2023] [Indexed: 05/26/2023] Open
Abstract
Random walks on networks are widely used to model stochastic processes such as search strategies, transportation problems or disease propagation. A prominent example of such process is the dynamics of naive T cells within the lymph node while they are scanning for antigens. The observed T cells trajectories in small sub-volumes of the lymph node are well modeled as a random walk and they have been shown to follow the lymphatic conduit network as substrate for migration. One can then ask how does the connectivity patterns of the lymph node conduit network affect the T cells collective exploration behavior. In particular, does the network display properties that are uniform across the whole volume of the lymph node or can we distinguish some heterogeneities? We propose a workflow to accurately and efficiently define and compute these quantities on large networks, which enables us to characterize heterogeneities within a very large published dataset of Lymph Node Conduit Network. To establish the significance of our results, we compared the results obtained on the lymph node to null models of varying complexity. We identified significantly heterogeneous regions characterized as "remote regions" at the poles and next to the medulla, while a large portion of the network promotes uniform exploration by T cells.
Collapse
Affiliation(s)
- Solène Song
- Aix Marseille Univ, Université de Toulon, CNRS, LIS Turing Centre for Living Systems, Marseille, France
| | - Malek Senoussi
- Aix Marseille Univ, Université de Toulon, CNRS, LIS Turing Centre for Living Systems, Marseille, France
| | - Paul Escande
- Aix Marseille Univ, CNRS, Centrale Marseille, I2M, Marseille, France
| | - Paul Villoutreix
- Aix Marseille Univ, Université de Toulon, CNRS, LIS Turing Centre for Living Systems, Marseille, France
| |
Collapse
|
13
|
Song J, Deshpande T, Zhang X, Hannocks MJ, Lycke N, Cardell SL, Sorokin L. The extracellular matrix of lymph node reticular fibers modulates follicle border interactions and germinal center formation. iScience 2023; 26:106753. [PMID: 37234087 PMCID: PMC10206498 DOI: 10.1016/j.isci.2023.106753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/15/2022] [Accepted: 04/23/2023] [Indexed: 05/27/2023] Open
Abstract
Germinal center (GC) formation and antibody production in lymph node follicles require coordinated interactions between B-cells, T-cells and dendritic cells (DCs), orchestrated by the extracellular matrix-rich reticular fiber (RF) network. We describe a unique laminin 523-containing RF network around and between follicles that associates with PDGFrecβhighCCL19lowgp38low fibroblastic reticular cells (FRC). In the absence of FRC expression of laminin α5 (pdgfrb-cre:Lama5fl/fl), pre-Tfh-cells, B-cells and DCs are displaced from follicle borders, correlating with fewer Tfh-cells and GC B-cells. Total DCs are not altered in pdgfrb-cre:Lama5fl/fl mice, but cDC2s, which localize to laminin α5 in RFs at follicle borders, are reduced. In addition, PDGFrecβhighCCL19lowgp38low FRCs show lower Ch25h expression, required for 7α,25-dihydroxycholesterol synthesis that attracts pre-Tfh-cells, B-cells and DCs to follicle borders. We propose that RF basement membrane components represent a type of tissue memory that guides the localization and differentiation of both specialized FRC and DC populations, required for normal lymph node function.
Collapse
Affiliation(s)
- Jian Song
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Interfaculty Centre (CIMIC), University of Muenster, 48149 Muenster, Germany
| | - Tushar Deshpande
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Interfaculty Centre (CIMIC), University of Muenster, 48149 Muenster, Germany
| | - Xueli Zhang
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Interfaculty Centre (CIMIC), University of Muenster, 48149 Muenster, Germany
| | - Melanie-Jane Hannocks
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Interfaculty Centre (CIMIC), University of Muenster, 48149 Muenster, Germany
| | - Nils Lycke
- Department of Microbiology and Immunology, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Susanna L. Cardell
- Department of Microbiology and Immunology, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Interfaculty Centre (CIMIC), University of Muenster, 48149 Muenster, Germany
| |
Collapse
|
14
|
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: 0] [Impact Index Per Article: 0] [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
|
15
|
Ozulumba T, Montalbine AN, Ortiz-Cárdenas JE, Pompano RR. New tools for immunologists: models of lymph node function from cells to tissues. Front Immunol 2023; 14:1183286. [PMID: 37234163 PMCID: PMC10206051 DOI: 10.3389/fimmu.2023.1183286] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
The lymph node is a highly structured organ that mediates the body's adaptive immune response to antigens and other foreign particles. Central to its function is the distinct spatial assortment of lymphocytes and stromal cells, as well as chemokines that drive the signaling cascades which underpin immune responses. Investigations of lymph node biology were historically explored in vivo in animal models, using technologies that were breakthroughs in their time such as immunofluorescence with monoclonal antibodies, genetic reporters, in vivo two-photon imaging, and, more recently spatial biology techniques. However, new approaches are needed to enable tests of cell behavior and spatiotemporal dynamics under well controlled experimental perturbation, particularly for human immunity. This review presents a suite of technologies, comprising in vitro, ex vivo and in silico models, developed to study the lymph node or its components. We discuss the use of these tools to model cell behaviors in increasing order of complexity, from cell motility, to cell-cell interactions, to organ-level functions such as vaccination. Next, we identify current challenges regarding cell sourcing and culture, real time measurements of lymph node behavior in vivo and tool development for analysis and control of engineered cultures. Finally, we propose new research directions and offer our perspective on the future of this rapidly growing field. We anticipate that this review will be especially beneficial to immunologists looking to expand their toolkit for probing lymph node structure and function.
Collapse
Affiliation(s)
- Tochukwu Ozulumba
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Alyssa N. Montalbine
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Jennifer E. Ortiz-Cárdenas
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia (UVA) Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
16
|
The structure of biological complexity: Comment on "Networks behind the morphology and structural design of living systems" by Gosak et al. Phys Life Rev 2023; 44:73-76. [PMID: 36543074 DOI: 10.1016/j.plrev.2022.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
|
17
|
Ganusov VV, Zenkov VS, Majumder B. Correlation between speed and turning naturally arises for sparsely sampled cell movements. Phys Biol 2023; 20:10.1088/1478-3975/acb18c. [PMID: 36623315 PMCID: PMC9918869 DOI: 10.1088/1478-3975/acb18c] [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: 09/20/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
Mechanisms regulating cell movement are not fully understood. One feature of cell movement that determines how far cells displace from an initial position is persistence, the ability to perform movements in a direction similar to the previous movement direction. Persistence is thus determined by turning angles (TA) between two sequential displacements and can be characterized by an average TA or persistence time. Recent studies documenting T cell movement in zebrafish found that a cell's average speed and average TA are negatively correlated, suggesting a fundamental cell-intrinsic program whereby cells with a lower TA (and larger persistence time) are intrinsically faster (or faster cells turn less). In this paper we confirm the existence of the correlation between turning and speed for six different datasets on 3D movement of CD8 T cells in murine lymph nodes or liver. Interestingly, the negative correlation between TA and speed was observed in experiments in which liver-localized CD8 T cells rapidly displace due to floating with the blood flow, suggesting that other mechanisms besides cell-intrinsic program may be at play. By simulating correlated random walks using two different frameworks (one based on the von Mises-Fisher (vMF) distribution and another based on the Ornstein-Uhlenbeck (OU) process) we show that the negative correlation between speed and turning naturally arises when cell trajectories are sub-sampled, i.e. when the frequency of sampling is lower than frequency at which cells typically make movements. This effect is strongest when the sampling frequency is of the order of magnitude of the inverse of persistence time of cells and when cells vary in persistence time. The effect arises in part due to the sensitivity of estimated cell speeds to the frequency of imaging whereby less frequent imaging results in slower speeds. Interestingly, by using estimated persistence times for cells in two of our datasets and simulating cell movements using the OU process, we could partially reproduce the experimentally observed correlation between TA and speed without a cell-intrinsic program linking the two processes. Our results thus suggest that sub-sampling may contribute to (and perhaps fully explains) the observed correlation between speed and turning at least for some cell trajectory data and emphasize the role of sampling frequency in the inference of critical cellular parameters of cell motility such as speeds.
