1
|
M. S. Barron A, Fabre T, De S. Distinct fibroblast functions associated with fibrotic and immune-mediated inflammatory diseases and their implications for therapeutic development. F1000Res 2024; 13:54. [PMID: 38681509 PMCID: PMC11053351 DOI: 10.12688/f1000research.143472.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/28/2023] [Indexed: 05/01/2024] Open
Abstract
Fibroblasts are ubiquitous cells that can adopt many functional states. As tissue-resident sentinels, they respond to acute damage signals and shape the earliest events in fibrotic and immune-mediated inflammatory diseases. Upon sensing an insult, fibroblasts produce chemokines and growth factors to organize and support the response. Depending on the size and composition of the resulting infiltrate, these activated fibroblasts may also begin to contract or relax thus changing local stiffness within the tissue. These early events likely contribute to the divergent clinical manifestations of fibrotic and immune-mediated inflammatory diseases. Further, distinct changes to the cellular composition and signaling dialogue in these diseases drive progressive fibroblasts specialization. In fibrotic diseases, fibroblasts support the survival, activation and differentiation of myeloid cells, granulocytes and innate lymphocytes, and produce most of the pathogenic extracellular matrix proteins. Whereas, in immune-mediated inflammatory diseases, sequential accumulation of dendritic cells, T cells and B cells programs fibroblasts to support local, destructive adaptive immune responses. Fibroblast specialization has clear implications for the development of effective induction and maintenance therapies for patients with these clinically distinct diseases.
Collapse
Affiliation(s)
- Alexander M. S. Barron
- Inflammation & Immunology Research Unit, Pfizer, Inc., Cambridge, Massachusetts, 02139, USA
| | - Thomas Fabre
- Inflammation & Immunology Research Unit, Pfizer, Inc., Cambridge, Massachusetts, 02139, USA
| | - Saurav De
- Inflammation & Immunology Research Unit, Pfizer, Inc., Cambridge, Massachusetts, 02139, USA
| |
Collapse
|
2
|
Li L, Wu L, Kensiski A, Zhao J, Shirkey MW, Song Y, Piao W, Zhang T, Mei Z, Gavzy SJ, Ma B, Saxena V, Lee YS, Xiong Y, Li X, Fan X, Abdi R, Bromberg JS. FRC transplantation restores lymph node conduit defects in laminin α4-deficient mice. JCI Insight 2023; 8:e167816. [PMID: 37092548 PMCID: PMC10243809 DOI: 10.1172/jci.insight.167816] [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: 12/06/2022] [Accepted: 03/03/2023] [Indexed: 04/25/2023] Open
Abstract
Fibroblastic reticular cells (FRCs) play important roles in tolerance by producing laminin α4 (Lama4) and altering lymph node (LN) structure and function. The present study revealed the specific roles of extracellular matrix Lama4 in regulating LN conduits using FRC-specific KO mouse strains. FRC-derived Lama4 maintained conduit fiber integrity, as its depletion altered conduit morphology and structure and reduced homeostatic conduit flow. Lama4 regulated the lymphotoxin β receptor (LTβR) pathway, which is critical for conduit and LN integrity. Depleting LTβR in FRCs further reduced conduits and impaired reticular fibers. Lama4 was indispensable for FRC generation and survival, as FRCs lacking Lama4 displayed reduced proliferation but upregulated senescence and apoptosis. During acute immunization, FRC Lama4 deficiency increased antigen flow through conduits. Importantly, adoptive transfer of WT FRCs to FRC Lama4-deficient mice rescued conduit structure, ameliorated Treg and chemokine distribution, and restored transplant allograft acceptance, which were all impaired by FRC Lama4 depletion. Single-cell RNA sequencing analysis of LN stromal cells indicated that the laminin and collagen signaling pathways linked crosstalk among FRC subsets and endothelial cells. This study demonstrated that FRC Lama4 is responsible for maintaining conduits by FRCs and can be harnessed to potentiate FRC-based immunomodulation.
Collapse
Affiliation(s)
- Lushen Li
- Department of Surgery, and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Long Wu
- Department of Surgery, and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Allison Kensiski
- Department of Surgery, and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jing Zhao
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marina W. Shirkey
- Department of Surgery, and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Wenji Piao
- Department of Surgery, and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | - Samuel J. Gavzy
- Department of Surgery, and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Bing Ma
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Vikas Saxena
- Department of Surgery, and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Young S. Lee
- Department of Surgery, and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yanbao Xiong
- Department of Surgery, and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Xiaofei Li
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaoxuan Fan
- Flow Cytometry Shared Service, Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan S. Bromberg
- Department of Surgery, and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Vella G, Hua Y, Bergers G. High endothelial venules in cancer: Regulation, function, and therapeutic implication. Cancer Cell 2023; 41:527-545. [PMID: 36827979 DOI: 10.1016/j.ccell.2023.02.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/12/2023] [Accepted: 02/01/2023] [Indexed: 02/25/2023]
Abstract
The lack of sufficient intratumoral CD8+ T lymphocytes is a significant obstacle to effective immunotherapy in cancer. High endothelial venules (HEVs) are organ-specific and specialized postcapillary venules uniquely poised to facilitate the transmigration of lymphocytes to lymph nodes (LNs) and other secondary lymphoid organs (SLOs). HEVs can also form in human and murine cancer (tumor HEVs [TU-HEVs]) and contribute to the generation of diffuse T cell-enriched aggregates or tertiary lymphoid structures (TLSs), which are commonly associated with a good prognosis. Thus, therapeutic induction of TU-HEVs may provide attractive avenues to induce and sustain the efficacy of immunotherapies by overcoming the major restriction of T cell exclusion from the tumor microenvironment. In this review, we provide current insight into the commonalities and discrepancies of HEV formation and regulation in LNs and tumors and discuss the specific function and significance of TU-HEVs in eliciting, predicting, and aiding anti-tumoral immunity.
