1
|
Najibi AJ, Lane RS, Sobral MC, Bovone G, Kang S, Freedman BR, Gutierrez Estupinan J, Elosegui-Artola A, Tringides CM, Dellacherie MO, Williams K, Ijaz H, Müller S, Turley SJ, Mooney DJ. Durable lymph-node expansion is associated with the efficacy of therapeutic vaccination. Nat Biomed Eng 2024; 8:1226-1242. [PMID: 38710838 PMCID: PMC11485260 DOI: 10.1038/s41551-024-01209-3] [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: 01/27/2023] [Accepted: 03/30/2024] [Indexed: 05/08/2024]
Abstract
Following immunization, lymph nodes dynamically expand and contract. The mechanical and cellular changes enabling the early-stage expansion of lymph nodes have been characterized, yet the durability of such responses and their implications for adaptive immunity and vaccine efficacy are unknown. Here, by leveraging high-frequency ultrasound imaging of the lymph nodes of mice, we report more potent and persistent lymph-node expansion for animals immunized with a mesoporous silica vaccine incorporating a model antigen than for animals given bolus immunization or standard vaccine formulations such as alum, and that durable and robust lymph-node expansion was associated with vaccine efficacy and adaptive immunity for 100 days post-vaccination in a mouse model of melanoma. Immunization altered the mechanical and extracellular-matrix properties of the lymph nodes, drove antigen-dependent proliferation of immune and stromal cells, and altered the transcriptional features of dendritic cells and inflammatory monocytes. Strategies that robustly maintain lymph-node expansion may result in enhanced vaccination outcomes.
Collapse
Affiliation(s)
- Alexander J Najibi
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Ryan S Lane
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Miguel C Sobral
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Giovanni Bovone
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Shawn Kang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Benjamin R Freedman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Joel Gutierrez Estupinan
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Alberto Elosegui-Artola
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
- Institute for Bioengineering of Catalonia, Barcelona, Spain
| | - Christina M Tringides
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
- Harvard Program in Biophysics, Harvard University, Cambridge, MA, USA
| | - Maxence O Dellacherie
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Katherine Williams
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Hamza Ijaz
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Sören Müller
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Shannon J Turley
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.
| |
Collapse
|
2
|
Doan TA, Forward TS, Schafer JB, Lucas ED, Fleming I, Uecker-Martin A, Ayala E, Guthmiller JJ, Hesselberth JR, Morrison TE, Tamburini BAJ. Immunization-induced antigen archiving enhances local memory CD8+ T cell responses following an unrelated viral infection. NPJ Vaccines 2024; 9:66. [PMID: 38514656 PMCID: PMC10957963 DOI: 10.1038/s41541-024-00856-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 02/29/2024] [Indexed: 03/23/2024] Open
Abstract
Antigens from viruses or immunizations can persist or are archived in lymph node stromal cells such as lymphatic endothelial cells (LEC) and fibroblastic reticular cells (FRC). Here, we find that, during the time frame of antigen archiving, LEC apoptosis caused by a second, but unrelated, innate immune stimulus such as vaccina viral infection or CpG DNA administration resulted in cross-presentation of archived antigens and boosted memory CD8 + T cells specific to the archived antigen. In contrast to "bystander" activation associated with unrelated infections, the memory CD8 + T cells specific to the archived antigen from the immunization were significantly higher than memory CD8 + T cells of a different antigen specificity. Finally, the boosted memory CD8 + T cells resulted in increased protection against Listeria monocytogenes expressing the antigen from the immunization, but only for the duration that the antigen was archived. These findings outline an important mechanism by which lymph node stromal cell archived antigens, in addition to bystander activation, can augment memory CD8 + T cell responses during repeated inflammatory insults.
Collapse
Affiliation(s)
- Thu A Doan
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
- Immunology Graduate Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Tadg S Forward
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Johnathon B Schafer
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Erin D Lucas
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
- Immunology Graduate Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ira Fleming
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Aspen Uecker-Martin
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Edgardo Ayala
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jenna J Guthmiller
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jay R Hesselberth
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Beth A Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA.
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
3
|
Tamburini B, Doan T, Forward T, Lucas E, Fleming I, Uecker-Martin A, Hesselberth J, Morrison T. Vaccine-induced antigen archiving enhances local memory CD8+ T cell responses following an unrelated viral infection. RESEARCH SQUARE 2023:rs.3.rs-3307809. [PMID: 37841845 PMCID: PMC10571600 DOI: 10.21203/rs.3.rs-3307809/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Viral and vaccine antigens persist or are archived in lymph node stromal cells (LNSC) such as lymphatic endothelial cells (LEC) and fibroblastic reticular cells (FRC). Here, we find that, during the time frame of antigen archiving, LEC apoptosis caused by a second, but unrelated, innate immune stimulus such as vaccina viral infection or CpG DNA administration boosted memory CD8+ T cells specific to the archived antigen. In contrast to "bystander" activation associated with unrelated infections, the memory CD8+ T cells specific to the vaccine archived antigen were significantly higher than memory CD8+ T cells of a different antigen specificity. Finally, the boosted memory CD8+ T cells resulted in increased protection against Listeria monocytogenes expressing the vaccine antigen, but only for the duration that the vaccine antigen was archived. These findings outline a novel mechanism by which LNSC archived antigens, in addition to bystander activation, can augment memory CD8+ T cell responses during repeated inflammatory insults.
Collapse
Affiliation(s)
| | - Thu Doan
- University of Colorado Anschutz Medical Campus
| | | | - Erin Lucas
- University of Colorado Anschutz Medical Campus
| | - Ira Fleming
- University of Colorado Anschutz Medical Campus
| | | | | | | |
Collapse
|
4
|
Daniel L, Counoupas C, Bhattacharyya ND, Triccas JA, Britton WJ, Feng CG. L-selectin-dependent and -independent homing of naïve lymphocytes through the lung draining lymph node support T cell response to pulmonary Mycobacterium tuberculosis infection. PLoS Pathog 2023; 19:e1011460. [PMID: 37405965 DOI: 10.1371/journal.ppat.1011460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 06/05/2023] [Indexed: 07/07/2023] Open
Abstract
Recruiting large numbers of naïve lymphocytes to lymph nodes is critical for mounting an effective adaptive immune response. While most naïve lymphocytes utilize homing molecule L-selectin to enter lymph nodes, some circulating cells can traffic to the lung-draining mediastinal lymph node (mLN) through lymphatics via the intermediate organ, lung. However, whether this alternative trafficking mechanism operates in infection and contributes to T cell priming are unknown. We report that in pulmonary Mycobacterium tuberculosis-infected mice, homing of circulating lymphocytes to the mLN is significantly less efficient than to non-draining lymph node. CD62L blockade only partially reduced the homing of naïve T lymphocytes, consistent with L-selectin-independent routing of naïve lymphocytes to the site. We further demonstrated that lymphatic vessels in infected mLN expanded significantly and inhibiting lymphangiogenesis with a vascular endothelial growth factor receptor 3 kinase inhibitor reduced the recruitment of intravenously injected naïve lymphocytes to the mLN. Finally, mycobacterium-specific T cells entering via the L-selectin-independent route were readily activated in the mLN. Our study suggests that both L-selectin-dependent and -independent pathways contribute to naïve lymphocyte entry into mLN during M. tuberculosis infection and the latter pathway may represent an important mechanism for orchestrating host defence in the lungs.
Collapse
Affiliation(s)
- Lina Daniel
- Immunology and Host Defence Group, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centenary Institute, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Claudio Counoupas
- Centenary Institute, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Microbial Pathogenesis and Immunity Group, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Nayan D Bhattacharyya
- Immunology and Host Defence Group, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centenary Institute, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - James A Triccas
- Centenary Institute, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Microbial Pathogenesis and Immunity Group, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, Australia
| | - Warwick J Britton
- Centenary Institute, The University of Sydney, Sydney, Australia
- The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, Australia
- Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, Sydney, Australia
| | - Carl G Feng
- Immunology and Host Defence Group, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centenary Institute, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, Australia
| |
Collapse
|
5
|
Arroz-Madeira S, Bekkhus T, Ulvmar MH, Petrova TV. Lessons of Vascular Specialization From Secondary Lymphoid Organ Lymphatic Endothelial Cells. Circ Res 2023; 132:1203-1225. [PMID: 37104555 PMCID: PMC10144364 DOI: 10.1161/circresaha.123.322136] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023]
Abstract
Secondary lymphoid organs, such as lymph nodes, harbor highly specialized and compartmentalized niches. These niches are optimized to facilitate the encounter of naive lymphocytes with antigens and antigen-presenting cells, enabling optimal generation of adaptive immune responses. Lymphatic vessels of lymphoid organs are uniquely specialized to perform a staggering variety of tasks. These include antigen presentation, directing the trafficking of immune cells but also modulating immune cell activation and providing factors for their survival. Recent studies have provided insights into the molecular basis of such specialization, opening avenues for better understanding the mechanisms of immune-vascular interactions and their applications. Such knowledge is essential for designing better treatments for human diseases given the central role of the immune system in infection, aging, tissue regeneration and repair. In addition, principles established in studies of lymphoid organ lymphatic vessel functions and organization may be applied to guide our understanding of specialization of vascular beds in other organs.
Collapse
Affiliation(s)
- Silvia Arroz-Madeira
- Department of Oncology, University of Lausanne, Switzerland (S.A.M., T.V.P.)