Collapse
Affiliation(s)
- Vitaly V. Ganusov
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
- Department of Mathematics, University of Tennessee, Knoxville, TN 37996, USA
| | - Viktor S. Zenkov
- Department of Electrical Engineering and Computer Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Barun Majumder
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
18
|
Fairchild RL. Fibroblastic reticular cells orchestrate long-term graft survival following recipient treatment with CD40 ligand-targeted costimulatory blockade. J Clin Invest 2022; 132:e165174. [PMID: 36519546 PMCID: PMC9753987 DOI: 10.1172/jci165174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Fibroblastic reticular cells (FRCs) maintain the architecture of secondary lymphoid organs, which optimize interactions between antigen-presenting dendritic cells and reactive naive T cells. In this issue of the JCI, Zhao, Jung, and colleagues investigated CD4+FoxP3+ regulatory T cell development and long-term heart allograft survival in recipients treated with peritransplant costimulatory blockade to inhibit CD40/CD40 ligand (CD40L) signaling. Treatment with an anti-CD40L monoclonal antibody (mAb) increased the lymph node (LN) population of Madcam1+ FRCs and altered their transcription profile to express immunoregulatory mediators. Administration of nanoparticles, containing the anti-CD40L mAb and a targeting antibody against high endothelial venules, delivered the treatment into LNs of allograft recipients. Direct LN delivery of the costimulatory blockade allowed decreased dosing and increased the efficacy in extending graft survival. The results provide insights into mechanisms by which FRCs can promote donor-reactive tolerance, and establish a strategy for administering costimulation-blocking reagents that circumvent systemic effects and improve allograft outcomes.
Collapse
|
19
|
Tan Z, Wang L, Li X. Composition and regulation of the immune microenvironment of salivary gland in Sjögren’s syndrome. Front Immunol 2022; 13:967304. [PMID: 36177010 PMCID: PMC9513852 DOI: 10.3389/fimmu.2022.967304] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Primary Sjögren’s syndrome (pSS) is a systemic autoimmune disease characterized by exocrine gland dysfunction and inflammation. Patients often have dry mouth and dry eye symptoms, which seriously affect their lives. Improving dry mouth and eye symptoms has become a common demand from patients. For this reason, researchers have conducted many studies on external secretory glands. In this paper, we summarize recent studies on the salivary glands of pSS patients from the perspective of the immune microenvironment. These studies showed that hypoxia, senescence, and chronic inflammation are the essential characteristics of the salivary gland immune microenvironment. In the SG of pSS, genes related to lymphocyte chemotaxis, antigen presentation, and lymphocyte activation are upregulated. Interferon (IFN)-related genes, DNA methylation, sRNA downregulation, and mitochondrial-related differentially expressed genes are also involved in forming the immune microenvironment of pSS, while multiple signaling pathways are involved in regulation. We further elucidated the regulation of the salivary gland immune microenvironment in pSS and relevant, targeted treatments.
Collapse
|
20
|
Horsnell HL, Tetley RJ, De Belly H, Makris S, Millward LJ, Benjamin AC, Heeringa LA, de Winde CM, Paluch EK, Mao Y, Acton SE. Lymph node homeostasis and adaptation to immune challenge resolved by fibroblast network mechanics. Nat Immunol 2022; 23:1169-1182. [PMID: 35882934 PMCID: PMC9355877 DOI: 10.1038/s41590-022-01272-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 06/15/2022] [Indexed: 11/23/2022]
Abstract
Emergent physical properties of tissues are not readily understood by reductionist studies of their constituent cells. Here, we show molecular signals controlling cellular, physical, and structural properties and collectively determine tissue mechanics of lymph nodes, an immunologically relevant adult tissue. Lymph nodes paradoxically maintain robust tissue architecture in homeostasis yet are continually poised for extensive expansion upon immune challenge. We find that in murine models of immune challenge, cytoskeletal mechanics of a cellular meshwork of fibroblasts determine tissue tension independently of extracellular matrix scaffolds. We determine that C-type lectin-like receptor 2 (CLEC-2)-podoplanin signaling regulates the cell surface mechanics of fibroblasts, providing a mechanically sensitive pathway to regulate lymph node remodeling. Perturbation of fibroblast mechanics through genetic deletion of podoplanin attenuates T cell activation. We find that increased tissue tension through the fibroblastic stromal meshwork is required to trigger the initiation of fibroblast proliferation and restore homeostatic cellular ratios and tissue structure through lymph node expansion.
Collapse
Affiliation(s)
- Harry L Horsnell
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Robert J Tetley
- Tissue Mechanics Group, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Henry De Belly
- Physiological Laboratory, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Spyridon Makris
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Lindsey J Millward
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Agnesska C Benjamin
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Lucas A Heeringa
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Charlotte M de Winde
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Ewa K Paluch
- Physiological Laboratory, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Yanlan Mao
- Tissue Mechanics Group, MRC Laboratory for Molecular Cell Biology, University College London, London, UK
- Institute for the Physics of Living Systems, University College London, London, UK
| | - Sophie E Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, London, UK.
| |
Collapse
|
21
|
Dubreil L, Ledevin M, Hervet C, Menard D, Philippe C, Michel FJ, Larcher T, Meurens F, Bertho N. The Internal Conduit System of the Swine Inverted Lymph Node. Front Immunol 2022; 13:869384. [PMID: 35734172 PMCID: PMC9207403 DOI: 10.3389/fimmu.2022.869384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/12/2022] [Indexed: 12/04/2022] Open
Abstract
Lymph nodes (LN) are the crossroad where naïve lymphocytes, peripheral antigens and antigen presenting cells contact together in order to mount an adaptive immune response. For this purpose, LN are highly organized convergent hubs of blood and lymphatic vessels that, in the case of B lymphocytes, lead to the B cell follicles. Herein take place the selection and maturation of B cell clones producing high affinity antibodies directed against various antigens. Whereas the knowledge on the murine and human LN distribution systems have reached an exquisite precision those last years, the organization of the antigens and cells circulation into the inverted porcine LN remains poorly described. Using up to date microscopy tools, we described the complex interconnections between afferent lymphatics and blood vessels, perifollicular macrophages, follicular B cells and efferent blood vessels. We observed that afferent lymphatic sinuses presented an asymmetric Lyve-1 expression similar to the one observed in murine LN, whereas specialized perifollicular sinuses connect the main afferent lymphatic sinus to the B cell follicles. Finally, whereas it was long though that mature B cells egress from the inverted LN in the T cell zone through HEV, our observations are in agreement with mature B cells accessing the efferent blood circulation in the efferent, subcapsular area. This understanding of the inverted porcine LN circuitry will allow a more accurate exploration of swine pathogens interactions with the immune cells inside the LN structures. Moreover, the mix between similarities and differences of porcine inverted LN circuitry with mouse and human normal LN shall enable to better apprehend the functions and malfunctions of normal LN from a new perspective.