Collapse
Affiliation(s)
- Gerlanda Vella
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Yichao Hua
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
4
|
Milutinovic S, Abe J, Jones E, Kelch I, Smart K, Lauder SN, Somerville M, Ware C, Godkin A, Stein JV, Bogle G, Gallimore A. Three-dimensional Imaging Reveals Immune-driven Tumor-associated High Endothelial Venules as a Key Correlate of Tumor Rejection Following Depletion of Regulatory T Cells. CANCER RESEARCH COMMUNICATIONS 2022; 2:1641-1656. [PMID: 36704666 PMCID: PMC7614106 DOI: 10.1158/2767-9764.crc-21-0123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 06/29/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022]
Abstract
High endothelial venules (HEV) are specialized post capillary venules that recruit naïve T cells and B cells into secondary lymphoid organs (SLO) such as lymph nodes (LN). Expansion of HEV networks in SLOs occurs following immune activation to support development of an effective immune response. In this study, we used a carcinogen-induced model of fibrosarcoma to examine HEV remodeling after depletion of regulatory T cells (Treg). We used light sheet fluorescence microscopy imaging to visualize entire HEV networks, subsequently applying computational tools to enable topological mapping and extraction of numerical descriptors of the networks. While these analyses revealed profound cancer- and immune-driven alterations to HEV networks within LNs, these changes did not identify successful responses to treatment. The presence of HEV networks within tumors did however clearly distinguish responders from nonresponders. Finally, we show that a successful treatment response is dependent on coupling tumor-associated HEV (TA-HEV) development to T-cell activation implying that T-cell activation acts as the trigger for development of TA-HEVs which subsequently serve to amplify the immune response by facilitating extravasation of T cells into the tumor mass.
Collapse
Affiliation(s)
- Stefan Milutinovic
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Jun Abe
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Emma Jones
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Inken Kelch
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Kathryn Smart
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Sarah N. Lauder
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michelle Somerville
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Carl Ware
- Laboratory of Molecular Immunology, Sanford Burnham Prebys, La Jolla, California
| | - Andrew Godkin
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Jens V. Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Gib Bogle
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Awen Gallimore
- Systems Immunity University Research Institute, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
5
|
Lim AR, Ghajar CM. Thorny ground, rocky soil: Tissue-specific mechanisms of tumor dormancy and relapse. Semin Cancer Biol 2022; 78:104-123. [PMID: 33979673 PMCID: PMC9595433 DOI: 10.1016/j.semcancer.2021.05.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023]
Abstract
Disseminated tumor cells (DTCs) spread systemically yet distinct patterns of metastasis indicate a range of tissue susceptibility to metastatic colonization. Distinctions between permissive and suppressive tissues are still being elucidated at cellular and molecular levels. Although there is a growing appreciation for the role of the microenvironment in regulating metastatic success, we have a limited understanding of how diverse tissues regulate DTC dormancy, the state of reversible quiescence and subsequent awakening thought to contribute to delayed relapse. Several themes of microenvironmental regulation of dormancy are beginning to emerge, including vascular association, co-option of pre-existing niches, metabolic adaptation, and immune evasion, with tissue-specific nuances. Conversely, DTC awakening is often associated with injury or inflammation-induced activation of the stroma, promoting a proliferative environment with DTCs following suit. We review what is known about tissue-specific regulation of tumor dormancy on a tissue-by-tissue basis, profiling major metastatic organs including the bone, lung, brain, liver, and lymph node. An aerial view of the barriers to metastatic growth may reveal common targets and dependencies to inform the therapeutic prevention of relapse.
Collapse
Affiliation(s)
- Andrea R Lim
- Public Health Sciences Division/Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Graduate Program in Molecular and Cellular Biology, University of Washington/Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| | - Cyrus M Ghajar
- Public Health Sciences Division/Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| |
Collapse
|
6
|
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
|
7
|
Sim JH, Ambler WG, Sollohub IF, Howlader MJ, Li TM, Lee HJ, Lu TT. Immune Cell-Stromal Circuitry in Lupus Photosensitivity. THE JOURNAL OF IMMUNOLOGY 2021; 206:302-309. [PMID: 33397744 DOI: 10.4049/jimmunol.2000905] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022]
Abstract
Photosensitivity is a sensitivity to UV radiation (UVR) commonly found in systemic lupus erythematosus (SLE) patients who have cutaneous disease. Upon even ambient UVR exposure, patients can develop inflammatory skin lesions that can reduce the quality of life. Additionally, UVR-exposed skin lesions can be associated with systemic disease flares marked by rising autoantibody titers and worsening kidney disease. Why SLE patients are photosensitive and how skin sensitivity leads to systemic disease flares are not well understood, and treatment options are limited. In recent years, the importance of immune cell-stromal interactions in tissue function and maintenance is being increasingly recognized. In this review, we discuss SLE as an anatomic circuit and review recent findings in the pathogenesis of photosensitivity with a focus on immune cell-stromal circuitry in tissue health and disease.
Collapse
Affiliation(s)
- Ji Hyun Sim
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021.,Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065
| | - William G Ambler
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021.,Pediatric Rheumatology, Hospital for Special Surgery, New York, NY 10021
| | - Isabel F Sollohub
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021
| | - Mir J Howlader
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021.,Biochemistry and Structural Biology, Cell Biology, Developmental Biology, and Molecular Biology Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065; and
| | - Thomas M Li
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021
| | - Henry J Lee
- Department of Dermatology, Weill Cornell Medical College, New York, NY 10065
| | - Theresa T Lu
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021; .,Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065.,Pediatric Rheumatology, Hospital for Special Surgery, New York, NY 10021
| |
Collapse
|
8
|
Zou M, Wiechers C, Huehn J. Lymph node stromal cell subsets-Emerging specialists for tailored tissue-specific immune responses. Int J Med Microbiol 2021; 311:151492. [PMID: 33676241 DOI: 10.1016/j.ijmm.2021.151492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/04/2021] [Accepted: 02/23/2021] [Indexed: 12/17/2022] Open
Abstract
The effective priming of adaptive immune responses depends on the precise dispatching of lymphocytes and antigens into and within lymph nodes (LNs), which are strategically dispersed throughout the body. Over the past decade, a growing body of evidence has advanced our understanding of lymph node stromal cells (LNSCs) from viewing them as mere accessory cells to seeing them as critical cellular players for the modulation of adaptive immune responses. In this review, we summarize current advances on the pivotal roles that LNSCs play in orchestrating adaptive immune responses during homeostasis and infection, and highlight the imprinting of location-specific information by micro-environmental cues into LNSCs, thereby tailoring tissue-specific immune responses.
Collapse
Affiliation(s)
- Mangge Zou
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124 Braunschweig, Germany
| | - Carolin Wiechers
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124 Braunschweig, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124 Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| |
Collapse
|
9
|
Milutinovic S, Abe J, Godkin A, Stein JV, Gallimore A. The Dual Role of High Endothelial Venules in Cancer Progression versus Immunity. Trends Cancer 2021; 7:214-225. [PMID: 33132107 PMCID: PMC9213382 DOI: 10.1016/j.trecan.2020.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/17/2022]
Abstract
Secondary lymphoid organs (SLOs) are important initiators and regulators of immunity. To carry out this function, the blood vasculature must deliver oxygen and nutrients and recruit circulating lymphocytes into the SLO parenchyma, where they encounter cognate antigen. High endothelial venules (HEVs) are specialised postcapillary venules that specifically serve this function and are found in all SLOs except spleen. It is becoming clear that alterations to HEV network density and/or morphology can result in immune activation or, as recently implicated, in providing an exit route for tumour cell dissemination and metastases. In this review, the structural plasticity of HEVs, the regulatory pathways underpinning this plasticity, and the relevance of these pathways to cancer progression will be discussed.