- Ludwig Institute for Cancer Research Lausanne, Switzerland (S.A.M., T.V.P.)
| | - Tove Bekkhus
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden (T.B., M.H.U.)
| | - Maria H. Ulvmar
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden (T.B., M.H.U.)
| | - Tatiana V. Petrova
- Department of Oncology, University of Lausanne, Switzerland (S.A.M., T.V.P.)
- Ludwig Institute for Cancer Research Lausanne, Switzerland (S.A.M., T.V.P.)
| |
Collapse
|
6
|
Schafer JB, Lucas ED, Dzieciatkowska M, Forward T, Tamburini BAJ. Programmed death ligand 1 intracellular interactions with STAT3 and focal adhesion protein Paxillin facilitate lymphatic endothelial cell remodeling. J Biol Chem 2022; 298:102694. [PMID: 36375639 PMCID: PMC9761386 DOI: 10.1016/j.jbc.2022.102694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/20/2022] [Accepted: 10/22/2022] [Indexed: 11/13/2022] Open
Abstract
Lymphatic endothelial cells (LECs) comprise lymphatic capillaries and vessels that guide immune cells to lymph nodes (LNs) and form the subcapsular sinus and cortical and medullary lymphatic structures of the LN. During an active immune response, the lymphatics remodel to accommodate the influx of immune cells from the tissue, but factors involved in remodeling are unclear. Here, we determined that a TSS motif within the cytoplasmic domain of programmed death ligand 1 (PD-L1), expressed by LECs in the LN, participates in lymphatic remodeling. Mutation of the TSS motif to AAA does not affect surface expression of PD-L1, but instead causes defects in LN cortical and medullary lymphatic organization following immunostimulant, Poly I:C, administration in vivo. Supporting this observation, in vitro treatment of the LEC cell line, SVEC4-10, with cytokines TNFα and IFNα significantly impeded SVEC4-10 movement in the presence of the TSS-AAA cytoplasmic mutation. The cellular movement defects coincided with reduced F-actin polymerization, consistent with differences previously found in dendritic cells. Here, in addition to loss of actin polymerization, we define STAT3 and Paxillin as important PD-L1 binding partners. STAT3 and Paxillin were previously demonstrated to be important at focal adhesions for cellular motility. We further demonstrate the PD-L1 TSS-AAA motif mutation reduced the amount of pSTAT3 and Paxillin bound to PD-L1 both before and after exposure to TNFα and IFNα. Together, these findings highlight PD-L1 as an important component of a membrane complex that is involved in cellular motility, which leads to defects in lymphatic organization.
Collapse
Affiliation(s)
- Johnathon B Schafer
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, Colorado, USA; Molecular Biology Graduate Program, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Erin D Lucas
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, Colorado, USA; Immunology Graduate Program, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Tadg Forward
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Beth A Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, Colorado, USA; Molecular Biology Graduate Program, University of Colorado School of Medicine, Aurora, Colorado, USA; Immunology Graduate Program, University of Colorado School of Medicine, Aurora, Colorado, USA; Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA.
| |
Collapse
|
7
|
Ozawa M, Nakajima S, Kobayashi D, Tomii K, Li NJ, Watarai T, Suzuki R, Watanabe S, Kanda Y, Takeuchi A, Katakai T. Micro- and Macro-Anatomical Frameworks of Lymph Nodes Indispensable for the Lymphatic System Filtering Function. Front Cell Dev Biol 2022; 10:902601. [PMID: 35794860 PMCID: PMC9251010 DOI: 10.3389/fcell.2022.902601] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/19/2022] [Indexed: 01/01/2023] Open
Abstract
In the lymphatic vascular system, lymph nodes (LNs) play a pivotal role in filtering and removing lymph-borne substances. The filtering function of LNs involves resident macrophages tightly associated with unique lymphatic sinus structures. Moreover, an intermittently arranged LN in the lymphatic pathway is considered to cooperatively prevent lymph-borne substances from entering blood circulation. However, the functional significance of tissue microarchitecture, cellular composition, and individual LNs in the “LN chain” system is not fully understood. To explore the mechanistic and histo-anatomical significance of LNs as lymph fluid filters, we subcutaneously injected fluorescent tracers into mice and examined the details of lymphatic transport to the LNs qualitatively and quantitatively. Lymph-borne tracers were selectively accumulated in the MARCO+ subcapsular-medullary sinus border (SMB) region of the LN, in which reticular lymphatic endothelial cells and CD169+F4/80+ medullary sinus macrophages construct a dense meshwork of the physical barrier, forming the main body to capture the tracers. We also demonstrated stepwise filtration via the LN chain in the lymphatic basin, which prevented tracer leakage into the blood. Furthermore, inflammatory responses that induce the remodeling of LN tissue as well as the lymphatic pathway reinforce the overall filtering capacity of the lymphatic basin. Taken together, specialized tissue infrastructure in the LNs and their systematic orchestration constitute an integrated filtering system for lymphatic recirculation.
Collapse
Affiliation(s)
- Madoka Ozawa
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shihori Nakajima
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Daichi Kobayashi
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Koichi Tomii
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Nan-Jun Li
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tomoya Watarai
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ryo Suzuki
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yasuhiro Kanda
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Arata Takeuchi
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Department of Immunology, Tokyo Medical University, Tokyo, Japan
| | - Tomoya Katakai
- Department of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- *Correspondence: Tomoya Katakai,
| |
Collapse
|
8
|
Lobov GI. Role of Endogenous Hydrogen Sulfide in Relaxation of the Lymph Node Capsule in LPS-induced Inflammation. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021060156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
9
|
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
|
10
|
Fibroblasts as immune regulators in infection, inflammation and cancer. Nat Rev Immunol 2021; 21:704-717. [PMID: 33911232 DOI: 10.1038/s41577-021-00540-z] [Citation(s) in RCA: 267] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2021] [Indexed: 02/07/2023]
Abstract
In chronic infection, inflammation and cancer, the tissue microenvironment controls how local immune cells behave, with tissue-resident fibroblasts emerging as a key cell type in regulating activation or suppression of an immune response. Fibroblasts are heterogeneous cells, encompassing functionally distinct populations, the phenotypes of which differ according to their tissue of origin and type of inciting disease. Their immunological properties are also diverse, ranging from the maintenance of a potent inflammatory environment in chronic inflammation to promoting immunosuppression in malignancy, and encapsulating and incarcerating infectious agents within tissues. In this Review, we compare the mechanisms by which fibroblasts control local immune responses, as well as the factors regulating their inflammatory and suppressive profiles, in different tissues and pathological settings. This cross-disease perspective highlights the importance of tissue context in determining fibroblast-immune cell interactions, as well as potential therapeutic avenues to exploit this knowledge for the benefit of patients with chronic infection, inflammation and cancer.
Collapse
|
11
|
Stritt S, Koltowska K, Mäkinen T. Homeostatic maintenance of the lymphatic vasculature. Trends Mol Med 2021; 27:955-970. [PMID: 34332911 DOI: 10.1016/j.molmed.2021.07.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022]
Abstract
The lymphatic vasculature is emerging as a multifaceted regulator of tissue homeostasis and regeneration. Lymphatic vessels drain fluid, macromolecules, and immune cells from peripheral tissues to lymph nodes (LNs) and the systemic circulation. Their recently uncovered functions extend beyond drainage and include direct modulation of adaptive immunity and paracrine regulation of organ growth. The developmental mechanisms controlling lymphatic vessel growth have been described with increasing precision. It is less clear how the essential functional features of lymphatic vessels are established and maintained. We discuss the mechanisms that maintain lymphatic vessel integrity in adult tissues and control vessel repair and regeneration. This knowledge is crucial for understanding the pathological vessel changes that contribute to disease, and provides an opportunity for therapy development.
Collapse
Affiliation(s)
- Simon Stritt
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden
| | - Katarzyna Koltowska
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden
| | - Taija Mäkinen
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden.
| |
Collapse
|
12
|
Li L, Wu J, Abdi R, Jewell CM, Bromberg JS. Lymph node fibroblastic reticular cells steer immune responses. Trends Immunol 2021; 42:723-734. [PMID: 34256989 PMCID: PMC8324561 DOI: 10.1016/j.it.2021.06.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 02/06/2023]
Abstract
Lymph nodes (LNs), where immune responses are initiated, are organized into distinctive compartments by fibroblastic reticular cells (FRCs). FRCs imprint immune responses by supporting LN architecture, recruiting immune cells, coordinating immune cell crosstalk, and presenting antigens. Recent high-resolution transcriptional and histological analyses have enriched our knowledge of LN FRC genetic and spatial heterogeneities. Here, we summarize updated anatomic, phenotypic, and functional identities of FRC subsets, delve into topological and transcriptional remodeling of FRCs in inflammation, and illustrate the crosstalk between FRCs and immune cells. Discussing FRC functions in immunity and tolerance, we highlight state-of-the-art FRC-based therapeutic approaches for maintaining physiological homeostasis, steering protective immunity, inducing transplantation tolerance, and treating diverse immune-related diseases.