Collapse
Affiliation(s)
| | | | | | | | - Claire Philippe
- APEX, PAnTher, INRAE, Oniris, Nantes, France
- BIOEPAR, INRAE, Oniris, Nantes, France
| | | | | | - François Meurens
- BIOEPAR, INRAE, Oniris, Nantes, France
- Department of Veterinary Microbiology and Immunology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Nicolas Bertho
- BIOEPAR, INRAE, Oniris, Nantes, France
- *Correspondence: Nicolas Bertho,
| |
Collapse
|
22
|
Antonioli L, Haskó G. May be adenosine an immuno-quorum-sensing signal? Purinergic Signal 2022; 18:205-209. [PMID: 35501535 PMCID: PMC9123119 DOI: 10.1007/s11302-022-09866-2] [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: 02/21/2022] [Accepted: 04/21/2022] [Indexed: 10/18/2022] Open
Abstract
Quorum sensing indicates a communication process between bacteria based on a coordinate variation in gene expression aimed at coordinating a collective comportment related to the bacterial population density. Increasing pieces of evidence pointed out that a quorum-sensing system can be a regulatory program also used in the immune field to organize the density of the various immune cell populations and to calibrate their responses. In particular, such equilibrium is achieved by the ability of immune cells to perceive the density of their own populations or those of other cells in their environment, through the release of several mediators able to finely shape the cell density via coordinated changes in gene expression and protein signaling. In this regard, adenosine displays the typical characteristics of a mediator involved in the regulation of quorum sensing, thus suggesting a putative role of this nucleoside in shaping the balance between diverse immune cell populations.
Collapse
Affiliation(s)
- Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
23
|
Shou Y, Johnson SC, Quek YJ, Li X, Tay A. Integrative lymph node-mimicking models created with biomaterials and computational tools to study the immune system. Mater Today Bio 2022; 14:100269. [PMID: 35514433 PMCID: PMC9062348 DOI: 10.1016/j.mtbio.2022.100269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/16/2022] [Accepted: 04/18/2022] [Indexed: 11/17/2022] Open
Abstract
The lymph node (LN) is a vital organ of the lymphatic and immune system that enables timely detection, response, and clearance of harmful substances from the body. Each LN comprises of distinct substructures, which host a plethora of immune cell types working in tandem to coordinate complex innate and adaptive immune responses. An improved understanding of LN biology could facilitate treatment in LN-associated pathologies and immunotherapeutic interventions, yet at present, animal models, which often have poor physiological relevance, are the most popular experimental platforms. Emerging biomaterial engineering offers powerful alternatives, with the potential to circumvent limitations of animal models, for in-depth characterization and engineering of the lymphatic and adaptive immune system. In addition, mathematical and computational approaches, particularly in the current age of big data research, are reliable tools to verify and complement biomaterial works. In this review, we first discuss the importance of lymph node in immunity protection followed by recent advances using biomaterials to create in vitro/vivo LN-mimicking models to recreate the lymphoid tissue microstructure and microenvironment, as well as to describe the related immuno-functionality for biological investigation. We also explore the great potential of mathematical and computational models to serve as in silico supports. Furthermore, we suggest how both in vitro/vivo and in silico approaches can be integrated to strengthen basic patho-biological research, translational drug screening and clinical personalized therapies. We hope that this review will promote synergistic collaborations to accelerate progress of LN-mimicking systems to enhance understanding of immuno-complexity.
Collapse
Key Words
- ABM, agent-based model
- APC, antigen-presenting cell
- BV, blood vessel
- Biomaterials
- CPM, Cellular Potts model
- Computational models
- DC, dendritic cell
- ECM, extracellular matrix
- FDC, follicular dendritic cell
- FRC, fibroblastic reticular cell
- Immunotherapy
- LEC, lymphatic endothelial cell
- LN, lymph node
- LV, lymphatic vessel
- Lymph node
- Lymphatic system
- ODE, ordinary differential equation
- PDE, partial differential equation
- PDMS, polydimethylsiloxane
Collapse
Affiliation(s)
- Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Sarah C. Johnson
- Department of Bioengineering, Stanford University, CA, 94305, USA
- Department of Bioengineering, Imperial College London, South Kensington, SW72AZ, UK
| | - Ying Jie Quek
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research, 138648, Singapore
| | - Xianlei Li
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
| |
Collapse
|
24
|
Networks behind the morphology and structural design of living systems. Phys Life Rev 2022; 41:1-21. [DOI: 10.1016/j.plrev.2022.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/04/2022] [Indexed: 01/06/2023]
|
25
|
Rajakaruna H, O'Connor JH, Cockburn IA, Ganusov VV. Liver Environment-Imposed Constraints Diversify Movement Strategies of Liver-Localized CD8 T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1292-1304. [PMID: 35131868 PMCID: PMC9250760 DOI: 10.4049/jimmunol.2100842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/17/2021] [Indexed: 05/11/2023]
Abstract
Pathogen-specific CD8 T cells face the problem of finding rare cells that present their cognate Ag either in the lymph node or in infected tissue. Although quantitative details of T cell movement strategies in some tissues such as lymph nodes or skin have been relatively well characterized, we still lack quantitative understanding of T cell movement in many other important tissues, such as the spleen, lung, liver, and gut. We developed a protocol to generate stable numbers of liver-located CD8 T cells, used intravital microscopy to record movement patterns of CD8 T cells in livers of live mice, and analyzed these and previously published data using well-established statistical and computational methods. We show that, in most of our experiments, Plasmodium-specific liver-localized CD8 T cells perform correlated random walks characterized by transiently superdiffusive displacement with persistence times of 10-15 min that exceed those observed for T cells in lymph nodes. Liver-localized CD8 T cells typically crawl on the luminal side of liver sinusoids (i.e., are in the blood); simulating T cell movement in digital structures derived from the liver sinusoids illustrates that liver structure alone is sufficient to explain the relatively long superdiffusive displacement of T cells. In experiments when CD8 T cells in the liver poorly attach to the sinusoids (e.g., 1 wk after immunization with radiation-attenuated Plasmodium sporozoites), T cells also undergo Lévy flights: large displacements occurring due to cells detaching from the endothelium, floating with the blood flow, and reattaching at another location. Our analysis thus provides quantitative details of movement patterns of liver-localized CD8 T cells and illustrates how structural and physiological details of the tissue may impact T cell movement patterns.
Collapse
Affiliation(s)
| | - James H O'Connor
- Division of Immunology, Inflammation and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australia; and
- Australian National University Medical School, Acton, Australian Capital Territory, Australia
| | - Ian A Cockburn
- Division of Immunology, Inflammation and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australia; and
| | - Vitaly V Ganusov
- Department of Microbiology, University of Tennessee, Knoxville, TN;
| |
Collapse
|
26
|
Makris S, de Winde CM, Horsnell HL, Cantoral-Rebordinos JA, Finlay RE, Acton SE. Immune function and dysfunction are determined by lymphoid tissue efficacy. Dis Model Mech 2022; 15:dmm049256. [PMID: 35072206 PMCID: PMC8807573 DOI: 10.1242/dmm.049256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Lymphoid tissue returns to a steady state once each immune response is resolved, and although this occurs multiple times throughout life, its structural integrity and functionality remain unaffected. Stromal cells orchestrate cellular interactions within lymphoid tissue, and any changes to the microenvironment can have detrimental outcomes and drive disease. A breakdown in lymphoid tissue homeostasis can lead to a loss of tissue structure and function that can cause aberrant immune responses. This Review highlights recent advances in our understanding of lymphoid tissue function and remodelling in adaptive immunity and in disease states. We discuss the functional role of lymphoid tissue in disease progression and explore the changes to lymphoid tissue structure and function driven by infection, chronic inflammatory conditions and cancer. Understanding the role of lymphoid tissues in immune responses to a wide range of pathologies allows us to take a fuller systemic view of disease progression.