Collapse
Affiliation(s)
- Stefan Milutinovic
- Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Jun Abe
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Andrew Godkin
- Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Awen Gallimore
- Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK.
| |
Collapse
|
10
|
Cabeza-Cabrerizo M, Cardoso A, Minutti CM, Pereira da Costa M, Reis E Sousa C. Dendritic Cells Revisited. Annu Rev Immunol 2021; 39:131-166. [PMID: 33481643 DOI: 10.1146/annurev-immunol-061020-053707] [Citation(s) in RCA: 435] [Impact Index Per Article: 108.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dendritic cells (DCs) possess the ability to integrate information about their environment and communicate it to other leukocytes, shaping adaptive and innate immunity. Over the years, a variety of cell types have been called DCs on the basis of phenotypic and functional attributes. Here, we refocus attention on conventional DCs (cDCs), a discrete cell lineage by ontogenetic and gene expression criteria that best corresponds to the cells originally described in the 1970s. We summarize current knowledge of mouse and human cDC subsets and describe their hematopoietic development and their phenotypic and functional attributes. We hope that our effort to review the basic features of cDC biology and distinguish cDCs from related cell types brings to the fore the remarkable properties of this cell type while shedding some light on the seemingly inordinate complexity of the DC field.
Collapse
Affiliation(s)
- Mar Cabeza-Cabrerizo
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | - Ana Cardoso
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | - Carlos M Minutti
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | | | - Caetano Reis E Sousa
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| |
Collapse
|
11
|
Pikor NB, Cheng HW, Onder L, Ludewig B. Development and Immunological Function of Lymph Node Stromal Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:257-263. [PMID: 33397739 DOI: 10.4049/jimmunol.2000914] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/16/2020] [Indexed: 01/07/2023]
Abstract
Stromal cells have for a long time been viewed as structural cells that support distinct compartments within lymphoid tissues and little more. Instead, an active cross-talk between endothelial and fibroblastic stromal cells drives the maturation of lymphoid niches, a relationship that is recapitulated during lymph node organogenesis, steady-state conditions, and following inflammation. In this review, we go over recent advances in genetic models and high-resolution transcriptomic analyses that have propelled the finer resolution of the stromal cell infrastructure of lymph nodes, revealing that the distinct subsets are strategically positioned to deliver a catered mixture of niche factors to interacting immune cell populations. Moreover, we discuss how changes in the activation state of poised stromal cell-underpinned niches rather than on-demand differentiation of new stromal cell subsets govern the efficient interaction of Ag, APC, and cognate B and T lymphocytes during adaptive immune responses.
Collapse
Affiliation(s)
- Natalia Barbara Pikor
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, CH-9007 St. Gallen, Switzerland; and
| | - Hung-Wei Cheng
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, CH-9007 St. Gallen, Switzerland; and
| | - Lucas Onder
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, CH-9007 St. Gallen, Switzerland; and
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Center, Kantonsspital St. Gallen, CH-9007 St. Gallen, Switzerland; and .,Institute of Experimental Immunology, University of Zürich, 8006 Zürich, Switzerland
| |
Collapse
|
12
|
Dasoveanu DC, Park HJ, Ly CL, Shipman WD, Chyou S, Kumar V, Tarlinton D, Ludewig B, Mehrara BJ, Lu TT. Lymph node stromal CCL2 limits antibody responses. Sci Immunol 2020; 5:5/45/eaaw0693. [PMID: 32198221 DOI: 10.1126/sciimmunol.aaw0693] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 11/26/2019] [Accepted: 02/11/2020] [Indexed: 12/12/2022]
Abstract
Nonhematopoietic stromal cells in lymph nodes such as fibroblastic reticular cells (FRCs) can support the survival of plasmablasts and plasma cells [together, antibody-forming cells (AFCs)]. However, a regulatory function for the stromal compartment in AFC accumulation has not been appreciated. Here, we show that chemokine ligand 2 (CCL2)-expressing stromal cells limit AFC survival. FRCs express high levels of CCL2 in vessel-rich areas of the T cell zone and the medulla, where AFCs are located. FRC CCL2 is up-regulated during AFC accumulation, and we use lymph node transplantation to show that CCL2 deficiency in BP3+ FRCs and lymphatic endothelial cells increases AFC survival without affecting B or germinal center cell numbers. Monocytes are key expressers of the CCL2 receptor CCR2, as monocyte depletion and transfer late in AFC responses increases and decreases AFC accumulation, respectively. Monocytes express reactive oxygen species (ROS) in an NADPH oxidase 2 (NOX2)-dependent manner, and NOX2-deficient monocytes fail to reduce AFC numbers. Stromal CCL2 modulates both monocyte accumulation and ROS production, and is regulated, in part, by manipulations that modulate vascular permeability. Together, our results reveal that the lymph node stromal compartment, by influencing monocyte accumulation and functional phenotype, has a regulatory role in AFC survival. Our results further suggest a role for inflammation-induced vascular activity in tuning the lymph node microenvironment. The understanding of stromal-mediated AFC regulation in vessel-rich environments could potentially be harnessed to control antibody-mediated autoimmunity.