Collapse
Affiliation(s)
- Lushen Li
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jing Wu
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Jonathan S Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
13
|
Ocansey DKW, Pei B, Xu X, Zhang L, Olovo CV, Mao F. Cellular and molecular mediators of lymphangiogenesis in inflammatory bowel disease. J Transl Med 2021; 19:254. [PMID: 34112196 PMCID: PMC8190852 DOI: 10.1186/s12967-021-02922-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Background Recent studies reporting the intricate crosstalk between cellular and molecular mediators and the lymphatic endothelium in the development of inflammatory bowel diseases (IBD) suggest altered inflammatory cell drainage and lymphatic vasculature, implicating the lymphatic system as a player in the occurrence, development, and recurrence of intestinal diseases. This article aims to review recent data on the modulatory functions of cellular and molecular components of the IBD microenvironment on the lymphatic system, particularly lymphangiogenesis. It serves as a promising therapeutic target for IBD management and treatment. The interaction with gut microbiota is also explored. Main text Evidence shows that cells of the innate and adaptive immune system and certain non-immune cells participate in the complex processes of inflammatory-induced lymphangiogenesis through the secretion of a wide spectrum of molecular factors, which vary greatly among the various cells. Lymphangiogenesis enhances lymphatic fluid drainage, hence reduced infiltration of immunomodulatory cells and associated-inflammatory cytokines. Interestingly, some of the cellular mediators, including mast cells, neutrophils, basophils, monocytes, and lymphatic endothelial cells (LECs), are a source of lymphangiogenic molecules, and a target as they express specific receptors for lymphangiogenic factors. Conclusion The effective target of lymphangiogenesis is expected to provide novel therapeutic interventions for intestinal inflammatory conditions, including IBD, through both immune and non-immune cells and based on cellular and molecular mechanisms of lymphangiogenesis that facilitate inflammation resolution.
Collapse
Affiliation(s)
- Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.,Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Bing Pei
- Department of Clinical Laboratory, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, 223800, Jiangsu, People's Republic of China
| | - Xinwei Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Lu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Chinasa Valerie Olovo
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.,Department of Microbiology, University of Nigeria, Nsukka, 410001, Nigeria
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| |
Collapse
|
14
|
Poholek AC. Tissue-Specific Contributions to Control of T Cell Immunity. Immunohorizons 2021; 5:410-423. [PMID: 34103371 PMCID: PMC10876086 DOI: 10.4049/immunohorizons.2000103] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/17/2021] [Indexed: 11/19/2022] Open
Abstract
T cells are critical for orchestrating appropriate adaptive immune responses and maintaining homeostasis in the face of persistent nonpathogenic Ags. T cell function is controlled in part by environmental signals received upon activation and derived from the tissue environment in which Ag is encountered. Indeed, tissue-specific environments play important roles in controlling the T cell response to Ag, and recent evidence suggests that tissue draining lymph nodes can mirror those local differences. Thus, tissue-specific immunity may begin at priming in secondary lymph nodes, where local signals have an important role in T cell fate. In this study, we discuss the tissue-specific signals that may impact T cell differentiation and function, including the microbiome, metabolism, and tissue-specific innate cell imprinting. We argue that these individual contributions create tissue-specific niches that likely play important roles in T cell differentiation and function controlling the outcome of the response to Ags.
Collapse
Affiliation(s)
- Amanda C Poholek
- Division of Pediatric Rheumatology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA; and Department of Immunology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
15
|
Cribb MT, Sestito LF, Rockson SG, Nicolls MR, Thomas SN, Dixon JB. The Kinetics of Lymphatic Dysfunction and Leukocyte Expansion in the Draining Lymph Node during LTB 4 Antagonism in a Mouse Model of Lymphedema. Int J Mol Sci 2021; 22:ijms22094455. [PMID: 33923272 PMCID: PMC8123113 DOI: 10.3390/ijms22094455] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/23/2022] Open
Abstract
The mechanisms of lymphedema development are not well understood, but emerging evidence highlights the crucial role the immune system plays in driving its progression. It is well known that lymphatic function deteriorates as lymphedema progresses; however, the connection between this progressive loss of function and the immune-driven changes that characterize the disease has not been well established. In this study, we assess changes in leukocyte populations in lymph nodes within the lymphatic drainage basin of the tissue injury site (draining lymph nodes, dLNs) using a mouse tail model of lymphedema in which a pair of draining collecting vessels are left intact. We additionally quantify lymphatic pump function using established near infrared (NIR) lymphatic imaging methods and lymph-draining nanoparticles (NPs) synthesized and employed by our team for lymphatic tissue drug delivery applications to measure lymphatic transport to and resulting NP accumulation within dLNs associated with swelling following surgery. When applied to assess the effects of the anti-inflammatory drug bestatin, which has been previously shown to be a possible treatment for lymphedema, we find lymph-draining NP accumulation within dLNs and lymphatic function to increase as lymphedema progresses, but no significant effect on leukocyte populations in dLNs or tail swelling. These results suggest that ameliorating this loss of lymphatic function is not sufficient to reverse swelling in this surgically induced disease model that better recapitulates the extent of lymphatic injury seen in human lymphedema. It also suggests that loss of lymphatic function during lymphedema may be driven by immune-mediated mechanisms coordinated in dLNs. Our work indicates that addressing both lymphatic vessel dysfunction and immune cell expansion within dLNs may be required to prevent or reverse lymphedema when partial lymphatic function is sustained.
Collapse
Affiliation(s)
- Matthew T. Cribb
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA; (M.T.C.); (S.N.T.)
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Lauren F. Sestito
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Stanley G. Rockson
- Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA; (S.G.R.); (M.R.N.)
| | - Mark R. Nicolls
- Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA; (S.G.R.); (M.R.N.)
- VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Susan N. Thomas
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA; (M.T.C.); (S.N.T.)
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - J. Brandon Dixon
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA; (M.T.C.); (S.N.T.)
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA;
- Correspondence:
| |
Collapse
|
16
|
Yeo KP, Lim HY, Thiam CH, Azhar SH, Tan C, Tang Y, See WQ, Koh XH, Zhao MH, Phua ML, Balachander A, Tan Y, Lim SY, Chew HS, Ng LG, Angeli V. Efficient aortic lymphatic drainage is necessary for atherosclerosis regression induced by ezetimibe. SCIENCE ADVANCES 2020; 6:6/50/eabc2697. [PMID: 33310846 PMCID: PMC7732200 DOI: 10.1126/sciadv.abc2697] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 10/22/2020] [Indexed: 05/24/2023]
Abstract
A functional lymphatic vasculature is essential for tissue fluid homeostasis, immunity, and lipid clearance. Although atherosclerosis has been linked to adventitial lymphangiogenesis, the functionality of aortic lymphatic vessels draining the diseased aorta has never been assessed and the role of lymphatic drainage in atherogenesis is not well understood. We develop a method to measure aortic lymphatic transport of macromolecules and show that it is impaired during atherosclerosis progression, whereas it is ameliorated during lesion regression induced by ezetimibe. Disruption of aortic lymph flow by lymphatic ligation promotes adventitial inflammation and development of atherosclerotic plaque in hypercholesterolemic mice and inhibits ezetimibe-induced atherosclerosis regression. Thus, progression of atherosclerotic plaques may result not only from increased entry of atherogenic factors into the arterial wall but also from reduced lymphatic clearance of these factors as a result of aortic lymph stasis. Our findings suggest that promoting lymphatic drainage might be effective for treating atherosclerosis.
Collapse
Affiliation(s)
- Kim Pin Yeo
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Hwee Ying Lim
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Chung Hwee Thiam
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Syaza Hazwany Azhar
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Caris Tan
- Histology Core Facility, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Ya Tang
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Wei Qiang See
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Xuan Han Koh
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Ming Hao Zhao
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Meow Ling Phua
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Akhila Balachander
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Yingrou Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Sheau Yng Lim
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Hui Shang Chew
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Lai Guan Ng
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Veronique Angeli
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
17
|
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
|
18
|
Abstract
The influx and efflux of cells and antigens to and from the draining lymph nodes largely take place through the subcapsular, cortical and medullary sinus systems. Recent analyses in mice and humans have revealed unexpected diversity in the lymphatic endothelial cells, which form the distinct regions of the sinuses. As a semipermeable barrier, the lymphatic endothelial cells regulate the sorting of lymph-borne antigens to the lymph node parenchyma and can themselves serve as antigen-presenting cells. The leukocytes entering the lymph node via the sinus system and the lymphocytes egressing from the parenchyma migrate through the lymphatic endothelial cell layer. The sinus lymphatic endothelial cells also orchestrate the organogenesis of lymph nodes, and they undergo bidirectional signalling with other sinus-resident cells, such as subcapsular sinus macrophages, to generate a unique lymphatic niche. In this Review, we consider the structural and functional basis of how the lymph node sinus system coordinates immune responses under physiological conditions, and in inflammation and cancer.
Collapse
|
19
|
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
|
20
|
Jeucken KCM, Koning JJ, Mebius RE, Tas SW. The Role of Endothelial Cells and TNF-Receptor Superfamily Members in Lymphoid Organogenesis and Function During Health and Inflammation. Front Immunol 2019; 10:2700. [PMID: 31824495 PMCID: PMC6879661 DOI: 10.3389/fimmu.2019.02700] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/04/2019] [Indexed: 01/02/2023] Open
Abstract
Lymph nodes (LNs) are crucial for the orchestration of immune responses. LN reactions depend on interactions between incoming and local immune cells, and stromal cells. To mediate these cellular interactions an organized vascular network within the LN exists. In general, the LN vasculature can be divided into two components: blood vessels, which include the specialized high endothelial venules that recruit lymphocytes from the bloodstream, and lymphatic vessels. Signaling via TNF receptor (R) superfamily (SF) members has been implicated as crucial for the development and function of LNs and the LN vasculature. In recent years the role of cell-specific signaling of TNFRSF members in different endothelial cell (EC) subsets and their roles in development and maintenance of lymphoid organs has been elucidated. Here, we discuss recent insights into EC-specific TNFRSF member signaling and highlight its importance in different EC subsets in LN organogenesis and function during health, and in lymphocyte activation and tertiary lymphoid structure formation during inflammation.