Collapse
Affiliation(s)
- Spyridon Makris
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Charlotte M. de Winde
- Department for Molecular Cell Biology and Immunology, Amsterdam UMC, location VUmc, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands
| | - Harry L. Horsnell
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Jesús A. Cantoral-Rebordinos
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Rachel E. Finlay
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester M13 9PL, UK
| | - Sophie E. Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
27
|
Isringhausen S, Mun Y, Kovtonyuk L, Kräutler NJ, Suessbier U, Gomariz A, Spaltro G, Helbling PM, Wong HC, Nagasawa T, Manz MG, Oxenius A, Nombela-Arrieta C. Chronic viral infections persistently alter marrow stroma and impair hematopoietic stem cell fitness. J Exp Med 2021; 218:e20192070. [PMID: 34709350 PMCID: PMC8558839 DOI: 10.1084/jem.20192070] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 08/11/2021] [Accepted: 10/05/2021] [Indexed: 11/04/2022] Open
Abstract
Chronic viral infections are associated with hematopoietic suppression, bone marrow (BM) failure, and hematopoietic stem cell (HSC) exhaustion. However, how persistent viral challenge and inflammatory responses target BM tissues and perturb hematopoietic competence remains poorly understood. Here, we combine functional analyses with advanced 3D microscopy to demonstrate that chronic infection with lymphocytic choriomeningitis virus leads to (1) long-lasting decimation of the BM stromal network of mesenchymal CXCL12-abundant reticular cells, (2) proinflammatory transcriptional remodeling of remaining components of this key niche subset, and (3) durable functional defects and decreased competitive fitness in HSCs. Mechanistically, BM immunopathology is elicited by virus-specific, activated CD8 T cells, which accumulate in the BM via interferon-dependent mechanisms. Combined antibody-mediated inhibition of type I and II IFN pathways completely preempts degeneration of CARc and protects HSCs from chronic dysfunction. Hence, viral infections and ensuing immune reactions durably impact BM homeostasis by persistently decreasing the competitive fitness of HSCs and disrupting essential stromal-derived, hematopoietic-supporting cues.
Collapse
Affiliation(s)
- Stephan Isringhausen
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - YeVin Mun
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Larisa Kovtonyuk
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | | | - Ute Suessbier
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Alvaro Gomariz
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Gianluca Spaltro
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Patrick M. Helbling
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Hui Chyn Wong
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Takashi Nagasawa
- Department of Microbiology and Immunology, Osaka University, Osaka, Japan
| | - Markus G. Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | | | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
28
|
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: 8] [Impact Index Per Article: 2.0] [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
|
29
|
Johnson SC, Frattolin J, Edgar LT, Jafarnejad M, Moore Jr JE. Lymph node swelling combined with temporary effector T cell retention aids T cell response in a model of adaptive immunity. J R Soc Interface 2021; 18:20210464. [PMID: 34847790 PMCID: PMC8633806 DOI: 10.1098/rsif.2021.0464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 11/02/2021] [Indexed: 12/19/2022] Open
Abstract
Swelling of lymph nodes (LNs) is commonly observed during the adaptive immune response, yet the impact on T cell (TC) trafficking and subsequent immune response is not well known. To better understand the effect of macro-scale alterations, we developed an agent-based model of the LN paracortex, describing the TC proliferative response to antigen-presenting dendritic cells alongside inflammation-driven and swelling-induced changes in TC recruitment and egress, while also incorporating regulation of the expression of egress-modulating TC receptor sphingosine-1-phosphate receptor-1. Analysis of the effector TC response under varying swelling conditions showed that swelling consistently aided TC activation. However, subsequent effector CD8+ TC production was reduced in scenarios where swelling occurred too early in the TC proliferative phase or when TC cognate frequency was low due to increased opportunity for TC exit. Temporarily extending retention of newly differentiated effector TCs, mediated by sphingosine-1-phosphate receptor-1 expression, mitigated any negative effects of swelling by allowing facilitation of activation to outweigh increased access to exit areas. These results suggest that targeting temporary effector TC retention and egress associated with swelling offers new ways to modulate effector TC responses in, for example, immuno-suppressed patients and to optimize of vaccine design.
Collapse
Affiliation(s)
- Sarah C. Johnson
- Department of Bioengineering, Imperial College London, London, UK
| | | | - Lowell T. Edgar
- Department of Bioengineering, Imperial College London, London, UK
| | - Mohammad Jafarnejad
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
30
|
Abstract
The lymph node (LN) represents a key structural component of the lymphatic system network responsible for the fluid balance in tissues and the immune system functioning. Playing an important role in providing the immune defense of the host organism, LNs can also contribute to the progression of pathological processes, e.g., the spreading of cancer cells. To gain a deeper understanding of the transport function of LNs, experimental approaches are used. Mathematical modeling of the fluid transport through the LN represents a complementary tool for studying the LN functioning under broadly varying physiological conditions. We developed an artificial neural network (NN) model to describe the lymph node drainage function. The NN model predicts the flow characteristics through the LN, including the exchange with the blood vascular systems in relation to the boundary and lymphodynamic conditions, such as the afferent lymph flow, Darcy’s law constants and Starling’s equation parameters. The model is formulated as a feedforward NN with one hidden layer. The NN complements the computational physics-based model of a stationary fluid flow through the LN and the fluid transport across the blood vessel system of the LN. The physical model is specified as a system of boundary integral equations (IEs) equivalent to the original partial differential equations (PDEs; Darcy’s Law and Starling’s equation) formulations. The IE model has been used to generate the training dataset for identifying the NN model architecture and parameters. The computation of the output LN drainage function characteristics (the fluid flow parameters and the exchange with blood) with the trained NN model required about 1000-fold less central processing unit (CPU) time than computationally tracing the flow characteristics of interest with the physics-based IE model. The use of the presented computational models will allow for a more realistic description and prediction of the immune cell circulation, cytokine distribution and drug pharmacokinetics in humans under various health and disease states as well as assisting in the development of artificial LN-on-a-chip technologies.
Collapse
|
31
|
Poirot J, Medvedovic J, Trichot C, Soumelis V. Compartmentalized multicellular crosstalk in lymph nodes coordinates the generation of potent cellular and humoral immune responses. Eur J Immunol 2021; 51:3146-3160. [PMID: 34606627 PMCID: PMC9298410 DOI: 10.1002/eji.202048977] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/13/2021] [Accepted: 09/22/2021] [Indexed: 12/24/2022]
Abstract
Distributed throughout the body, lymph nodes (LNs) constitute an important crossroad where resident and migratory immune cells interact to initiate antigen‐specific immune responses supported by a dynamic 3‐dimensional network of stromal cells, that is, endothelial cells and fibroblastic reticular cells (FRCs). LNs are organized into four major subanatomically separated compartments: the subcapsular sinus (SSC), the paracortex, the cortex, and the medulla. Each compartment is underpinned by particular FRC subsets that physically support LN architecture and delineate functional immune niches by appropriately providing environmental cues, nutrients, and survival factors to the immune cell subsets they interact with. In this review, we discuss how FRCs drive the structural and functional organization of each compartment to give rise to prosperous interactions and coordinate immune cell activities. We also discuss how reciprocal communication makes FRCs and immune cells perfect compatible partners for the generation of potent cellular and humoral immune responses.
Collapse
Affiliation(s)
- Justine Poirot
- Université de Paris, INSERM U976, Paris, France.,Université Paris-Saclay, Saint Aubin, France
| | | | | | - Vassili Soumelis
- Université de Paris, INSERM U976, Paris, France.,AP-HP, Hôpital Saint-Louis, Laboratoire d'Immunologie-Histocompatibilité, Paris, France
| |
Collapse
|
32
|
Acton SE, Onder L, Novkovic M, Martinez VG, Ludewig B. Communication, construction, and fluid control: lymphoid organ fibroblastic reticular cell and conduit networks. Trends Immunol 2021; 42:782-794. [PMID: 34362676 DOI: 10.1016/j.it.2021.07.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 01/16/2023]
Abstract
Fibroblastic reticular cells (FRCs) are a crucial part of the stromal cell infrastructure of secondary lymphoid organs (SLOs). Lymphoid organ fibroblasts form specialized niches for immune cell interactions and thereby govern lymphocyte activation and differentiation. Moreover, FRCs produce and ensheath a network of extracellular matrix (ECM) microfibers called the conduit system. FRC-generated conduits contribute to fluid and immune cell control by funneling fluids containing antigens and inflammatory mediators through the SLOs. We review recent progress in FRC biology that has advanced our understanding of immune cell functions and interactions. We discuss the intricate relationships between the cellular FRC and the fibrillar conduit networks, which together form the basis for efficient communication between immune cells and the tissues they survey.