Collapse
Affiliation(s)
- Dragos C Dasoveanu
- Physiology Biophysics and Systems Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA.,Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, New York, NY 10021, USA
| | - Hyeung Ju Park
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Catherine L Ly
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - William D Shipman
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, New York, NY 10021, USA.,Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA.,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Susan Chyou
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, New York, NY 10021, USA
| | - Varsha Kumar
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, New York, NY 10021, USA
| | - David Tarlinton
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3004, Australia
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen CH-9007, Switzerland.,Institute of Experimental Immunology, University of Zürich, Zürich CH-8057, Switzerland
| | - Babak J Mehrara
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Theresa T Lu
- Autoimmunity and Inflammation Program, Hospital for Special Surgery Research Institute, New York, NY 10021, USA. .,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA.,Pediatric Rheumatology, Hospital for Special Surgery, New York, NY 10021, USA.,Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
13
|
Menzel L, Höpken UE, Rehm A. Angiogenesis in Lymph Nodes Is a Critical Regulator of Immune Response and Lymphoma Growth. Front Immunol 2020; 11:591741. [PMID: 33343570 PMCID: PMC7744479 DOI: 10.3389/fimmu.2020.591741] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor-induced remodeling of the microenvironment in lymph nodes (LNs) includes the formation of blood vessels, which goes beyond the regulation of metabolism, and shaping a survival niche for tumor cells. In contrast to solid tumors, which primarily rely on neo-angiogenesis, hematopoietic malignancies usually grow within pre-vascularized autochthonous niches in secondary lymphatic organs or the bone marrow. The mechanisms of vascular remodeling in expanding LNs during infection-induced responses have been studied in more detail; in contrast, insights into the conditions of lymphoma growth and lodging remain enigmatic. Based on previous murine studies and clinical trials in human, we conclude that there is not a universal LN-specific angiogenic program applicable. Instead, signaling pathways that are tightly connected to autochthonous and infiltrating cell types contribute variably to LN vascular expansion. Inflammation related angiogenesis within LNs relies on dendritic cell derived pro-inflammatory cytokines stimulating vascular endothelial growth factor-A (VEGF-A) expression in fibroblastic reticular cells, which in turn triggers vessel growth. In high-grade B cell lymphoma, angiogenesis correlates with poor prognosis. Lymphoma cells immigrate and grow in LNs and provide pro-angiogenic growth factors themselves. In contrast to infectious stimuli that impact on LN vasculature, they do not trigger the typical inflammatory and hypoxia-related stroma-remodeling cascade. Blood vessels in LNs are unique in selective recruitment of lymphocytes via high endothelial venules (HEVs). The dissemination routes of neoplastic lymphocytes are usually disease stage dependent. Early seeding via the blood stream requires the expression of the homeostatic chemokine receptor CCR7 and of L-selectin, both cooperate to facilitate transmigration of tumor and also of protective tumor-reactive lymphocytes via HEV structures. In this view, the HEV route is not only relevant for lymphoma cell homing, but also for a continuous immunosurveillance. We envision that HEV functional and structural alterations during lymphomagenesis are not only key to vascular remodeling, but also impact on tumor cell accessibility when targeted by T cell-mediated immunotherapies.
Collapse
Affiliation(s)
- Lutz Menzel
- Translational Tumor Immunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Uta E. Höpken
- Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Armin Rehm
- Translational Tumor Immunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
14
|
Zhao SJ, Liu H, Chen J, Qian DF, Kong FQ, Jie J, Yin GY, Li QQ, Fan J. Macrophage GIT1 Contributes to Bone Regeneration by Regulating Inflammatory Responses in an ERK/NRF2-Dependent Way. J Bone Miner Res 2020; 35:2015-2031. [PMID: 32460388 PMCID: PMC7689802 DOI: 10.1002/jbmr.4099] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022]
Abstract
Despite the best treatment, approximately 10% of fractures still face undesirable repair. Recently, many studies have focused on the importance of macrophages in bone repair; however, the cellular mechanisms by which they work are not yet fully understood. In this study, we explored the functions of macrophage G-protein-coupled receptor interacting protein 1 (GIT1) in healing a tibial monocortical defect model. Using GIT1flox/flox Lyz2-Cre (GIT1 CKO) mice, we observed that a GIT1 deficiency in the macrophages led to an exacerbation of interleukin 1β (IL1β) production, more M1-like macrophage infiltration, and impaired intramembranous ossification in vivo. The results of in vitro assays further indicated that the macrophage GIT1 plays a critical role in several cellular processes in response to lipopolysaccharide (LPS), such as anti-oxidation, IL1β production alleviation, and glycolysis control. Although GIT1 has been recognized as a scaffold protein, our data clarified that GIT1-mediated extracellular-signal-regulated kinase (ERK) phosphorylation could activate nuclear factor (erythroid-derived 2)-like 2 (NRF2) in macrophages after LPS treatment. Moreover, we demonstrated that macrophage GIT1-activated ERK/NRF2 negatively regulates the 6-phosphofructo-2-kinase/fructose-2, 6-biphosphatase 3 (PFKFB3), facilitating the decrease of glycolysis. Our findings uncovered a previously unrecognized role of GIT1 in regulating ERK/NRF2 in macrophages to control the inflammatory response, suggesting that macrophage GIT1 could be a potential target to improve bone regeneration. © 2020 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research..
Collapse
Affiliation(s)
- Shu-Jie Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ding-Fei Qian
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fan-Qi Kong
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Jie
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Orthopedics, Pukou Branch of Jiangsu Province Hospital (Nanjing Pukou Central Hospital), Nanjing, China
| | - Guo-Yong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qing-Qing Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Shipman WD, Sandoval MJ, Veiga K, Donlin LT, Lu TT. Fibroblast subtypes in tissues affected by autoimmunity: with lessons from lymph node fibroblasts. Curr Opin Immunol 2020; 64:63-70. [PMID: 32387902 DOI: 10.1016/j.coi.2020.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/04/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
The recent advent of single-cell technologies has fast-tracked the discovery of multiple fibroblast subsets in tissues affected by autoimmune disease. In recent years, interest in lymph node fibroblasts that support and regulate immune cells has also grown, leading to an expanding framework of stromal cell subsets with distinct spatial, transcriptional, and functional characteristics. Inflammation can drive tissue fibroblasts to adopt a lymphoid tissue stromal cell phenotype, suggesting that fibroblasts in diseased tissues can have counterparts in lymphoid tissues. Here, we examine fibroblast subsets in tissues affected by autoimmunity in the context of knowledge gained from studies on lymph node fibroblasts, with the ultimate aim to better understand stromal cell heterogeneity in these immunologically reactive tissues.
Collapse
Affiliation(s)
- William D Shipman
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA; Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Marvin J Sandoval
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Keila Veiga
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA; Pediatric Rheumatology, Hospital for Special Surgery, New York, NY 10021, USA
| | - Laura T Donlin
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA; Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Theresa T Lu
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA; Pediatric Rheumatology, Hospital for Special Surgery, New York, NY 10021, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
16
|
Lymph node stromal cells: cartographers of the immune system. Nat Immunol 2020; 21:369-380. [PMID: 32205888 DOI: 10.1038/s41590-020-0635-3] [Citation(s) in RCA: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/17/2020] [Indexed: 01/03/2023]
Abstract
Lymph nodes (LNs) are strategically positioned at dedicated sites throughout the body to facilitate rapid and efficient immunity. Central to the structural integrity and framework of LNs, and the recruitment and positioning of leukocytes therein, are mesenchymal and endothelial lymph node stromal cells (LNSCs). Advances in the last decade have expanded our understanding and appreciation of LNSC heterogeneity, and the role they play in coordinating immunity has grown rapidly. In this review, we will highlight the functional contributions of distinct stromal cell populations during LN development in maintaining immune homeostasis and tolerance and in the activation and control of immune responses.
