101
|
Listeria-based hepatocellular carcinoma vaccine facilitates anti-PD-1 therapy by regulating macrophage polarization. Oncogene 2019; 39:1429-1444. [PMID: 31659256 DOI: 10.1038/s41388-019-1072-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/05/2019] [Accepted: 10/11/2019] [Indexed: 12/24/2022]
Abstract
Recently, patients with advanced cancers have been benefited greatly from immune checkpoint blockade immunotherapy. However, immune checkpoint blockade is still suboptimal in HCC treatment and more immune modifications are needed to achieve an efficient therapeutic goal. Here, we investigated the combined administration of a Listeria-based HCC vaccine, Lmdd-MPFG, and the anti-PD-1 immune checkpoint blockade antibody. We found that Lmdd-MPFG promoted the expression of PD-L1 in HCC cells but resensitized the tumor local T cell to respond to the anti-PD-1 immunotherapy. Mechanistically, the Lmdd-MPFG vaccine activates the NF-κB pathway in the tumor-associated macrophages (TAMs) through the TLR2 and MyD88 pathway, and recruits p62 to activate the autophagy pathway. The overall effect is skewing the TAMs from M2-polarized TAMs into the M1-polarized TAMs. Most importantly, it skewed the cytokine profiles into antitumor one in the tumor microenvironment (TME). This change restores the T-cell reactivity to the anti-PD-1 blockade. Our results suggested that Lmdd-MPFG combined with PD-1 blockade exerted synergistic antitumor effects through modifying TAMs in the TME and removing T-cell inhibitory signals, thereby providing a new potential strategy for HCC treatment.
Collapse
|
102
|
Takeda A, Hollmén M, Dermadi D, Pan J, Brulois KF, Kaukonen R, Lönnberg T, Boström P, Koskivuo I, Irjala H, Miyasaka M, Salmi M, Butcher EC, Jalkanen S. Single-Cell Survey of Human Lymphatics Unveils Marked Endothelial Cell Heterogeneity and Mechanisms of Homing for Neutrophils. Immunity 2019; 51:561-572.e5. [PMID: 31402260 DOI: 10.1016/j.immuni.2019.06.027] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/17/2019] [Accepted: 06/27/2019] [Indexed: 01/28/2023]
Abstract
Lymphatic vessels form a critical component in the regulation of human health and disease. While their functional significance is increasingly being recognized, the comprehensive heterogeneity of lymphatics remains uncharacterized. Here, we report the profiling of 33,000 lymphatic endothelial cells (LECs) in human lymph nodes (LNs) by single-cell RNA sequencing. Unbiased clustering revealed six major types of human LECs. LECs lining the subcapsular sinus (SCS) of LNs abundantly expressed neutrophil chemoattractants, whereas LECs lining the medullary sinus (MS) expressed a C-type lectin CD209. Binding of a carbohydrate Lewis X (CD15) to CD209 mediated neutrophil binding to the MS. The neutrophil-selective homing by MS LECs may retain neutrophils in the LN medulla and allow lymph-borne pathogens to clear, preventing their spread through LNs in humans. Our study provides a comprehensive characterization of LEC heterogeneity and unveils a previously undefined role for medullary LECs in human immunity.
Collapse
Affiliation(s)
- Akira Takeda
- MediCity Research Laboratory and Institute of Biomedicine, University of Turku, Turku, Finland
| | - Maija Hollmén
- MediCity Research Laboratory and Institute of Biomedicine, University of Turku, Turku, Finland
| | - Denis Dermadi
- Laboratory of Immunology and Vascular Biology, Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Junliang Pan
- Laboratory of Immunology and Vascular Biology, Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA; Veterans Affairs Palo Alto Health Care System and The Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Kevin Francis Brulois
- Laboratory of Immunology and Vascular Biology, Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Riina Kaukonen
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - Tapio Lönnberg
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - Pia Boström
- Department of Pathology, Turku University Hospital, Turku, Finland
| | - Ilkka Koskivuo
- Department of Plastic and General Surgery, Turku University Hospital, Turku, Finland
| | - Heikki Irjala
- Department of Otorhinolaryngology, Turku University Hospital and University of Turku, Finland
| | - Masayuki Miyasaka
- MediCity Research Laboratory and Institute of Biomedicine, University of Turku, Turku, Finland; Interdisciplinary Program for Biomedical Sciences, Institute for Academic Initiatives, Osaka University, Suita, Japan
| | - Marko Salmi
- MediCity Research Laboratory and Institute of Biomedicine, University of Turku, Turku, Finland
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA; Veterans Affairs Palo Alto Health Care System and The Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Sirpa Jalkanen
- MediCity Research Laboratory and Institute of Biomedicine, University of Turku, Turku, Finland.