Collapse
Affiliation(s)
- Kim C M Jeucken
- Amsterdam Rheumatology and Immunology Center (ARC), Department of Rheumatology and Clinical Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jasper J Koning
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center (ARC), Department of Rheumatology and Clinical Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
21
|
Sestito LF, Thomas SN. Biomaterials for Modulating Lymphatic Function in Immunoengineering. ACS Pharmacol Transl Sci 2019; 2:293-310. [PMID: 32259064 DOI: 10.1021/acsptsci.9b00047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Indexed: 12/13/2022]
Abstract
Immunoengineering is a rapidly growing and interdisciplinary field focused on developing tools to study and understand the immune system, then employing that knowledge to modulate immune response for the treatment of disease. Because of its roles in housing a substantial fraction of the body's lymphocytes, in facilitating immune cell trafficking, and direct immune modulatory functions, among others, the lymphatic system plays multifaceted roles in immune regulation. In this review, the potential for biomaterials to be applied to regulate the lymphatic system and its functions to achieve immunomodulation and the treatment of disease are described. Three related processes-lymphangiogenesis, lymphatic vessel contraction, and lymph node remodeling-are specifically explored. The molecular regulation of each process and their roles in pathologies are briefly outlined, with putative therapeutic targets and the lymphatic remodeling that can result from disease highlighted. Applications of biomaterials that harness these pathways for the treatment of disease via immunomodulation are discussed.
Collapse
Affiliation(s)
- Lauren F Sestito
- Wallace H. Coulter Department of Biomedical Engineering, George W. Woodruff School of Mechanical Engineering, and Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, Georgia 30332, United States.,Department of Biomedical Engineering, Emory University, 201 Dowman Drive, Atlanta, Georgia 30322, United States
| | - Susan N Thomas
- Wallace H. Coulter Department of Biomedical Engineering, George W. Woodruff School of Mechanical Engineering, and Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, Georgia 30332, United States.,Department of Biomedical Engineering, Emory University, 201 Dowman Drive, Atlanta, Georgia 30322, United States.,Wallace H. Coulter Department of Biomedical Engineering, George W. Woodruff School of Mechanical Engineering, and Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, Georgia 30332, United States.,Wallace H. Coulter Department of Biomedical Engineering, George W. Woodruff School of Mechanical Engineering, and Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, Georgia 30332, United States.,Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road NW, Atlanta, Georgia 30322, United States
| |
Collapse
|
22
|
Farnsworth RH, Karnezis T, Maciburko SJ, Mueller SN, Stacker SA. The Interplay Between Lymphatic Vessels and Chemokines. Front Immunol 2019; 10:518. [PMID: 31105685 PMCID: PMC6499173 DOI: 10.3389/fimmu.2019.00518] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/26/2019] [Indexed: 12/21/2022] Open
Abstract
Chemokines are a family of small protein cytokines that act as chemoattractants to migrating cells, in particular those of the immune system. They are categorized functionally as either homeostatic, constitutively produced by tissues for basal levels of cell migration, or inflammatory, where they are generated in association with a pathological inflammatory response. While the extravasation of leukocytes via blood vessels is a key step in cells entering the tissues, the lymphatic vessels also serve as a conduit for cells that are recruited and localized through chemoattractant gradients. Furthermore, the growth and remodeling of lymphatic vessels in pathologies is influenced by chemokines and their receptors expressed by lymphatic endothelial cells (LECs) in and around the pathological tissue. In this review we summarize the diverse role played by specific chemokines and their receptors in shaping the interaction of lymphatic vessels, immune cells, and other pathological cell types in physiology and disease.
Collapse
Affiliation(s)
- Rae H Farnsworth
- Tumor Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Tara Karnezis
- Lymphatic and Regenerative Medicine Laboratory, O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Simon J Maciburko
- Lymphatic and Regenerative Medicine Laboratory, O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Melbourne, VIC, Australia
| | - Steven A Stacker
- Tumor Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
23
|
Tay MHD, Lim SYJ, Leong YFI, Thiam CH, Tan KW, Torta FT, Narayanaswamy P, Wenk M, Angeli V. Halted Lymphocyte Egress via Efferent Lymph Contributes to Lymph Node Hypertrophy During Hypercholesterolemia. Front Immunol 2019; 10:575. [PMID: 30972070 PMCID: PMC6446103 DOI: 10.3389/fimmu.2019.00575] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/04/2019] [Indexed: 11/13/2022] Open
Abstract
Dyslipidemia is a central component of atherosclerosis and metabolic syndrome linked to chronic inflammation and immune dysfunction. Previously, we showed that hypercholesterolemic apolipoprotein E knock out (apoE−/−) mice exhibit systemic effects including skin inflammation and hypertrophic lymph nodes (LNs). However, the mechanisms accounting for LN hypertrophy in these mice remain unknown. Here, we show that hypercholesterolemia led to the accumulation of lymphocytes in LNs. We excluded that the increased number of lymphocytes in expanded LNs resulted from increased lymphocyte proliferation or entry into those LNs. Instead, we demonstrated that the egress of lymphocytes from the enlarged LN of apoE−/− mice was markedly decreased. Impairment in efferent lymphatic emigration of lymphocytes from LNs resulted from an aberrant expansion of cortical and medullary sinuses that became hyperplastic. Moreover, CCL21 was more abundant on these enlarged sinuses whereas lymph levels of sphingosine 1 phosphate (S1P) were decreased in apoE−/− mice. Normal LN size, lymphatic density and S1P levels were restored by reversing hypercholesterolemia. Thus, systemic changes in cholesterol can sequester lymphocytes in tissue draining LNs through the extensive remodeling of lymphatic sinuses and alteration of the balance between retention/egress signals leading to LN hypertrophy which subsequently may contribute to poor immunity. This study further illustrates the role of lymphatic vessels in immunity through the regulation of immune cell trafficking.
Collapse
Affiliation(s)
- Meng Hwee Daniel Tay
- Immunology Programme, Department of Microbiology and Immunology, Life Science Institute, Yoon Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Swee Yeng Jason Lim
- Immunology Programme, Department of Microbiology and Immunology, Life Science Institute, Yoon Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yew Fai Ivan Leong
- Immunology Programme, Department of Microbiology and Immunology, Life Science Institute, Yoon Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chung Hwee Thiam
- Immunology Programme, Department of Microbiology and Immunology, Life Science Institute, Yoon Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kar Wai Tan
- Immunology Programme, Department of Microbiology and Immunology, Life Science Institute, Yoon Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Federico Tesio Torta
- Department of Biochemistry, Life Science Institute, SLING-Singapore Lipidomics Incubator, Yoon Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Pradeep Narayanaswamy
- Department of Biochemistry, Life Science Institute, SLING-Singapore Lipidomics Incubator, Yoon Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Markus Wenk
- Department of Biochemistry, Life Science Institute, SLING-Singapore Lipidomics Incubator, Yoon Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Véronique Angeli
- Department of Biochemistry, Life Science Institute, SLING-Singapore Lipidomics Incubator, Yoon Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
24
|
Lucas ED, Tamburini BAJ. Lymph Node Lymphatic Endothelial Cell Expansion and Contraction and the Programming of the Immune Response. Front Immunol 2019; 10:36. [PMID: 30740101 PMCID: PMC6357284 DOI: 10.3389/fimmu.2019.00036] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/08/2019] [Indexed: 12/12/2022] Open
Abstract
Lymphatic endothelial cells (LECs) form the structure of the lymphatic vessels and the sinuses of the lymph nodes, positioning them to be key players in many different aspects of the immune response. Following an inflammatory stimulus, LECs produce chemokines that recruit immune cells to the lymph nodes. The recruitment of immune cells aids in the coordination of both LEC and lymph node expansion and contraction. More recent data has demonstrated that to coordinate LEC division and death, cell surface molecules, such as PD-L1 and interferon receptors, are required. During homeostasis, LECs use PD-L1 to maintain peripheral tolerance by presenting specific peripheral tissue antigens in order to eliminate tissue specific responses. LECs also have the capacity to acquire, present, and exchange foreign antigens following viral infection or immunization. Here we will review how lymph node LECs require immune cells to expand and contract in response to an immune stimulus, the factors involved and how direct LEC-immune cell interactions are important for programming immunity.