Collapse
|
17
|
Noah AC, Li TM, Martinez LM, Wada S, Swanson JB, Disser NP, Sugg KB, Rodeo SA, Lu TT, Mendias CL. Adaptive and innate immune cell responses in tendons and lymph nodes after tendon injury and repair. J Appl Physiol (1985) 2020; 128:473-482. [PMID: 31944888 DOI: 10.1152/japplphysiol.00682.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tendon injuries are a common clinical condition with limited treatment options. The cellular components of the innate immune system, such as neutrophils and macrophages, have been studied in tendon injuries. However, the adaptive immune system, comprising specialized lymphocytes, plays an important role in orchestrating the healing of numerous tissues, but less is known about these cells in tendon healing. To gain a greater understanding of the biological processes that regulate tendon healing, we determined how the cellular components of the adaptive and innate immune system respond to a tendon injury using two-month-old male mice. We observed that lymphatic vasculature is present in the epitenon and superficial regions of Achilles tendons, and that the lymphatics drain into the popliteal lymph node. We then created an acute Achilles tenotomy followed by repair, and collected tendons and popliteal lymph nodes 1, 2, and 4 wk after injury. Tendon injury resulted in a robust adaptive immune cell response that followed an initial innate immune cell response in tendons and lymph nodes. Monocytes, neutrophils, and macrophages initially accumulated at 1 wk after injury in tendons, while dendritic cells and CD4+ T cells peaked at 2 wk after injury. B cells and CD8+ T cells progressively increased over time. In parallel, immune cells of the popliteal lymph node demonstrated a similarly coordinated response to the injury. These results suggest that there is an adaptive immune response to tendon injury, and adaptive immune cells may play a role in regulating tendon healing.NEW & NOTEWORTHY While the innate immune system, consisting of macrophages and related hematopoietic cells, has been studied in tendon injury, less is known about the adaptive immune system. Using a mouse model of Achilles tendon tenotomy and repair, we observed an adaptive immune cell response, consisting of CD4+ and CD8+ T cells, and B cells, which occur through 4 wk after tendon injury. This response appeared to be coordinated by the draining popliteal lymph node.
Collapse
Affiliation(s)
| | - Thomas M Li
- Hospital for Special Surgery, New York, New York
| | | | - Susumu Wada
- Hospital for Special Surgery, New York, New York
| | | | | | - Kristoffer B Sugg
- Section of Plastic Surgery, Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Scott A Rodeo
- Hospital for Special Surgery, New York, New York.,Department of Orthopaedic Surgery, Weill Cornell Medical College, New York, New York
| | - Theresa T Lu
- Hospital for Special Surgery, New York, New York.,Department of Microbiology & Immunology, Weill Cornell Medical College, New York, New York
| | - Christopher L Mendias
- Hospital for Special Surgery, New York, New York.,Department of Orthopaedic Surgery, Weill Cornell Medical College, New York, New York.,Department of Physiology & Biophysics, Weill Cornell Medical College, New York, New York
| |
Collapse
|
18
|
Thierry GR, Gentek R, Bajenoff M. Remodeling of reactive lymph nodes: Dynamics of stromal cells and underlying chemokine signaling. Immunol Rev 2020; 289:42-61. [PMID: 30977194 DOI: 10.1111/imr.12750] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 01/28/2019] [Accepted: 01/31/2019] [Indexed: 12/19/2022]
Abstract
Lymph nodes (LNs) are secondary immune organs dispersed throughout the body. They are primarily composed of lymphocytes, "transient passengers" that are only present for a few hours. During this time, they extensively interact with a meshwork of stromal cells. Although these cells constitute less than 5% of all LN cells, they are integral to LN function: Stromal cells create a three-dimensional network that provides a rigid backbone for the transport of lymph and generates "roads" for lymphocyte migration. Beyond structural support, the LN stroma also produces survival signals for lymphocytes and provides nutrients, soluble factors, antigens, and immune cells collectively required for immune surveillance and the generation of adaptive immune responses. A unique feature of LNs is their ability to considerably and rapidly change size: the volume and cellularity of inflamed LNs can increase up to 20-fold before returning to homeostatic levels. This cycle will be repeated many times during life and is accommodated by stromal cells. The dynamics underlying this dramatic remodeling are subject of this review. We will first introduce the main types of LN stromal cells and explain their known functions. We will then discuss how these cells enable LN growth during immune responses, with a particular focus on underlying cellular mechanisms and molecular cues. Similarly, we will elaborate on stromal dynamics mediating the return to LN homeostasis, a process that is mechanistically much less understood than LN expansion.
Collapse
Affiliation(s)
- Guilhem R Thierry
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, Marseille, France
| | - Rebecca Gentek
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, Marseille, France
| | - Marc Bajenoff
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, Marseille, France
| |
Collapse
|
19
|
Gloger M, Menzel L, Grau M, Vion AC, Anagnostopoulos I, Zapukhlyak M, Gerlach K, Kammertöns T, Hehlgans T, Zschummel M, Lenz G, Gerhardt H, Höpken UE, Rehm A. Lymphoma Angiogenesis Is Orchestrated by Noncanonical Signaling Pathways. Cancer Res 2020; 80:1316-1329. [PMID: 31932457 DOI: 10.1158/0008-5472.can-19-1493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/04/2019] [Accepted: 01/08/2020] [Indexed: 11/16/2022]
Abstract
Tumor-induced remodeling of the microenvironment relies on the formation of blood vessels, which go beyond the regulation of metabolism, shaping a maladapted survival niche for tumor cells. In high-grade B-cell lymphoma, angiogenesis correlates with poor prognosis, but attempts to target established proangiogenic pathways within the vascular niche have been inefficient. Here, we analyzed Myc-driven B-cell lymphoma-induced angiogenesis in mice. A few lymphoma cells were sufficient to activate the angiogenic switch in lymph nodes. A unique morphology of dense microvessels emerged without obvious tip cell guidance and reliance on blood endothelial cell (BEC) proliferation. The transcriptional response of BECs was inflammation independent. Conventional HIF1α or Notch signaling routes prevalent in solid tumors were not activated. Instead, a nonconventional hypersprouting morphology was orchestrated by lymphoma-provided VEGFC and lymphotoxin (LT). Interference with VEGF receptor-3 and LTβ receptor signaling pathways abrogated lymphoma angiogenesis, thus revealing targets to block lymphomagenesis. SIGNIFICANCE: In lymphoma, transcriptomes and morphogenic patterns of the vasculature are distinct from processes in inflammation and solid tumors. Instead, LTβR and VEGFR3 signaling gain leading roles and are targets for lymphomagenesis blockade.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/80/6/1316/F1.large.jpg.