Collapse
Affiliation(s)
- Sophie E Acton
- Stromal Immunology Group, Medical Research Council (MRC) Laboratory for Molecular Cell Biology, University College London, London, UK.
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Mario Novkovic
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Victor G Martinez
- Molecular Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.
| |
Collapse
|
33
|
Cupovic J, Ring SS, Onder L, Colston JM, Lütge M, Cheng HW, De Martin A, Provine NM, Flatz L, Oxenius A, Scandella E, Krebs P, Engeler D, Klenerman P, Ludewig B. Adenovirus vector vaccination reprograms pulmonary fibroblastic niches to support protective inflating memory CD8 + T cells. Nat Immunol 2021; 22:1042-1051. [PMID: 34267375 PMCID: PMC7611414 DOI: 10.1038/s41590-021-00969-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 06/07/2021] [Indexed: 02/07/2023]
Abstract
Pathogens and vaccines that produce persisting antigens can generate expanded pools of effector memory CD8+ T cells, described as memory inflation. While properties of inflating memory CD8+ T cells have been characterized, the specific cell types and tissue factors responsible for their maintenance remain elusive. Here, we show that clinically applied adenovirus vectors preferentially target fibroblastic stromal cells in cultured human tissues. Moreover, we used cell-type-specific antigen targeting to define critical cells and molecules that sustain long-term antigen presentation and T cell activity after adenovirus vector immunization in mice. While antigen targeting to myeloid cells was insufficient to activate antigen-specific CD8+ T cells, genetic activation of antigen expression in Ccl19-cre-expressing fibroblastic stromal cells induced inflating CD8+ T cells. Local ablation of vector-targeted cells revealed that lung fibroblasts support the protective function and metabolic fitness of inflating memory CD8+ T cells in an interleukin (IL)-33-dependent manner. Collectively, these data define a critical fibroblastic niche that underpins robust protective immunity operating in a clinically important vaccine platform.
Collapse
Affiliation(s)
- Jovana Cupovic
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany
| | - Sandra S Ring
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Julia M Colston
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mechthild Lütge
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Hung-Wei Cheng
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Angelina De Martin
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Nicholas M Provine
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | | | - Elke Scandella
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Philippe Krebs
- Institute of Pathology, University of Berne, Berne, Switzerland
| | - Daniel Engeler
- Department of Urology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
34
|
de Winde CM, Makris S, Millward LJ, Cantoral-Rebordinos JA, Benjamin AC, Martínez VG, Acton SE. Fibroblastic reticular cell response to dendritic cells requires coordinated activity of podoplanin, CD44 and CD9. J Cell Sci 2021; 134:jcs258610. [PMID: 34184727 PMCID: PMC8325952 DOI: 10.1242/jcs.258610] [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: 03/04/2021] [Accepted: 06/22/2021] [Indexed: 12/18/2022] Open
Abstract
In adaptive immunity, CLEC-2+ dendritic cells (DCs) contact fibroblastic reticular cells (FRCs) inhibiting podoplanin-dependent actomyosin contractility, permitting FRC spreading and lymph node expansion. The molecular mechanisms controlling lymph node remodelling are incompletely understood. We asked how podoplanin is regulated on FRCs in the early phase of lymph node expansion, and which other proteins are required for the FRC response to DCs. We find that podoplanin and its partner proteins CD44 and CD9 are differentially expressed by specific lymph node stromal populations in vivo, and their expression in FRCs is coregulated by CLEC-2 (encoded by CLEC1B). Both CD44 and CD9 suppress podoplanin-dependent contractility. We find that beyond contractility, podoplanin is required for FRC polarity and alignment. Independently of podoplanin, CD44 and CD9 affect FRC-FRC interactions. Furthermore, our data show that remodelling of the FRC cytoskeleton in response to DCs is a two-step process requiring podoplanin partner proteins CD44 and CD9. Firstly, CLEC-2 and podoplanin binding inhibits FRC contractility, and, secondly, FRCs form protrusions and spread, which requires both CD44 and CD9. Together, we show a multi-faceted FRC response to DCs, which requires CD44 and CD9 in addition to podoplanin.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sophie E. Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
35
|
Lütge M, Pikor NB, Ludewig B. Differentiation and activation of fibroblastic reticular cells. Immunol Rev 2021; 302:32-46. [PMID: 34046914 PMCID: PMC8361914 DOI: 10.1111/imr.12981] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/17/2021] [Accepted: 04/30/2021] [Indexed: 12/29/2022]
Abstract
Secondary lymphoid organs (SLO) are underpinned by fibroblastic reticular cells (FRC) that form dedicated microenvironmental niches to secure induction and regulation of innate and adaptive immunity. Distinct FRC subsets are strategically positioned in SLOs to provide niche factors and govern efficient immune cell interaction. In recent years, the use of specialized mouse models in combination with single-cell transcriptomics has facilitated the elaboration of the molecular FRC landscape at an unprecedented resolution. While single-cell RNA-sequencing has advanced the resolution of FRC subset characterization and function, the high dimensionality of the generated data necessitates careful analysis and validation. Here, we reviewed novel findings from high-resolution transcriptomic analyses that refine our understanding of FRC differentiation and activation processes in the context of infection and inflammation. We further discuss concepts, strategies, and limitations for the analysis of single-cell transcriptome data from FRCs and the wide-ranging implications for our understanding of stromal cell biology.
Collapse
Affiliation(s)
- Mechthild Lütge
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Natalia B Pikor
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, St. Gallen, Switzerland.,Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
36
|
Joy DA, Libby ARG, McDevitt TC. Deep neural net tracking of human pluripotent stem cells reveals intrinsic behaviors directing morphogenesis. Stem Cell Reports 2021; 16:1317-1330. [PMID: 33979602 PMCID: PMC8185472 DOI: 10.1016/j.stemcr.2021.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 01/09/2023] Open
Abstract
Lineage tracing is a powerful tool in developmental biology to interrogate the evolution of tissue formation, but the dense, three-dimensional nature of tissue limits the assembly of individual cell trajectories into complete reconstructions of development. Human induced pluripotent stem cells (hiPSCs) can recapitulate aspects of developmental processes, providing an in vitro platform to assess the dynamic collective behaviors directing tissue morphogenesis. Here, we trained an ensemble of neural networks to track individual hiPSCs in time-lapse microscopy, generating longitudinal measures of cell and cellular neighborhood properties on timescales from minutes to days. Our analysis reveals that, while individual cell parameters are not strongly affected by pluripotency maintenance conditions or morphogenic cues, regional changes in cell behavior predict cell fate and colony organization. By generating complete multicellular reconstructions of hiPSC behavior, our tracking pipeline enables fine-grained understanding of morphogenesis by elucidating the role of regional behavior in early tissue formation.