Collapse
Affiliation(s)
- Erin D Lucas
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Beth A J Tamburini
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
25
|
Yang X, Lian K, Meng T, Liu X, Miao J, Tan Y, Yuan H, Hu F. Immune Adjuvant Targeting Micelles Allow Efficient Dendritic Cell Migration to Lymph Nodes for Enhanced Cellular Immunity. ACS APPLIED MATERIALS & INTERFACES 2018; 10:33532-33544. [PMID: 30192498 DOI: 10.1021/acsami.8b10081] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Cellular immunity is essential for the effectiveness of vaccines against cancer. After capture of vaccines, dendritic cells (DCs) have to migrate to lymph nodes via chemokine receptor type 7 (CCR7). Subsequently, DCs present cytosolic antigens via major histocompatibility complex class I (MHC I) molecules to induce cellular immunity. However, various vaccines fail to induce potent cellular immunity due to insufficient MHC I-restricted antigen presentation and limitations of immune adjuvants. Hence, we constructed novel immune adjuvant targeting micelles (M-COSA) to targeted codeliver antigen ovalbumin (OVA) and plasmid DNA encoding CCR7 (CCR7 pDNA) to the cytosol of DCs, thus promoting DC migration to lymph nodes to boost MHC I-restricted antigen presentation. M-COSA exhibited adjuvant activity and demonstrated more efficient DC cellular uptake compared with COSA. M-COSA/OVA/pDNA increased costimulatory molecule expression and cytokine secretion, resulting in DC activation and maturation. Moreover, antigens and pDNA, which were encapsulated in micelles, escaped from the endosome into the cytoplasm to achieve MHC I-restricted antigen presentation and increase CCR7 expression. The number of CD8+ T cells, which was positively correlated with tumor rejection, was notably increased and tumor growth was dramatically suppressed after vaccination with M-COSA/OVA/pDNA. In summary, M-COSA/OVA/pDNA micelles, which allow DC targeting and efficient DC migration to lymph nodes to enhance cellular immunity, exhibit effective tumor inhibition and lay the foundation for novel vaccine design.
Collapse
Affiliation(s)
| | | | | | | | | | - Yanan Tan
- Ocean College , Zhejiang University , Zhoushan 316021 , China
| | | | | |
Collapse
|
26
|
Lucas ED, Finlon JM, Burchill MA, McCarthy MK, Morrison TE, Colpitts TM, Tamburini BAJ. Type 1 IFN and PD-L1 Coordinate Lymphatic Endothelial Cell Expansion and Contraction during an Inflammatory Immune Response. THE JOURNAL OF IMMUNOLOGY 2018; 201:1735-1747. [PMID: 30045970 DOI: 10.4049/jimmunol.1800271] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/09/2018] [Indexed: 12/16/2022]
Abstract
Lymph node (LN) expansion during an immune response is a complex process that involves the relaxation of the fibroblastic network, germinal center formation, and lymphatic vessel growth. These processes require the stromal cell network of the LN to act deliberately to accommodate the influx of immune cells to the LN. The molecular drivers of these processes are not well understood. Therefore, we asked whether the immediate cytokines type 1 IFN produced during viral infection influence the lymphatic network of the LN in mice. We found that following an IFN-inducing stimulus such as viral infection or polyI:C, programmed cell death ligand 1 (PD-L1) expression is dynamically upregulated on lymphatic endothelial cells (LECs). We found that reception of type 1 IFN by LECs is important for the upregulation of PD-L1 of mouse and human LECs and the inhibition of LEC expansion in the LN. Expression of PD-L1 by LECs is also important for the regulation of LN expansion and contraction after an IFN-inducing stimulus. We demonstrate a direct role for both type 1 IFN and PD-L1 in inhibiting LEC division and in promoting LEC survival. Together, these data reveal a novel mechanism for the coordination of type 1 IFN and PD-L1 in manipulating LEC expansion and survival during an inflammatory immune response.
Collapse
Affiliation(s)
- Erin D Lucas
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045.,Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Jeffrey M Finlon
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Matthew A Burchill
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Mary K McCarthy
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Thomas E Morrison
- Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Tonya M Colpitts
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118; and.,Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| | - Beth A Jirón Tamburini
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; .,Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
27
|
Feng X, Classon C, Terán G, Yang Y, Li L, Chan S, Ribacke U, Rothfuchs AG, Coquet JM, Nylén S. Atrophy of skin-draining lymph nodes predisposes for impaired immune responses to secondary infection in mice with chronic intestinal nematode infection. PLoS Pathog 2018; 14:e1007008. [PMID: 29772005 PMCID: PMC5957330 DOI: 10.1371/journal.ppat.1007008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 04/03/2018] [Indexed: 11/18/2022] Open
Abstract
Intestinal nematodes suppress immune responses in the context of allergy, gut inflammation, secondary infection and vaccination. Several mechanisms have been proposed for this suppression including alterations in Th2 cell differentiation and increased Treg cell suppressive function. In this study, we show that chronic nematode infection leads to reduced peripheral responses to vaccination because of a generalized reduction in the available responsive lymphocyte pool. We found that superficial skin-draining lymph nodes (LNs) in mice that are chronically infected with the intestinal nematode Heligmosomides polygyrus, do not reach the same cellularity as worm-free mice upon subsequent BCG infection in the skin. B cells and T cells, all declined in skin-draining LN of H. polygyrus-infected mice, resulting in LNs atrophy and altered lymphocyte composition. Importantly, anti-helminthic treatment improved lymphocyte numbers in skin-draining LN, indicating that time after de-worming is critical to regain full-scale LN cellularity. De-worming, and time for the skin LN to recover cellularity, also mended responses to Bacille Calmette-Guerin (BCG) in the LN draining the footpad injection site. Thus, our findings show that chronic nematode infection leads to a paucity of lymphocytes in peripheral lymph nodes, which acts to reduce the efficacy of immune responses at these sites.
Collapse
Affiliation(s)
- Xiaogang Feng
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Cajsa Classon
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Graciela Terán
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lei Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Sherwin Chan
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Ulf Ribacke
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | | | - Jonathan M. Coquet
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Susanne Nylén
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
28
|
Kedl RM, Lindsay RS, Finlon JM, Lucas ED, Friedman RS, Tamburini BAJ. Migratory dendritic cells acquire and present lymphatic endothelial cell-archived antigens during lymph node contraction. Nat Commun 2017; 8:2034. [PMID: 29229919 PMCID: PMC5725486 DOI: 10.1038/s41467-017-02247-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 11/16/2017] [Indexed: 12/20/2022] Open
Abstract
Antigens derived from viral infection or vaccination can persist within a host for many weeks after resolution of the infection or vaccine responses. We previously identified lymphatic endothelial cells (LEC) as the repository for this antigen archival, yet LECs are unable to present their archived antigens to CD8+ T cells, and instead transfer their antigens to CD11c+ antigen-presenting cells (APC). Here we show that the exchange of archived antigens between LECs and APCs is mediated by migratory dendritic cells (DC). After vaccination, both migratory basic leucine zipper ATF-like transcription factor 3 (BatF3)-dependent and BatF3-independent DCs are responsible for antigen exchange and cross-presentation. However, exchange of archived viral antigens is mediated only by BatF3-dependent migratory DCs potentially acquiring apoptotic LECs. In conclusion, LEC-archived antigens are exchanged with migratory DCs, both directly and through LEC apoptosis, to cross-present archived antigens to circulating T cells. Viral infection and vaccination both induce lasting persistence of antigens for protective responses. Here the authors show that migratory dendritic cells, independent of the transcription factor BatF3 for their development, contribute to “archived antigen” exchange with lymphatic endothelial cells.
Collapse
Affiliation(s)
- Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Aurora, CO, 80045, USA.
| | - Robin S Lindsay
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Aurora, CO, 80045, USA.,Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA
| | - Jeffrey M Finlon
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, School of Medicine, 12700 E. 19th Ave., Aurora, CO, 80045, USA
| | - Erin D Lucas
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Aurora, CO, 80045, USA.,Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, School of Medicine, 12700 E. 19th Ave., Aurora, CO, 80045, USA
| | - Rachel S Friedman
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Aurora, CO, 80045, USA.,Department of Biomedical Research, National Jewish Health, 1400 Jackson Street, Denver, CO, 80206, USA
| | - Beth A Jirón Tamburini
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, 12800 E. 19th Ave, Aurora, CO, 80045, USA. .,Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, School of Medicine, 12700 E. 19th Ave., Aurora, CO, 80045, USA.
| |
Collapse
|
29
|
Fish scale-derived collagen patch promotes growth of blood and lymphatic vessels in vivo. Acta Biomater 2017; 63:246-260. [PMID: 28888665 DOI: 10.1016/j.actbio.2017.09.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/28/2017] [Accepted: 09/01/2017] [Indexed: 01/11/2023]
Abstract
In this study, Type I collagen was extracted from fish scales asa potential alternative source of collagen for tissue engineering applications. Since unmodified collagen typically has poor mechanical and degradation stability both in vitro and in vivo, additional methylation modification and 1,4-butanediol diglycidyl ether (BDE) crosslinking steps were used to improve the physicochemical properties of fish scale-derived collagen. Subsequently, in vivo studies using a murine model demonstrated the biocompatibility of the different fish scale-derived collagen patches. In general, favorable integration of the collagen patches to the surrounding tissues, with good infiltration of cells, blood vessels (BVs) and lymphatic vessels (LVs) were observed under growth factor-free conditions. Interestingly, significantly higher (p<0.05) number of LVs was found to be more abundant around collagen patches with methylation modification and BDE crosslinking. Overall, we have demonstrated the potential application of fish scale-derived collagen as a promising scaffolding material for various biomedical applications. STATEMENT OF SIGNIFICANCE Currently the most common sources of collagen are of bovine and porcine origins, although the industrial use of collagen obtained from non-mammalian species is growing in importance, particularly since they have a lower risk of disease transmission and are not subjected to any cultural or religious constraints. However, unmodified collagen typically has poor mechanical and degradation stability both in vitro and in vivo. Hence, in this study, Type I collagen was successfully extracted from fish scales and chemically modified and crosslinked. In vitro studies showed overall improvement in the physicochemical properties of the material, whilst in vivo implantation studies showed improvements in the growth of blood and lymphatic host vessels in the vicinity of the implants.