Collapse
Affiliation(s)
- Marleen Gloger
- Translational Tumorimmunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Lutz Menzel
- Translational Tumorimmunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Michael Grau
- Department of Medicine A, and Cluster of Excellence EXC 1003, University Hospital Münster, Münster, Germany
| | - Anne-Clemence Vion
- Integrative Vascular Biology Lab, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | - Myroslav Zapukhlyak
- Department of Medicine A, and Cluster of Excellence EXC 1003, University Hospital Münster, Münster, Germany
| | - Kerstin Gerlach
- Translational Tumorimmunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Thomas Kammertöns
- Institute of Immunology, Charité -University Medicine Berlin, Berlin, Germany
| | - Thomas Hehlgans
- Regensburg Center for Interventional Immunology, University Hospital Regensburg, Regensburg, Germany
| | - Maria Zschummel
- Microenvironmental Regulation in Autoimmunity and Cancer, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Georg Lenz
- Department of Medicine A, and Cluster of Excellence EXC 1003, University Hospital Münster, Münster, Germany
| | - Holger Gerhardt
- Integrative Vascular Biology Lab, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Uta E Höpken
- Microenvironmental Regulation in Autoimmunity and Cancer, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
| | - Armin Rehm
- Translational Tumorimmunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|
20
|
Abstract
BACKGROUND Periprosthetic joint infection (PJI) is one of the most devastating complications of total joint arthroplasty. Given the mortality and morbidity associated with PJI and the challenges in treating it, there has been increased interest in risk factors that can be modified before surgery. In this study, we used a novel mouse model to consider the role of the gut microbiome as a risk factor for PJI. QUESTIONS/PURPOSES (1) Does the state of the gut microbiota before surgery influence the likelihood of developing an established infection in a mouse model of PJI? (2) How does the state of the gut microbiota before surgery influence the local and systemic response to the presence of an established infection in a mouse model of PJI? METHODS Male C57Bl/6 mice were divided into two groups: those with modified microbiome [INCREMENT]microbiome (n = 40) and untreated mice (n = 42). In [INCREMENT]microbiome mice, the gut flora were modified using oral neomycin and ampicillin from 4 weeks to 16 weeks of age. Mice received a titanium tibial implant to mimic a joint implant and a local inoculation of Staphylococcus aureus in the synovial space (10 colony forming units [CFUs]). The proportion of animals developing an established infection in each group was determined by CFU count. The local and systemic response to established infection was determined using CFU counts in surrounding joint tissues, analysis of gait, radiographs, body weight, serum markers of inflammation, and immune cell profiles and was compared with animals that received the inoculation but resisted infection. RESULTS A greater proportion of animals with disrupted gut microbiota had infection (29 of 40 [73%]) than did untreated animals (21 of 42 [50%]; odds ratio, 2.63, 95% CI, 1.04-6.61; p = 0.035). The immune response to established infection in mice with altered microbiota was muted; serum amyloid A, a marker of systemic infection in mice, was greater than in mice with disrupted gut microbiota with infection (689 µg/dL; range, 68-2437 µg/dL, p < 0.05); infection associated increases in monocytes and neutrophils in the spleen and local lymph node in untreated mice but not were not observed in mice with disrupted gut microbiota. CONCLUSIONS The findings from this in vivo mouse model suggest that the gut microbiota may influence susceptibility to PJI. CLINICAL RELEVANCE These preclinical findings support the idea that the state of the gut microbiome before surgery may influence the development of PJI and justify further preclinical and clinical studies to develop appropriate microbiome-based interventions.
Collapse
|
21
|
Barron AMS, Mantero JC, Ho JD, Nazari B, Horback KL, Bhawan J, Lafyatis R, Lam C, Browning JL. Perivascular Adventitial Fibroblast Specialization Accompanies T Cell Retention in the Inflamed Human Dermis. THE JOURNAL OF IMMUNOLOGY 2018; 202:56-68. [PMID: 30510068 DOI: 10.4049/jimmunol.1801209] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Perivascular accumulation of lymphocytes can be a prominent histopathologic feature of various human inflammatory skin diseases. Select examples include systemic sclerosis, spongiotic dermatitis, and cutaneous lupus. Although a large body of work has described various aspects of the endothelial and vascular smooth muscle layers in these diseases, the outer adventitial compartment is poorly explored. The goal of the current study was to characterize perivascular adventitial fibroblast states in inflammatory human skin diseases and relate these states to perivascular lymphocyte accumulation. In normal skin, adventitial fibroblasts are distinguished by CD90 expression, and dense perivascular lymphocytic infiltrates are uncommon. In systemic sclerosis, this compartment expands, but lymphocyte infiltrates remain sparse. In contrast, perivascular adventitial fibroblast expression of VCAM1 is upregulated in spongiotic dermatitis and lupus and is associated with a dense perivascular T cell infiltrate. VCAM1 expression marks transitioned fibroblasts that show some resemblance to the reticular stromal cells in secondary lymphoid organs. Expanded adventitial compartments with perivascular infiltrates similar to the human settings were not seen in the inflamed murine dermis. This species difference may hinder the dissection of aspects of perivascular adventitial pathology. The altered perivascular adventitial compartment and its associated reticular network form a niche for lymphocytes and appear to be fundamental in the development of an inflammatory pattern.
Collapse
Affiliation(s)
- Alexander M S Barron
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| | - Julio C Mantero
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| | - Jonathan D Ho
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Banafsheh Nazari
- Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118
| | - Katharine L Horback
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115; and
| | - Jag Bhawan
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Robert Lafyatis
- Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118.,Division of Rheumatology and Clinical Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213
| | - Christina Lam
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118
| | - Jeffrey L Browning
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118; .,Section of Rheumatology, Boston University School of Medicine, Boston, MA 02118
| |
Collapse
|
22
|
Song A, Zhu L, Gorantla G, Berdysz O, Amici SA, Guerau-de-Arellano M, Madalena KM, Lerch JK, Liu X, Quan N. Salient type 1 interleukin 1 receptor expression in peripheral non-immune cells. Sci Rep 2018; 8:723. [PMID: 29335509 PMCID: PMC5768710 DOI: 10.1038/s41598-018-19248-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/21/2017] [Indexed: 12/26/2022] Open
Abstract
Interleukin 1 is a pleiotropic cytokine that mediates diverse functions through its receptor, type I interleukin 1 receptor (IL-1R1). Most previous studies have focused on the expression and function of IL-1R1 in immune cells. Here we performed a comprehensive mapping of IL-1R1 distribution in multiple peripheral tissues using our IL-1R1 reporter (IL-1R1GR/GR) mice. This method yielded the highest sensitivity of in situ detection of IL-1R1 mRNA and protein. Besides validating previously reported IL-1R1 expression in the endocrine tissues including pituitary and pancreas, our results refuted previously reported exclusive IL-1R1 expression in neurons of the spinal cord dorsal horn and dorsal root ganglia (DRG). Instead, IL-1R1 expression was detected in endothelial cells within DRG, spinal cord, pancreas, colon, muscles and many immune organs. In addition, gp38+ fibroblastic reticular cells (FRCs), rather than tissue macrophages or other immune cells, were found to express high levels of IL-1R1 in colon and many immune organs. A functional test of spleen FRCs showed that they responded rapidly to systemic IL-1β stimulation in vivo. Taken together, this study provides a rigorous re-examination of IL-1R1 expression in peripheral tissues and reveals tissue FRCs as a previously unappreciated novel high IL-1R1-expressing cell type in peripheral IL-1 signaling.