Collapse
Affiliation(s)
- David A Joy
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, San Francisco, CA, USA; Gladstone Institutes, San Francisco, CA, USA
| | - Ashley R G Libby
- Gladstone Institutes, San Francisco, CA, USA; Developmental and Stem Cell Biology PhD Program, University of California, San Francisco, San Francisco, CA, USA
| | - Todd C McDevitt
- Gladstone Institutes, San Francisco, CA, USA; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
37
|
Ferreira BO, Gamarra LF, Nucci MP, Oliveira FA, Rego GNA, Marti L. LN-Derived Fibroblastic Reticular Cells and Their Impact on T Cell Response—A Systematic Review. Cells 2021; 10:1150. [DOI: https:/doi.org/10.3390/cells10051150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Fibroblastic reticular cells (FRCs), usually found and isolated from the T cell zone of lymph nodes, have recently been described as much more than simple structural cells. Originally, these cells were described to form a conduit system called the “reticular fiber network” and for being responsible for transferring the lymph fluid drained from tissues through afferent lymphatic vessels to the T cell zone. However, nowadays, these cells are described as being capable of secreting several cytokines and chemokines and possessing the ability to interfere with the immune response, improving it, and also controlling lymphocyte proliferation. Here, we performed a systematic review of the several methods employed to investigate the mechanisms used by fibroblastic reticular cells to control the immune response, as well as their ability in determining the fate of T cells. We searched articles indexed and published in the last five years, between 2016 and 2020, in PubMed, Scopus, and Cochrane, following the PRISMA guidelines. We found 175 articles published in the literature using our searching strategies, but only 24 articles fulfilled our inclusion criteria and are discussed here. Other articles important in the built knowledge of FRCs were included in the introduction and discussion. The studies selected for this review used different strategies in order to access the contribution of FRCs to different mechanisms involved in the immune response: 21% evaluated viral infection in this context, 13% used a model of autoimmunity, 8% used a model of GvHD or cancer, 4% used a model of Ischemic-reperfusion injury (IRI). Another four studies just targeted a particular signaling pathway, such as MHC II expression, FRC microvesicles, FRC secretion of IL-15, FRC network, or ablation of the lysophosphatidic acid (LPA)-producing ectoenzyme autotaxin. In conclusion, our review shows the strategies used by several studies to isolate and culture fibroblastic reticular cells, the models chosen by each one, and dissects their main findings and implications in homeostasis and disease.
Collapse
|
38
|
LN-Derived Fibroblastic Reticular Cells and Their Impact on T Cell Response-A Systematic Review. Cells 2021; 10:cells10051150. [PMID: 34068712 PMCID: PMC8151444 DOI: 10.3390/cells10051150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/11/2021] [Accepted: 04/23/2021] [Indexed: 12/27/2022] Open
Abstract
Fibroblastic reticular cells (FRCs), usually found and isolated from the T cell zone of lymph nodes, have recently been described as much more than simple structural cells. Originally, these cells were described to form a conduit system called the “reticular fiber network” and for being responsible for transferring the lymph fluid drained from tissues through afferent lymphatic vessels to the T cell zone. However, nowadays, these cells are described as being capable of secreting several cytokines and chemokines and possessing the ability to interfere with the immune response, improving it, and also controlling lymphocyte proliferation. Here, we performed a systematic review of the several methods employed to investigate the mechanisms used by fibroblastic reticular cells to control the immune response, as well as their ability in determining the fate of T cells. We searched articles indexed and published in the last five years, between 2016 and 2020, in PubMed, Scopus, and Cochrane, following the PRISMA guidelines. We found 175 articles published in the literature using our searching strategies, but only 24 articles fulfilled our inclusion criteria and are discussed here. Other articles important in the built knowledge of FRCs were included in the introduction and discussion. The studies selected for this review used different strategies in order to access the contribution of FRCs to different mechanisms involved in the immune response: 21% evaluated viral infection in this context, 13% used a model of autoimmunity, 8% used a model of GvHD or cancer, 4% used a model of Ischemic-reperfusion injury (IRI). Another four studies just targeted a particular signaling pathway, such as MHC II expression, FRC microvesicles, FRC secretion of IL-15, FRC network, or ablation of the lysophosphatidic acid (LPA)-producing ectoenzyme autotaxin. In conclusion, our review shows the strategies used by several studies to isolate and culture fibroblastic reticular cells, the models chosen by each one, and dissects their main findings and implications in homeostasis and disease.
Collapse
|
39
|
Novkovic M, Onder L, Bocharov G, Ludewig B. Topological Structure and Robustness of the Lymph Node Conduit System. Cell Rep 2021; 30:893-904.e6. [PMID: 31968261 DOI: 10.1016/j.celrep.2019.12.070] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 11/26/2019] [Accepted: 12/18/2019] [Indexed: 02/07/2023] Open
Abstract
Fibroblastic reticular cells (FRCs) form a road-like cellular network in lymph nodes (LNs) that provides essential chemotactic, survival, and regulatory signals for immune cells. While the topological characteristics of the FRC network have been elaborated, the network properties of the micro-tubular conduit system generated by FRCs, which drains lymph fluid through a pipeline-like system to distribute small molecules and antigens, has remained unexplored. Here, we quantify the crucial 3D morphometric parameters and determine the topological properties governing the structural organization of the intertwined networks. We find that the conduit system exhibits lesser small-worldness and lower resilience to perturbation compared to the FRC network, while the robust topological organization of both networks is maintained in a lymphotoxin-β-receptor-independent manner. Overall, the high-resolution topological analysis of the "roads-and-pipes" networks highlights essential parameters underlying the functional organization of LN micro-environments and will, hence, advance the development of multi-scale LN models.
Collapse
Affiliation(s)
- Mario Novkovic
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Gennady Bocharov
- Marchuk Institute of Numerical Mathematics, Russian Academy of Sciences, Moscow 119333, Russia; Institute for Personalized Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland.
| |
Collapse
|
40
|
Dertschnig S, Evans P, Santos E Sousa P, Manzo T, Ferrer IR, Stauss HJ, Bennett CL, Chakraverty R. Graft-versus-host disease reduces lymph node display of tissue-restricted self-antigens and promotes autoimmunity. J Clin Invest 2020; 130:1896-1911. [PMID: 31917684 PMCID: PMC7108931 DOI: 10.1172/jci133102] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/03/2020] [Indexed: 12/28/2022] Open
Abstract
Acute graft-versus-host disease (GVHD) is initially triggered by alloreactive T cells, which damage peripheral tissues and lymphoid organs. Subsequent transition to chronic GVHD involves the emergence of autoimmunity, although the underlying mechanisms driving this process are unclear. Here, we tested the hypothesis that acute GVHD blocks peripheral tolerance of autoreactive T cells by impairing lymph node (LN) display of peripheral tissue–restricted antigens (PTAs). At the initiation of GVHD, LN fibroblastic reticular cells (FRCs) rapidly reduced expression of genes regulated by DEAF1, an autoimmune regulator-like transcription factor required for intranodal expression of PTAs. Subsequently, GVHD led to the selective elimination of the FRC population, and blocked the repair pathways required for its regeneration. We used a transgenic mouse model to show that the loss of presentation of an intestinal PTA by FRCs during GVHD resulted in the activation of autoaggressive T cells and gut injury. Finally, we show that FRCs normally expressed a unique PTA gene signature that was highly enriched for genes expressed in the target organs affected by chronic GVHD. In conclusion, acute GVHD damages and prevents repair of the FRC network, thus disabling an essential platform for purging autoreactive T cells from the repertoire.
Collapse
Affiliation(s)
- Simone Dertschnig
- UCL Cancer Institute, and.,Institute of Immunity and Transplantation, London, United Kingdom
| | - Pamela Evans
- UCL Cancer Institute, and.,Institute of Immunity and Transplantation, London, United Kingdom
| | - Pedro Santos E Sousa
- UCL Cancer Institute, and.,Institute of Immunity and Transplantation, London, United Kingdom
| | | | - Ivana R Ferrer
- UCL Cancer Institute, and.,Institute of Immunity and Transplantation, London, United Kingdom
| | - Hans J Stauss
- Institute of Immunity and Transplantation, London, United Kingdom
| | - Clare L Bennett
- UCL Cancer Institute, and.,Institute of Immunity and Transplantation, London, United Kingdom
| | - Ronjon Chakraverty
- UCL Cancer Institute, and.,Institute of Immunity and Transplantation, London, United Kingdom
| |
Collapse
|
41
|
Tfh Cells in Health and Immunity: Potential Targets for Systems Biology Approaches to Vaccination. Int J Mol Sci 2020; 21:ijms21228524. [PMID: 33198297 PMCID: PMC7696930 DOI: 10.3390/ijms21228524] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/16/2022] Open
Abstract
T follicular helper (Tfh) cells are a specialised subset of CD4+ T cells that play a significant role in the adaptive immune response, providing critical help to B cells within the germinal centres (GC) of secondary lymphoid organs. The B cell receptors of GC B cells undergo multiple rounds of somatic hypermutation and affinity maturation within the GC response, a process dependent on cognate interactions with Tfh cells. B cells that receive sufficient help from Tfh cells form antibody-producing long-lived plasma and memory B cells that provide the basis of decades of effective and efficient protection and are considered the gold standard in correlates of protection post-vaccination. However, the T cell response to vaccination has been understudied, and over the last 10 years, exponential improvements in the technological underpinnings of sampling techniques, experimental and analytical tools have allowed multidisciplinary characterisation of the role of T cells and the immune system as a whole. Of particular interest to the field of vaccinology are GCs and Tfh cells, representing a unique target for improving immunisation strategies. Here, we discuss recent insights into the unique journey of Tfh cells from thymus to lymph node during differentiation and their role in the production of high-quality antibody responses as well as their journey back to the periphery as a population of memory cells. Further, we explore their function in health and disease and the power of next-generation sequencing techniques to uncover their potential as modulators of vaccine-induced immunity.