Collapse
|
30
|
Interactions between fibroblastic reticular cells and B cells promote mesenteric lymph node lymphangiogenesis. Nat Commun 2017; 8:367. [PMID: 28848229 PMCID: PMC5573728 DOI: 10.1038/s41467-017-00504-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 06/27/2017] [Indexed: 01/18/2023] Open
Abstract
Lymphatic growth (lymphangiogenesis) within lymph nodes functions to promote dendritic cell entry and effector lymphocyte egress in response to infection or inflammation. Here we demonstrate a crucial role for lymphotoxin-beta receptor (LTβR) signaling to fibroblastic reticular cells (FRCs) by lymphotoxin-expressing B cells in driving mesenteric lymph node lymphangiogenesis following helminth infection. LTβR ligation on fibroblastic reticular cells leads to the production of B-cell-activating factor (BAFF), which synergized with interleukin-4 (IL-4) to promote the production of the lymphangiogenic factors, vascular endothelial growth factors (VEGF)-A and VEGF-C, by B cells. In addition, the BAFF-IL-4 synergy augments expression of lymphotoxin by antigen-activated B cells, promoting further B cell–fibroblastic reticular cell interactions. These results underlie the importance of lymphotoxin-dependent B cell–FRC cross talk in driving the expansion of lymphatic networks that function to promote and maintain immune responsiveness. The growth of lymph nodes in response to infection requires lymphangiogenesis. Dubey et al. show that the mesenteric lymph node lymphangiogenesis upon helminth infection depends on the signaling loop between the B and fibroblastic reticular cells (FRCs), whereby the FRCs respond to lymphotoxin secreted by B cells by releasing B cell activating factor.
Collapse
|
31
|
Magnuson AM, Regan DP, Fouts JK, Booth AD, Dow SW, Foster MT. Diet-induced obesity causes visceral, but not subcutaneous, lymph node hyperplasia via increases in specific immune cell populations. Cell Prolif 2017; 50. [PMID: 28762561 DOI: 10.1111/cpr.12365] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 06/06/2017] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES The spatial proximity of adipose depots to secondary lymph nodes allows a unique relation between the two systems. Obesity, predominately visceral adiposity, links to numerous diseases; hence, we postulate that secondary lymphatics within this region contributes to disease risk. MATERIAL AND METHODS Male C57BL/6 mice were fed standard CHOW (18% kcal fat) or Western diet (45% kcal fat) for 7 weeks. Visceral and subcutaneous lymph nodes and associated adipose depots they occupy were excised. Lymph node morphology and resident immune cell populations were characterized via histopathology, immunofluorescence and flow cytometry. Adipose tissue immune cell populations were also characterized. RESULTS Obesity caused lymph node expansion, increased viable cell number and deviations in immune cell populations. These alterations were exclusive to visceral lymph nodes. Notably, pro-inflammatory antigen presenting cells and regulatory T cells increased in number in the visceral lymph node. Obesity, however, reduced T regulatory cells in visceral lymph nodes. The visceral adipose depot also had greater reactivity towards HFD than subcutaneous, with a greater percent of macrophages, dendritic and CD8+ T cells. Immune cell number, in both the visceral and subcutaneous, however decreased as adipose depots enlarged. CONCLUSION Overall, HFD has a greater influence on visceral cavity than the subcutaneous. In the visceral lymph node, but not subcutaneous, HFD-induced obesity decreased cell populations that suppressed immune function while increasing those that regulate/activate immune response.
Collapse
Affiliation(s)
- A M Magnuson
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, 80523, USA
| | - D P Regan
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, 80523, USA
| | - J K Fouts
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, 80523, USA
| | - A D Booth
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, 80523, USA
| | - S W Dow
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, 80523, USA
| | - M T Foster
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, 80523, USA
| |
Collapse
|
32
|
Yeo KP, Angeli V. Bidirectional Crosstalk between Lymphatic Endothelial Cell and T Cell and Its Implications in Tumor Immunity. Front Immunol 2017; 8:83. [PMID: 28220121 PMCID: PMC5292621 DOI: 10.3389/fimmu.2017.00083] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 01/18/2017] [Indexed: 12/17/2022] Open
Abstract
Lymphatic vessels have been traditionally considered as passive transporters of fluid and lipids. However, it is apparent from recent literature that the function of lymphatic vessels is not only restricted to fluid balance homeostasis but also extends to regulation of immune cell trafficking, antigen presentation, tolerance, and immunity, all which may impact the progression of inflammatory responses and diseases such as cancer. The lymphatic system and the immune system are intimately connected, and there is emergent evidence for a crosstalk between T cell and lymphatic endothelial cell (LEC). This review describes how LECs in lymph nodes can affect multiple functional properties of T cells and the impact of these LEC-driven effects on adaptive immunity and, conversely, how T cells can modulate LEC growth. The significance of such crosstalk between T cells and LECs in cancer will also be discussed.
Collapse
Affiliation(s)
- Kim Pin Yeo
- Immunology Programme, Department of Microbiology and Immunology, Yoon Loo Lin School of Medicine, Life Science Institute, National University of Singapore , Singapore , Singapore
| | - Veronique Angeli
- Immunology Programme, Department of Microbiology and Immunology, Yoon Loo Lin School of Medicine, Life Science Institute, National University of Singapore , Singapore , Singapore
| |
Collapse
|
33
|
Abstract
The lymphatic vasculature is not considered a formal part of the immune system, but it is critical to immunity. One of its major roles is in the coordination of the trafficking of antigen and immune cells. However, other roles in immunity are emerging. Lymphatic endothelial cells, for example, directly present antigen or express factors that greatly influence the local environment. We cover these topics herein and discuss how other properties of the lymphatic vasculature, such as mechanisms of lymphatic contraction (which immunologists traditionally do not take into account), are nonetheless integral in the immune system. Much is yet unknown, and this nascent subject is ripe for exploration. We argue that to consider the impact of lymphatic biology in any given immunological interaction is a key step toward integrating immunology with organ physiology and ultimately many complex pathologies.
Collapse
Affiliation(s)
- Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110;
| | - Stoyan Ivanov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110;
| | - Bernd H Zinselmeyer
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110;
| | - Joshua P Scallan
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida 33612
| |
Collapse
|
34
|
Lymphadenectomy promotes tumor growth and cancer cell dissemination in the spontaneous RET mouse model of human uveal melanoma. Oncotarget 2016; 6:44806-18. [PMID: 26575174 PMCID: PMC4792593 DOI: 10.18632/oncotarget.6326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/23/2015] [Indexed: 01/01/2023] Open
Abstract
Resection of infiltrated tumor-draining lymph nodes (TDLNs) is a standard practice for the treatment of several cancers including breast cancer and melanoma. However, many randomized prospective trials have failed to show convincing clinical benefits associated with LN removal and the role of TDLNs in cancer dissemination is poorly understood. Here, we found in a well-characterized spontaneous mouse model of uveal melanoma that the growth of the primary tumor was accompanied by increased lymphangiogenesis and cancer cell colonization in the LNs draining the eyes. But, unexpectedly, early resection of the TDLNs increased the growth of the primary tumor and associated blood vessels as well as promoted cancer cell survival and dissemination. These effects were accompanied by increased tumor cell proliferation and expression of phosphorylated AKT. Topical application of a broad anti-inflammatory agent, Tobradex, or an oral treatment with cyclooxygenase-2 specific inhibitor, Celecoxib, reversed tumor progression observed after complete lymphadenectomy. Our study confirms the importance of tumor homeostasis in cancer progression by showing the enhancing effects of TDLN removal on tumor growth and cancer cell dissemination, and suggests that TDLN resection may only be beneficial if used in combination with anti-inflammatory drugs such as Tobradex and Celecoxib.
Collapse
|
35
|
Jafarnejad M, Woodruff MC, Zawieja DC, Carroll MC, Moore JE. Modeling Lymph Flow and Fluid Exchange with Blood Vessels in Lymph Nodes. Lymphat Res Biol 2016; 13:234-47. [PMID: 26683026 DOI: 10.1089/lrb.2015.0028] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Lymph nodes (LNs) are positioned strategically throughout the body as critical mediators of lymph filtration and immune response. Lymph carries cytokines, antigens, and cells to the downstream LNs, and their effective delivery to the correct location within the LN directly impacts the quality and quantity of immune response. Despite the importance of this system, the flow patterns in LN have never been quantified, in part because experimental characterization is so difficult. METHODS AND RESULTS To achieve a more quantitative knowledge of LN flow, a computational flow model has been developed based on the mouse popliteal LN, allowing for a parameter sensitivity analysis to identify the important system characteristics. This model suggests that about 90% of the lymph takes a peripheral path via the subcapsular and medullary sinuses, while fluid perfusing deeper into the paracortex is sequestered by parenchymal blood vessels. Fluid absorption by these blood vessels under baseline conditions was driven mainly by oncotic pressure differences between lymph and blood, although the magnitude of fluid transfer is highly dependent on blood vessel surface area. We also predict that the hydraulic conductivity of the medulla, a parameter that has never been experimentally measured, should be at least three orders of magnitude larger than that of the paracortex to ensure physiologic pressures across the node. CONCLUSIONS These results suggest that structural changes in the LN microenvironment, as well as changes in inflow/outflow conditions, dramatically alter the distribution of lymph, cytokines, antigens, and cells within the LN, with great potential for modulating immune response.