Collapse
Affiliation(s)
- Anping Song
- Department of Oncolgy, Tongji Hospital, Huazhong University of Science and Technology Tongji Medical College, Wuhan, Hubei, 430030, P. R. China
| | - Ling Zhu
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.,West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Gowthami Gorantla
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Olimpia Berdysz
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Stephanie A Amici
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.,School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Mireia Guerau-de-Arellano
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.,School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA.,Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA.,Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
| | - Kathryn M Madalena
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA.,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, 43210, USA
| | - Jessica K Lerch
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA.,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, 43210, USA
| | - Xiaoyu Liu
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA. .,Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, 43210, USA. .,Institute for Behavioral Medicine Research, 460 Medical Center Drive, Columbus, OH, 43210, USA.
| | - Ning Quan
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA. .,Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, 43210, USA. .,Institute for Behavioral Medicine Research, 460 Medical Center Drive, Columbus, OH, 43210, USA.
| |
Collapse
|
23
|
Leukocyte-Stromal Interactions Within Lymph Nodes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:1-22. [PMID: 30155619 DOI: 10.1007/978-3-319-78127-3_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lymph nodes play a crucial role in the formation and initiation of immune responses, allowing lymphocytes to efficiently scan for foreign antigens and serving as rendezvous points for leukocyte-antigen interactions. Here we describe the major stromal subsets found in lymph nodes, including fibroblastic reticular cells, lymphatic endothelial cells, blood endothelial cells, marginal reticular cells, follicular dendritic cells and other poorly defined subsets such as integrin alpha-7+ pericytes. We focus on biomedically relevant interactions with T cells, B cells and dendritic cells, describing pro-survival mechanisms of support for these cells, promotion of their migration and tolerance-inducing mechanisms that help keep the body free of autoimmune-mediated damage.
Collapse
|
24
|
Williams SP, Odell AF, Karnezis T, Farnsworth RH, Gould CM, Li J, Paquet-Fifield S, Harris NC, Walter A, Gregory JL, Lamont SF, Liu R, Takano EA, Nowell CJ, Bower NI, Resnick D, Smyth GK, Coultas L, Hogan BM, Fox SB, Mueller SN, Simpson KJ, Achen MG, Stacker SA. Genome-wide functional analysis reveals central signaling regulators of lymphatic endothelial cell migration and remodeling. Sci Signal 2017; 10:10/499/eaal2987. [DOI: 10.1126/scisignal.aal2987] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
25
|
Gentek R, Bajénoff M. Lymph Node Stroma Dynamics and Approaches for Their Visualization. Trends Immunol 2017; 38:236-247. [PMID: 28214099 DOI: 10.1016/j.it.2017.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 12/21/2022]
Abstract
Lymphoid stromal cells are best known as the architectural cells of lymphoid organs. For decades, they have been considered as inert elements of the immune system but this view has changed dramatically in recent years, when it was discovered that they are endowed with critical immunoregulatory functions. It is now accepted that without them, the adaptive immune response would be compromised, if not abrogated entirely. Here, we review the function of the major lymphoid stromal cell types; the way they remodel upon inflammation; discuss the available tools to track their behavior; and introduce several methodological approaches that we believe will help improving our knowledge of these pivotal cell types.
Collapse
Affiliation(s)
- Rebecca Gentek
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Marc Bajénoff
- Aix Marseille Université, CNRS, INSERM, CIML, Marseille, France.
| |
Collapse
|
26
|
Sautès-Fridman C, Lawand M, Giraldo NA, Kaplon H, Germain C, Fridman WH, Dieu-Nosjean MC. Tertiary Lymphoid Structures in Cancers: Prognostic Value, Regulation, and Manipulation for Therapeutic Intervention. Front Immunol 2016; 7:407. [PMID: 27752258 PMCID: PMC5046074 DOI: 10.3389/fimmu.2016.00407] [Citation(s) in RCA: 255] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/22/2016] [Indexed: 01/03/2023] Open
Abstract
Tertiary lymphoid structures (TLS) are ectopic lymphoid aggregates that reflect lymphoid neogenesis occurring in tissues at sites of inflammation. They are detected in tumors where they orchestrate local and systemic anti-tumor responses. A correlation has been found between high densities of TLS and prolonged patient's survival in more than 10 different types of cancer. TLS can be regulated by the same set of chemokines and cytokines that orchestrate lymphoid organogenesis and by regulatory T cells. Thus, TLS offer a series of putative new targets that could be used to develop therapies aiming to increase the anti-tumor immune response.
Collapse
Affiliation(s)
- Catherine Sautès-Fridman
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| | - Myriam Lawand
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| | - Nicolas A Giraldo
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| | - Hélène Kaplon
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| | - Claire Germain
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| | - Wolf Herman Fridman
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| | - Marie-Caroline Dieu-Nosjean
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| |
Collapse
|
27
|
Mondor I, Jorquera A, Sene C, Adriouch S, Adams R, Zhou B, Wienert S, Klauschen F, Bajénoff M. Clonal Proliferation and Stochastic Pruning Orchestrate Lymph Node Vasculature Remodeling. Immunity 2016; 45:877-888. [DOI: 10.1016/j.immuni.2016.09.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/08/2016] [Accepted: 08/21/2016] [Indexed: 01/06/2023]
|
28
|
Dasoveanu DC, Shipman WD, Chia JJ, Chyou S, Lu TT. Regulation of Lymph Node Vascular-Stromal Compartment by Dendritic Cells. Trends Immunol 2016; 37:764-777. [PMID: 27638128 DOI: 10.1016/j.it.2016.08.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/15/2016] [Accepted: 08/22/2016] [Indexed: 12/24/2022]
Abstract
During normal and pathologic immune responses, lymph nodes can swell considerably. The lymph node vascular-stromal compartment supports and regulates the developing immune responses and undergoes dynamic expansion and remodeling. Recent studies have shown that dendritic cells (DCs), best known for their antigen presentation roles, can directly regulate the vascular-stromal compartment, pointing to a new perspective on DCs as facilitators of lymphoid tissue function. Here, we review the phases of lymph node vascular-stromal growth and remodeling during immune responses, discuss the roles of DCs, and discuss how this understanding can potentially be used for developing novel therapeutic approaches.