Collapse
|
42
|
Cosgrove J, Novkovic M, Albrecht S, Pikor NB, Zhou Z, Onder L, Mörbe U, Cupovic J, Miller H, Alden K, Thuery A, O'Toole P, Pinter R, Jarrett S, Taylor E, Venetz D, Heller M, Uguccioni M, Legler DF, Lacey CJ, Coatesworth A, Polak WG, Cupedo T, Manoury B, Thelen M, Stein JV, Wolf M, Leake MC, Timmis J, Ludewig B, Coles MC. B cell zone reticular cell microenvironments shape CXCL13 gradient formation. Nat Commun 2020; 11:3677. [PMID: 32699279 PMCID: PMC7376062 DOI: 10.1038/s41467-020-17135-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 03/12/2020] [Indexed: 02/07/2023] Open
Abstract
Through the formation of concentration gradients, morphogens drive graded responses to extracellular signals, thereby fine-tuning cell behaviors in complex tissues. Here we show that the chemokine CXCL13 forms both soluble and immobilized gradients. Specifically, CXCL13+ follicular reticular cells form a small-world network of guidance structures, with computer simulations and optimization analysis predicting that immobilized gradients created by this network promote B cell trafficking. Consistent with this prediction, imaging analysis show that CXCL13 binds to extracellular matrix components in situ, constraining its diffusion. CXCL13 solubilization requires the protease cathepsin B that cleaves CXCL13 into a stable product. Mice lacking cathepsin B display aberrant follicular architecture, a phenotype associated with effective B cell homing to but not within lymph nodes. Our data thus suggest that reticular cells of the B cell zone generate microenvironments that shape both immobilized and soluble CXCL13 gradients.
Collapse
Affiliation(s)
- Jason Cosgrove
- York Computational Immunology Lab, University of York, York, UK
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, UK
- Department of Electronic Engineering, University of York, York, UK
| | - Mario Novkovic
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Stefan Albrecht
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Natalia B Pikor
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Zhaoukun Zhou
- Department of Biology, University of York, York, UK
- Biological Physical Sciences Institute (BPSI), University of York, York, UK
- Department of Physics, University of York, York, UK
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Urs Mörbe
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Jovana Cupovic
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Helen Miller
- Department of Biology, University of York, York, UK
- Biological Physical Sciences Institute (BPSI), University of York, York, UK
- Department of Physics, University of York, York, UK
| | - Kieran Alden
- York Computational Immunology Lab, University of York, York, UK
- Department of Electronic Engineering, University of York, York, UK
| | - Anne Thuery
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, UK
| | | | - Rita Pinter
- Kennedy Institute of Rheumatology at the University of Oxford, Oxford, UK
| | - Simon Jarrett
- Kennedy Institute of Rheumatology at the University of Oxford, Oxford, UK
| | - Emily Taylor
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, UK
| | - Daniel Venetz
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Manfred Heller
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Mariagrazia Uguccioni
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland
| | - Charles J Lacey
- York Computational Immunology Lab, University of York, York, UK
| | | | - Wojciech G Polak
- Department of Surgery, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Tom Cupedo
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Bénedicte Manoury
- Institut Necker Enfants Malades, INSERM U1151- CNRS UMR 8253, 149 rue de Sèvres 75015 Paris, France Université René Descartes, 75005, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Marlene Wolf
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Mark C Leake
- Department of Biology, University of York, York, UK.
- Biological Physical Sciences Institute (BPSI), University of York, York, UK.
- Department of Physics, University of York, York, UK.
| | - Jon Timmis
- York Computational Immunology Lab, University of York, York, UK.
- Department of Electronic Engineering, University of York, York, UK.
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.
| | - Mark C Coles
- York Computational Immunology Lab, University of York, York, UK.
- Kennedy Institute of Rheumatology at the University of Oxford, Oxford, UK.
| |
Collapse
|
43
|
Tissue-Engineered Stromal Reticula to Study Lymph Node Fibroblastic Reticular Cells in Type I Diabetes. Cell Mol Bioeng 2020; 13:419-434. [PMID: 33184575 PMCID: PMC7596159 DOI: 10.1007/s12195-020-00627-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/12/2020] [Indexed: 01/25/2023] Open
Abstract
Introduction Fibroblastic reticular cells (FRCs) support and remodel the lymph node (LN), express and present self-antigens to T cells to promote tolerance. In Type 1 diabetes (T1D), decrease in FRC frequency and in their expression of T1D-related self-antigens may hinder tolerogenic engagement of autoreactive T cells. FRC reticular organization in LNs is critical for adaptive immunity. Thus, we engineered LN-like FRC reticula to determine if FRC reticular properties were altered in T1D and to study engagement of autoreactive T cells in vitro. Methods We characterized FRC networks in pancreatic and skin-draining LNs of 4- and 12-week old non-obese diabetic (NOD) and diabetes resistant NOR mice by immunofluorescence. Murine FRCs isolated from NOR, NOD or human pancreatic LNs were cultured in collagen sponges for up to 21 days before immunofluorescence and flow cytometry analysis. NOD FRCs expressing T1D antigens were co-cultured with CellTrace-labeled specific T cells in 2D or in scaffolds. T cell engagement was quantified by CD25 upregulation, CellTrace dilution and by T cell tracking. Results FRC networks in both 4- and 12-week old NOD LNs displayed larger reticular pores than NOR controls. NOD FRCs had delayed scaffold remodeling compared to NOR FRCs. Expression of the gp38 FRC marker in NOD FRCs was lower than in NOR but improved in 3D. FRC reticula expressing T1D antigens promoted higher engagement of specific T cells than 2D. Conclusion We engineered LN-like FRC reticula that recapitulate FRC organization and phenotype of T1D LNs for studying tolerogenic autoreactive T cell engagement in T1D.
Collapse
|
44
|
Antonioli L, Fornai M, Pellegrini C, Masi S, Puxeddu I, Blandizzi C. Ectopic Lymphoid Organs and Immune-Mediated Diseases: Molecular Basis for Pharmacological Approaches. Trends Mol Med 2020; 26:1021-1033. [PMID: 32600794 DOI: 10.1016/j.molmed.2020.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/21/2020] [Accepted: 06/04/2020] [Indexed: 12/15/2022]
Abstract
Chronic inflammation is the result a persistent increase in the expression of several proinflammatory pathways with impaired inflammatory resolution. Ectopic lymphoid organs (ELOs), untypical lymphoid annexes, emerge during chronic inflammation and contribute to the physiopathology of chronic inflammatory disorders. This review discusses the pathophysiological role of ELOs in the progression of immune-mediated inflammatory diseases (IMIDs), including multiple sclerosis (MS), rheumatoid arthritis (RA), inflammatory bowel disease (IBD), atherosclerosis, and Sjögren syndrome (SSj). The molecular pathways underlying the emergence of ELOs are of interest for the development of novel pharmacological approaches for the management of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy.