Collapse
Affiliation(s)
| | | | - David C Zawieja
- 3 Department of Medical Physiology, Texas A&M Health Science Center , Temple, Texas
| | - Michael C Carroll
- 4 Program in Cellular and Molecular Medicine, Boston Childrens Hospital , Harvard Medical School, Boston, Massachusetts
| | - J E Moore
- 1 Department of Bioengineering, Imperial College , London, United Kingdom
| |
Collapse
|
36
|
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
|
37
|
Weinkopff T, Konradt C, Christian DA, Discher DE, Hunter CA, Scott P. Leishmania major Infection-Induced VEGF-A/VEGFR-2 Signaling Promotes Lymphangiogenesis That Controls Disease. THE JOURNAL OF IMMUNOLOGY 2016; 197:1823-31. [PMID: 27474074 DOI: 10.4049/jimmunol.1600717] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/29/2016] [Indexed: 01/09/2023]
Abstract
Cutaneous leishmaniasis causes a spectrum of diseases from self-healing to severe nonhealing lesions. Defining the factors contributing to lesion resolution may help in developing new therapies for those patients with chronic disease. We found that infection with Leishmania major increases the expression of vascular endothelial growth factor-A and vascular endothelial growth factor receptor (VEGFR)-2 and is associated with significant changes in the blood and lymphatic vasculature at the site of infection. Ab blockade of VEGFR-2 during infection led to a reduction in lymphatic endothelial cell proliferation and simultaneously increased lesion size without altering the parasite burden. These data show that L. major infection initiates enhanced vascular endothelial growth factor-A/VEGFR-2 signaling and suggest that VEGFR-2-dependent lymphangiogenesis is a mechanism that restricts tissue inflammation in leishmaniasis.
Collapse
Affiliation(s)
- Tiffany Weinkopff
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Christoph Konradt
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - David A Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Dennis E Discher
- Biophysical Eng'g Labs, University of Pennsylvania, Philadelphia, PA 19104
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| |
Collapse
|
38
|
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
|
39
|
Lymphoid Tissue Mesenchymal Stromal Cells in Development and Tissue Remodeling. Stem Cells Int 2016; 2016:8419104. [PMID: 27190524 PMCID: PMC4846763 DOI: 10.1155/2016/8419104] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 03/20/2016] [Indexed: 12/28/2022] Open
Abstract
Secondary lymphoid organs (SLOs) are sites that facilitate cell-cell interactions required for generating adaptive immune responses. Nonhematopoietic mesenchymal stromal cells have been shown to play a critical role in SLO function, organization, and tissue homeostasis. The stromal microenvironment undergoes profound remodeling to support immune responses. However, chronic inflammatory conditions can promote uncontrolled stromal cell activation and aberrant tissue remodeling including fibrosis, thus leading to tissue damage. Despite recent advancements, the origin and role of mesenchymal stromal cells involved in SLO development and remodeling remain unclear.
Collapse
|
40
|
Song E, Seo H, Choe K, Hwang Y, Ahn J, Ahn S, Kim P. Optical clearing based cellular-level 3D visualization of intact lymph node cortex. BIOMEDICAL OPTICS EXPRESS 2015; 6:4154-64. [PMID: 26504662 PMCID: PMC4605071 DOI: 10.1364/boe.6.004154] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/19/2015] [Accepted: 09/21/2015] [Indexed: 05/03/2023]
Abstract
Lymph node (LN) is an important immune organ that controls adaptive immune responses against foreign pathogens and abnormal cells. To facilitate efficient immune function, LN has highly organized 3D cellular structures, vascular and lymphatic system. Unfortunately, conventional histological analysis relying on thin-sliced tissue has limitations in 3D cellular analysis due to structural disruption and tissue loss in the processes of fixation and tissue slicing. Optical sectioning confocal microscopy has been utilized to analyze 3D structure of intact LN tissue without physical tissue slicing. However, light scattering within biological tissues limits the imaging depth only to superficial portion of LN cortex. Recently, optical clearing techniques have shown enhancement of imaging depth in various biological tissues, but their efficacy for LN are remained to be investigated. In this work, we established optical clearing procedure for LN and achieved 3D volumetric visualization of the whole cortex of LN. More than 4 times improvement in imaging depth was confirmed by using LN obtained from H2B-GFP/actin-DsRed double reporter transgenic mouse. With adoptive transfer of GFP expressing B cells and DsRed expressing T cells and fluorescent vascular labeling by anti-CD31 and anti-LYVE-1 antibody conjugates, we successfully visualized major cellular-level structures such as T-cell zone, B-cell follicle and germinal center. Further, we visualized the GFP expressing metastatic melanoma cell colony, vasculature and lymphatic vessels in the LN cortex.
Collapse
|
41
|
Visualization and dynamic analysis of host-pathogen interactions. Curr Opin Immunol 2014; 29:8-15. [PMID: 24705104 DOI: 10.1016/j.coi.2014.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 03/05/2014] [Accepted: 03/06/2014] [Indexed: 12/15/2022]
Abstract
To contain invading microbes, the immune system must efficiently recognize the presence of the invader, mobilize cells to the site of infection, and deploy effector function. Rare antigen-specific T cells must find small numbers of antigen-presenting cells, proliferate and differentiate in secondary lymphoid tissues, then traffic to the infected site and be activated by antigen again to contribute to host defense. Our understanding of the dynamic processes involved has benefited enormously from tools that enable the visualization of cell location and behavior in complex tissue environments. Here we summarize recent insights into T cell trafficking and migration through secondary lymphoid organs and at peripheral infection sites, highlighting cell-intrinsic and extrinsic factors optimizing antigen surveillance at steady-state and delivery of an effector response during infection.
Collapse
|
42
|
Benahmed F, Chyou S, Dasoveanu D, Chen J, Kumar V, Iwakura Y, Lu TT. Multiple CD11c+ cells collaboratively express IL-1β to modulate stromal vascular endothelial growth factor and lymph node vascular-stromal growth. THE JOURNAL OF IMMUNOLOGY 2014; 192:4153-63. [PMID: 24659690 DOI: 10.4049/jimmunol.1301765] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lymphadenopathy in autoimmune and other lymphoproliferative diseases is in part characterized by immunoblasts and vascular proliferation. The lymph node vasculature, along with the nonvascular stromal compartment, supports lymphocyte function, and targeting vascular-stromal expansion in inflamed nodes may modulate lymphocyte function in disease. CD11c(+) cells are essential for vascular-stromal proliferation and the upregulation of vascular endothelial growth factor (VEGF) needed for vascular proliferation. However, targetable CD11c(+) cell-derived molecular mediators, the identity of relevant CD11c(+) cells, and whether CD11c(+) cells directly stimulate VEGF-expressing stromal cells are poorly understood. In this study we show that CD11c(+) CD11b(+) CCR2-dependent monocytes and CCR7-dependent dendritic cells express IL-1β. IL-1β blockade, IL-1β deficiency in radiosensitive cells, and CCR2/CCR7 double deficiency but not single deficiency all attenuate immunization-induced vascular-stromal proliferation. gp38(+) stromal fibroblastic reticular cells (FRCs) that express VEGF are enriched for Thy1(+) cells and partially overlap with CCL21-expressing FRCs, and FRC VEGF is attenuated with IL-1β deficiency or blockade. IL-1β localizes to the outer borders of the T zone, where VEGF-expressing cells are also enriched. Ex vivo, CD11b(+) cells enriched for IL-1β(+) cells can directly induce cultured gp38(+)Thy1(+) FRCs to upregulate VEGF. Taken together, these results suggest a mechanism whereby multiple recruited CD11c(+) populations express IL-1β and directly modulate FRC function to help promote the initiation of vascular-stromal growth in stimulated lymph nodes. These data provide new insight into how CD11c(+) cells regulate the lymph node vascular-stromal compartment, add to the evolving understanding of functional stromal subsets, and suggest a possible utility for IL-1β blockade in preventing inflammatory lymph node growth.
Collapse
Affiliation(s)
- Fairouz Benahmed
- Autoimmunity and Inflammation Program and Pediatric Rheumatology, Hospital for Special Surgery, New York, NY 10021
| | | | | | | | | | | | | |
Collapse
|
43
|
Card CM, Yu SS, Swartz MA. Emerging roles of lymphatic endothelium in regulating adaptive immunity. J Clin Invest 2014; 124:943-52. [PMID: 24590280 DOI: 10.1172/jci73316] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Emerging research on the roles of stromal cells in modulating adaptive immune responses has included a new focus on lymphatic endothelial cells (LECs). LECs are presumably the first cells that come into direct contact with peripheral antigens, cytokines, danger signals, and immune cells travelling from peripheral tissues to lymph nodes. LECs can modulate dendritic cell function, present antigens to T cells on MHC class I and MHC class II molecules, and express immunomodulatory cytokines and receptors, which suggests that their roles in adaptive immunity are far more extensive than previously realized. This Review summarizes the emergent evidence that LECs are important in maintaining peripheral tolerance, limiting and resolving effector T cell responses, and modulating leukocyte function.