Collapse
Affiliation(s)
- Dragos C Dasoveanu
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA; Physiology, Biophysics and Systems Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - William D Shipman
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA; Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA
| | - Jennifer J Chia
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA; Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, NY 10065, USA
| | - Susan Chyou
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Theresa T Lu
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA; Pediatric Rheumatology, Hospital for Special Surgery, New York, NY 10021, USA; Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
29
|
Jordan-Williams KL, Ramanujam N, Farr AG, Ruddell A. The Lymphatic Endothelial mCLCA1 Antibody Induces Proliferation and Growth of Lymph Node Lymphatic Sinuses. PLoS One 2016; 11:e0156079. [PMID: 27224029 PMCID: PMC4880189 DOI: 10.1371/journal.pone.0156079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/09/2016] [Indexed: 12/17/2022] Open
Abstract
Lymphocyte- and leukocyte-mediated lymph node (LN) lymphatic sinus growth (lymphangiogenesis) is involved in immune responses and in diseases including cancer and arthritis. We previously discovered a 10.1.1 Ab that recognizes the lymphatic endothelial cell (LEC) surface protein mCLCA1, which is an interacting partner for LFA1 and Mac-1 that mediates lymphocyte adhesion to LECs. Here, we show that 10.1.1 Ab treatment specifically induces LEC proliferation, and influences migration and adhesion in vitro. Functional testing by injection of mice with 10.1.1 Ab but not control hamster Abs identified rapid induction of LN LEC proliferation and extensive lymphangiogenesis within 23 h. BrdU pulse-chase analysis demonstrated incorporation of proliferating LYVE-1-positive LEC into the growing medullary lymphatic sinuses. The 10.1.1 Ab-induced LN remodeling involved coordinate increases in LECs and also blood endothelial cells, fibroblastic reticular cells, and double negative stroma, as is observed during the LN response to inflammation. 10.1.1 Ab-induced lymphangiogenesis was restricted to LNs, as mCLCA1-expressing lymphatic vessels of the jejunum and dermis were unaffected by 23 h 10.1.1 Ab treatment. These findings demonstrate that 10.1.1 Ab rapidly and specifically induces proliferation and growth of LN lymphatic sinuses and stroma, suggesting a key role of mCLCA1 in coordinating LN remodeling during immune responses.
Collapse
Affiliation(s)
| | - Neela Ramanujam
- Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Andrew G Farr
- Department of Biological Structure, University of Washington, Seattle, WA, United States of America
| | - Alanna Ruddell
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States of America.,Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| |
Collapse
|
30
|
Kumar V, Dasoveanu DC, Chyou S, Tzeng TC, Rozo C, Liang Y, Stohl W, Fu YX, Ruddle NH, Lu TT. A dendritic-cell-stromal axis maintains immune responses in lymph nodes. Immunity 2015; 42:719-30. [PMID: 25902483 DOI: 10.1016/j.immuni.2015.03.015] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 01/17/2015] [Accepted: 02/28/2015] [Indexed: 12/23/2022]
Abstract
Within secondary lymphoid tissues, stromal reticular cells support lymphocyte function, and targeting reticular cells is a potential strategy for controlling pathogenic lymphocytes in disease. However, the mechanisms that regulate reticular cell function are not well understood. Here we found that during an immune response in lymph nodes, dendritic cells (DCs) maintain reticular cell survival in multiple compartments. DC-derived lymphotoxin beta receptor (LTβR) ligands were critical mediators, and LTβR signaling on reticular cells mediated cell survival by modulating podoplanin (PDPN). PDPN modulated integrin-mediated cell adhesion, which maintained cell survival. This DC-stromal axis maintained lymphocyte survival and the ongoing immune response. Our findings provide insight into the functions of DCs, LTβR, and PDPN and delineate a DC-stromal axis that can potentially be targeted in autoimmune or lymphoproliferative diseases.
Collapse
Affiliation(s)
- Varsha Kumar
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Dragos C Dasoveanu
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Susan Chyou
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Te-Chen Tzeng
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Cristina Rozo
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Yong Liang
- Department of Pathology, Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - William Stohl
- Department of Rheumatology, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Yang-Xin Fu
- Department of Pathology, Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Nancy H Ruddle
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06520, USA
| | - Theresa T Lu
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021, USA; Pediatric Rheumatology, Hospital for Special Surgery, New York, NY 10021, USA; Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, 10021, USA.
| |
Collapse
|
31
|
Germain C, Gnjatic S, Dieu-Nosjean MC. Tertiary Lymphoid Structure-Associated B Cells are Key Players in Anti-Tumor Immunity. Front Immunol 2015; 6:67. [PMID: 25755654 PMCID: PMC4337382 DOI: 10.3389/fimmu.2015.00067] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/02/2015] [Indexed: 12/25/2022] Open
Abstract
It is now admitted that the immune system plays a major role in tumor control. Besides the existence of tumor-specific T cells and B cells, many studies have demonstrated that high numbers of tumor-infiltrating lymphocytes are associated with good clinical outcome. In addition, not only the density but also the organization of tumor-infiltrating immune cells has been shown to determine patient survival. Indeed, more and more studies describe the development within the tumor microenvironment of tertiary lymphoid structures (TLS), whose presence has a positive impact on tumor prognosis. TLS are transient ectopic lymphoid aggregates displaying the same organization and functionality as canonical secondary lymphoid organs, with T-cell-rich and B-cell-rich areas that are sites for the differentiation of effector and memory T cells and B cells. However, factors favoring the emergence of such structures within tumors still need to be fully characterized. In this review, we survey the state of the art of what is known about the general organization, induction, and functionality of TLS during chronic inflammation, and more especially in cancer, with a particular focus on the B-cell compartment. We detail the role played by TLS B cells in anti-tumor immunity, both as antigen-presenting cells and tumor antigen-specific antibody-secreting cells, and raise the question of the capacity of chemotherapeutic and immunotherapeutic agents to induce the development of TLS within tumors. Finally, we explore how to take advantage of our knowledge on TLS B cells to develop new therapeutic tools.
Collapse
Affiliation(s)
- Claire Germain
- Laboratory Cancer, Immune Control and Escape, Cordeliers Research Center, INSERM UMRS1138 , Paris , France ; UMRS1138, University Pierre and Marie Curie , Paris , France ; UMRS1138, University Paris Descartes , Paris , France
| | - Sacha Gnjatic
- Division of Hematology, Oncology and Immunology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai , New York, NY , USA
| | - Marie-Caroline Dieu-Nosjean
- Laboratory Cancer, Immune Control and Escape, Cordeliers Research Center, INSERM UMRS1138 , Paris , France ; UMRS1138, University Pierre and Marie Curie , Paris , France ; UMRS1138, University Paris Descartes , Paris , France
| |
Collapse
|