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | | | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Ilaria Puxeddu
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
45
|
Hillmann A, Crane M, Ruskin HJ. Assessing the impact of HIV treatment interruptions using stochastic cellular Automata. J Theor Biol 2020; 502:110376. [PMID: 32574568 DOI: 10.1016/j.jtbi.2020.110376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 04/23/2020] [Accepted: 06/12/2020] [Indexed: 11/30/2022]
Abstract
Chronic HIV infection causes a progressive decrease in the ability to maintain homeostasis resulting, after some time, in eventual break down of immune functions. Recent clinical research has shed light on a significant contribution of the lymphatic tissues, where HIV causes accumulation of collagen, (fibrosis). Specifically, where tissue is populated by certain types of functional stromal cells designated Fibroblastic Reticular Cells (FRCs), these have been found to play a crucial role in balancing out apoptosis and regeneration of naïve T-cells through 2-way cellular signaling. Tissue fibrosis not only impedes this signaling, effectively reducing T-cell levels through increased apoptosis of cells of both T- and FRC type but has been found to be irreversible by current HIV standard treatment (cART). While the therapy aims to block the viral lifecycle, cART-associated increase of T-cell levels in blood appears to conceal existing FRC impairment through fibrosis. This hidden impairment can lead to adverse consequences if treatment is interrupted, e.g. due to poor adherence (missing doses) or through periods recovering from drug toxicities. Formal clinical studies on treatment interruption have indicated possible adverse effects, but quantification of those effects in relation to interruption protocol and patient predisposition remains unclear. Accordingly, the impact of treatment interruption on lymphatic tissue structure and T-cell levels is explored here by means of computer simulation. A novel Stochastic Cellular Automata model is proposed, which utilizes all sources of clinical detail available to us (though sparse in part) for model parametrization. Sources are explicitly referenced and conflicting evidence from previous studies explored. The main focus is on (i) spatial aspects of collagen build up, together with (ii) collagen increase after repeated treatment interruptions to explore the dynamics of HIV-induced fibrosis and T-cell loss.
Collapse
Affiliation(s)
- Andreas Hillmann
- Advanced Research Computing Centre for Complex Systems Modelling, School of Computing, Dublin City University, Dublin, Ireland.
| | - Martin Crane
- Advanced Research Computing Centre for Complex Systems Modelling, School of Computing, Dublin City University, Dublin, Ireland
| | - Heather J Ruskin
- Advanced Research Computing Centre for Complex Systems Modelling, School of Computing, Dublin City University, Dublin, Ireland
| |
Collapse
|
46
|
Fletcher AL, Baker AT, Lukacs-Kornek V, Knoblich K. The fibroblastic T cell niche in lymphoid tissues. Curr Opin Immunol 2020; 64:110-116. [PMID: 32497868 DOI: 10.1016/j.coi.2020.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/19/2020] [Accepted: 04/21/2020] [Indexed: 12/22/2022]
Abstract
Fibroblastic reticular cells (FRCs) are a necessary immunological component for T cell health. These myofibroblasts are specialized for immune cell support and develop in locations where T and B lymphocyte priming occurs, usually secondary lymphoid organs, but also tertiary lymphoid structures and sites of chronic inflammation. This review describes their dual supportive and suppressive functions and emerging evidence on the co-ordination required to balance these competing roles.
Collapse
Affiliation(s)
- Anne L Fletcher
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Australia; Institute of Immunology and Immunotherapy, University of Birmingham, UK.
| | - Alfie T Baker
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Australia
| | - Veronika Lukacs-Kornek
- Institute of Experimental Immunology, Rheinische-Friedrichs-Wilhelms University of Bonn, 53127, Bonn, Germany
| | - Konstantin Knoblich
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Australia; Institute of Immunology and Immunotherapy, University of Birmingham, UK
| |
Collapse
|
47
|
Environmental Restrictions: A New Concept Governing HIV-1 Spread Emerging from Integrated Experimental-Computational Analysis of Tissue-Like 3D Cultures. Cells 2020; 9:cells9051112. [PMID: 32365826 PMCID: PMC7291240 DOI: 10.3390/cells9051112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/22/2022] Open
Abstract
HIV-1 can use cell-free and cell-associated transmission modes to infect new target cells, but how the virus spreads in the infected host remains to be determined. We recently established 3D collagen cultures to study HIV-1 spread in tissue-like environments and applied iterative cycles of experimentation and computation to develop a first in silico model to describe the dynamics of HIV-1 spread in complex tissue. These analyses (i) revealed that 3D collagen environments restrict cell-free HIV-1 infection but promote cell-associated virus transmission and (ii) defined that cell densities in tissue dictate the efficacy of these transmission modes for virus spread. In this review, we discuss, in the context of the current literature, the implications of this study for our understanding of HIV-1 spread in vivo, which aspects of in vivo physiology this integrated experimental-computational analysis takes into account, and how it can be further improved experimentally and in silico.
Collapse
|
48
|
Lymph node stromal cells: cartographers of the immune system. Nat Immunol 2020; 21:369-380. [PMID: 32205888 DOI: 10.1038/s41590-020-0635-3] [Citation(s) in RCA: 193] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/17/2020] [Indexed: 01/03/2023]
Abstract
Lymph nodes (LNs) are strategically positioned at dedicated sites throughout the body to facilitate rapid and efficient immunity. Central to the structural integrity and framework of LNs, and the recruitment and positioning of leukocytes therein, are mesenchymal and endothelial lymph node stromal cells (LNSCs). Advances in the last decade have expanded our understanding and appreciation of LNSC heterogeneity, and the role they play in coordinating immunity has grown rapidly. In this review, we will highlight the functional contributions of distinct stromal cell populations during LN development in maintaining immune homeostasis and tolerance and in the activation and control of immune responses.
Collapse
|
49
|
Liu C, Ma Y, Zhao J, Nussinov R, Zhang YC, Cheng F, Zhang ZK. Computational network biology: Data, models, and applications. PHYSICS REPORTS 2020; 846:1-66. [DOI: 10.1016/j.physrep.2019.12.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
50
|
YAP/TAZ direct commitment and maturation of lymph node fibroblastic reticular cells. Nat Commun 2020; 11:519. [PMID: 31980640 PMCID: PMC6981200 DOI: 10.1038/s41467-020-14293-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 12/31/2019] [Indexed: 02/07/2023] Open
Abstract
Fibroblastic reticular cells (FRCs) are immunologically specialized myofibroblasts of lymphoid organ, and FRC maturation is essential for structural and functional properties of lymph nodes (LNs). Here we show that YAP and TAZ (YAP/TAZ), the final effectors of Hippo signaling, regulate FRC commitment and maturation. Selective depletion of YAP/TAZ in FRCs impairs FRC growth and differentiation and compromises the structural organization of LNs, whereas hyperactivation of YAP/TAZ enhances myofibroblastic characteristics of FRCs and aggravates LN fibrosis. Mechanistically, the interaction between YAP/TAZ and p52 promotes chemokine expression that is required for commitment of FRC lineage prior to lymphotoxin-β receptor (LTβR) engagement, whereas LTβR activation suppresses YAP/TAZ activity for FRC maturation. Our findings thus present YAP/TAZ as critical regulators of commitment and maturation of FRCs, and hold promise for better understanding of FRC-mediated pathophysiologic processes. Fibroblastic reticular cells (FRC) are important for lymph node (LN) structure and function. Here the authors show that the YAP/TAZ complex downstream of Hippo signalling regulates FRC commitment and maturation, with YAP/TAZ deficiency impairing FRC differentiation, while hyperactivation of YAZ/TAZ inducing myofibroblastic FRCs and LN fibrosis.
Collapse
|