Collapse
|
44
|
Kim H, Kataru RP, Koh GY. Inflammation-associated lymphangiogenesis: a double-edged sword? J Clin Invest 2014; 124:936-42. [PMID: 24590279 DOI: 10.1172/jci71607] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Lymphangiogenesis and lymphatic vessel remodeling are complex biological processes frequently observed during inflammation. Accumulating evidence indicates that inflammation-associated lymphangiogenesis (IAL) is not merely an endpoint event, but actually a phenomenon actively involved in the pathophysiology of various inflammatory disorders. The VEGF-C/VEGFR-3 and VEGF-A/VEGF-R2 signaling pathways are two of the best-studied pathways in IAL. Methods targeting these molecules, such as prolymphangiogenic or antilymphatic treatments, were found to be beneficial in various preclinical and/or clinical studies. This Review focuses on the most recent achievements in the fields of lymphatic biology relevant to inflammatory conditions. Additionally, preclinical and clinical therapies that modulate IAL are summarized.
Collapse
|
45
|
Lu TT, Browning JL. Role of the Lymphotoxin/LIGHT System in the Development and Maintenance of Reticular Networks and Vasculature in Lymphoid Tissues. Front Immunol 2014; 5:47. [PMID: 24575096 PMCID: PMC3920476 DOI: 10.3389/fimmu.2014.00047] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/27/2014] [Indexed: 01/08/2023] Open
Abstract
Lymphoid organs are meeting zones where lymphocytes come together and encounter antigens present in the blood and lymph or as delivered by cells migrating from the draining tissue bed. The exquisite efficiency of this process relies heavily on highly specialized anatomy to direct and position the various players. Gated entry and exit control access to these theaters and reticular networks and associated chemokines guide cells into the proper sections. Lymphoid tissues are remarkably plastic, being able to expand dramatically and then involute upon resolution of the danger. All of the reticular scaffolds and vascular and lymphatic components adapt accordingly. As such, the lymph node (LN) is a wonderful example of a physiologic remodeling process and is potentially a guide to study such elements in pathological settings such as fibrosis, chronic infection, and tumor metastasis. The lymphotoxin/LIGHT axis delivers critical differentiation signals that direct and hone differentiation of both reticular networks and the vasculature. Considerable progress has been made recently in understanding the mesenchymal differentiation pathways leading to these specialized networks and in the remodeling that occurs in reactive LNs. In this article, we will review some new advances in the area in terms of developmental, differentiation, and maintenance events mediated by this axis.
Collapse
Affiliation(s)
- Theresa T Lu
- Autoimmunity and Inflammation Program and Pediatric Rheumatology, Hospital for Special Surgery , New York, NY , USA ; Department of Microbiology and Immunology, Weill Cornell Medical College , New York, NY , USA
| | - Jeffrey L Browning
- Department of Microbiology and Section of Rheumatology, Boston University School of Medicine , Boston, MA , USA
| |
Collapse
|
46
|
Cohen JN, Tewalt EF, Rouhani SJ, Buonomo EL, Bruce AN, Xu X, Bekiranov S, Fu YX, Engelhard VH. Tolerogenic properties of lymphatic endothelial cells are controlled by the lymph node microenvironment. PLoS One 2014; 9:e87740. [PMID: 24503860 PMCID: PMC3913631 DOI: 10.1371/journal.pone.0087740] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 12/13/2013] [Indexed: 12/04/2022] Open
Abstract
Peripheral self-tolerance eliminates lymphocytes specific for tissue-specific antigens not encountered in the thymus. Recently, we demonstrated that lymphatic endothelial cells in mice directly express peripheral tissue antigens, including tyrosinase, and induce deletion of specific CD8 T cells via Programmed Death Ligand-1 (PD-L1). Here, we demonstrate that high-level expression of peripheral tissue antigens and PD-L1 is confined to lymphatic endothelial cells in lymph nodes, as opposed to tissue (diaphragm and colon) lymphatics. Lymphatic endothelial cells in the lymph node medullary sinus express the highest levels of peripheral tissue antigens and PD-L1, and are the only subpopulation that expresses tyrosinase epitope. The representation of lymphatic endothelial cells in the medullary sinus expressing high-level PD-L1, which is necessary for normal CD8 T cell deletion kinetics, is controlled by lymphotoxin-β receptor signaling and B cells. Lymphatic endothelial cells from neonatal mice do not express high-level PD-L1 or present tyrosinase epitope. This work uncovers a critical role for the lymph node microenvironment in endowing lymphatic endothelial cells with potent tolerogenic properties.
Collapse
Affiliation(s)
- Jarish N. Cohen
- Department of Microbiology, Immunology, and Cancer Biology and Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Eric F. Tewalt
- Department of Microbiology, Immunology, and Cancer Biology and Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Sherin J. Rouhani
- Department of Microbiology, Immunology, and Cancer Biology and Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Erica L. Buonomo
- Department of Microbiology, Immunology, and Cancer Biology and Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Amber N. Bruce
- Department of Microbiology, Immunology, and Cancer Biology and Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Xiaojiang Xu
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Stefan Bekiranov
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Yang-Xin Fu
- Department of Pathology, University of Chicago, Chicago, Illinois, United States of America
| | - Victor H. Engelhard
- Department of Microbiology, Immunology, and Cancer Biology and Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
47
|
Inflammatory lymphangiogenesis: cellular mediators and functional implications. Angiogenesis 2014; 17:373-81. [DOI: 10.1007/s10456-014-9419-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 01/15/2014] [Indexed: 10/25/2022]
|
48
|
Trapping of naive lymphocytes triggers rapid growth and remodeling of the fibroblast network in reactive murine lymph nodes. Proc Natl Acad Sci U S A 2013; 111:E109-18. [PMID: 24367096 DOI: 10.1073/pnas.1312585111] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Adaptive immunity is initiated in T-cell zones of secondary lymphoid organs. These zones are organized in a rigid 3D network of fibroblastic reticular cells (FRCs) that are a rich cytokine source. In response to lymph-borne antigens, draining lymph nodes (LNs) expand several folds in size, but the fate and role of the FRC network during immune response is not fully understood. Here we show that T-cell responses are accompanied by the rapid activation and growth of FRCs, leading to an expanded but similarly organized network of T-zone FRCs that maintains its vital function for lymphocyte trafficking and survival. In addition, new FRC-rich environments were observed in the expanded medullary cords. FRCs are activated within hours after the onset of inflammation in the periphery. Surprisingly, FRC expansion depends mainly on trapping of naïve lymphocytes that is induced by both migratory and resident dendritic cells. Inflammatory signals are not required as homeostatic T-cell proliferation was sufficient to trigger FRC expansion. Activated lymphocytes are also dispensable for this process, but can enhance the later growth phase. Thus, this study documents the surprising plasticity as well as the complex regulation of FRC networks allowing the rapid LN hyperplasia that is critical for mounting efficient adaptive immunity.
Collapse
|
49
|
Dieterich LC, Seidel CD, Detmar M. Lymphatic vessels: new targets for the treatment of inflammatory diseases. Angiogenesis 2013; 17:359-71. [PMID: 24212981 DOI: 10.1007/s10456-013-9406-1] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 10/30/2013] [Indexed: 11/29/2022]
Abstract
The lymphatic system plays an important role in the physiological control of the tissue fluid balance and in the initiation of immune responses. Recent studies have shown that lymphangiogenesis, the growth of new lymphatic vessels and/or the expansion of existing lymphatic vessels, is a characteristic feature of acute inflammatory reactions and of chronic inflammatory diseases. In these conditions, lymphatic vessel expansion occurs at the tissue level but also within the draining lymph nodes. Surprisingly, activation of lymphatic vessel function by delivery of vascular endothelial growth factor-C exerts anti-inflammatory effects in several models of cutaneous and joint inflammation. These effects are likely mediated by enhanced drainage of extravasated fluid and inflammatory cells, but also by lymphatic vessel-mediated modulation of immune responses. Although some of the underlying mechanisms are just beginning to be identified, lymphatic vessels have emerged as important targets for the development of new therapeutic strategies to treat inflammatory conditions. In this context, it is of great interest that some of the currently used anti-inflammatory drugs also potently activate lymphatic vessels.
Collapse
Affiliation(s)
- Lothar C Dieterich
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Wolfgang-Pauli-Strasse 10, HCI H 303, 8093, Zurich, Switzerland
| | | | | |
Collapse
|
50
|
Aebischer D, Iolyeva M, Halin C. The inflammatory response of lymphatic endothelium. Angiogenesis 2013; 17:383-93. [PMID: 24154862 DOI: 10.1007/s10456-013-9404-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 10/16/2013] [Indexed: 12/13/2022]
Abstract
Lymphatic vessels have traditionally been regarded as a rather inert drainage system, which just passively transports fluids, leukocytes and antigen. However, it is becoming increasingly clear that the lymphatic vasculature is highly dynamic and plays a much more active role in inflammatory and immune processes. Tissue inflammation induces a rapid, stimulus-specific upregulation of chemokines and adhesion molecules in lymphatic endothelial cells and a proliferative expansion of the lymphatic network in the inflamed tissue and in draining lymph nodes. Moreover, increasing evidence suggests that inflammation-induced changes in the lymphatic vasculature have a profound impact on the course of inflammatory and immune responses, by modulating fluid drainage, leukocyte migration or the removal of inflammatory mediators from tissues. In this review we will summarize and discuss current knowledge of the inflammatory response of lymphatic endothelium and of inflammation-induced lymphangiogenesis and the current perspective on the overall functional significance of these processes.
Collapse
Affiliation(s)
- David Aebischer
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Wolfgang-Pauli Str. 10, HCI H413, 8093, Zurich, Switzerland
| | | | | |
Collapse
|