| |
Collapse
|
103
|
Leachman SA, Hornyak TJ, Barsh G, Bastian BC, Brash DE, Cleaver JE, Cooper CD, D'Orazio JA, Fujita M, Holmen SL, Indra AK, Kraemer KH, Le Poole IC, Lo RS, Lund AW, Manga P, Pavan WJ, Setaluri V, Stemwedel CE, Kulesz-Martin MF. Melanoma to Vitiligo: The Melanocyte in Biology & Medicine-Joint Montagna Symposium on the Biology of Skin/PanAmerican Society for Pigment Cell Research Annual Meeting. J Invest Dermatol 2019; 140:269-274. [PMID: 31348921 DOI: 10.1016/j.jid.2019.03.1164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 10/26/2022]
Affiliation(s)
- Sancy A Leachman
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Thomas J Hornyak
- Research and Development Service, VA Maryland Health Care System, Baltimore, Maryland; Departments of Dermatology and Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Greg Barsh
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama; Department of Genetics, Stanford University, Stanford, California
| | - Boris C Bastian
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California; Department of Dermatology, University of California, San Francisco, San Francisco, California
| | - Douglas E Brash
- Departments of Therapeutic Radiology and Dermatology and Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - James E Cleaver
- Department of Dermatology, University of California, San Francisco, San Francisco, California
| | - Cynthia D Cooper
- School of Molecular Biosciences and College of Arts and Sciences, Washington State University Vancouver, Vancouver, Washington
| | - John A D'Orazio
- The Markey Cancer Center and the Departments of Toxicology and Cancer Biology and Pediatrics, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Mayumi Fujita
- Departments of Dermatology and Immunology & Microbiology, University of Colorado School of Medicine, Aurora, Colorado; Denver VA Medical Center, Denver, Colorado
| | - Sheri L Holmen
- Huntsman Cancer Institute and Departments of Oncological Sciences and Surgery, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Arup K Indra
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon; Department of Pharmaceutical Sciences, College of Pharmacy, Linus Pauling Institute, and Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon
| | - Kenneth H Kraemer
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - I Caroline Le Poole
- Oncology Research Institute, Loyola University Chicago, Maywood, Illinois; Departments of Dermatology and Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Roger S Lo
- Division of Dermatology, Department of Medicine, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
| | - Amanda W Lund
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon; Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon
| | - Prashiela Manga
- Ronald O. Perelman Department of Dermatology and Department of Cell Biology, New York University School of Medicine, New York, New York
| | - William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Clara E Stemwedel
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon
| | - Molly F Kulesz-Martin
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon; Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
104
|
Jiang X. Lymphatic vasculature in tumor metastasis and immunobiology. J Zhejiang Univ Sci B 2019; 21:3-11. [PMID: 31317681 PMCID: PMC6964999 DOI: 10.1631/jzus.b1800633] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/12/2019] [Indexed: 12/12/2022]
Abstract
Lymphatic vessels are essential for tissue fluid homeostasis, immune cell trafficking, and intestinal lipid absorption. The lymphatics have long been recognized to serve as conduits for distant tumor dissemination. However, recent findings suggest that the regional lymphatic vasculature also shapes the immune microenvironment of the tumor mass and potentiates immunotherapy. This review discusses the role of lymphatic vessels in tumor metastasis and tumor immunity.
Collapse
Affiliation(s)
- Xinguo Jiang
- VA Palo Alto Health Care System, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| |
Collapse
|
105
|
Abstract
Tumor-associated macrophages (TAMs) constitute a plastic and heterogeneous cell population of the tumor microenvironment (TME) that can account for up to 50% of some solid neoplasms. Most often, TAMs support disease progression and resistance to therapy by providing malignant cells with trophic and nutritional support. However, TAMs can mediate antineoplastic effects, especially in response to pharmacological agents that boost their phagocytic and oxidative functions. Thus, TAMs and their impact on the overall metabolic profile of the TME have a major influence on tumor progression and resistance to therapy, de facto constituting promising targets for the development of novel anticancer agents. Here, we discuss the metabolic circuitries whereby TAMs condition the TME to support tumor growth and how such pathways can be therapeutically targeted.
Collapse
|
106
|
Tamburini BAJ, Elder AM, Finlon JM, Winter AB, Wessells VM, Borges VF, Lyons TR. PD-1 Blockade During Post-partum Involution Reactivates the Anti-tumor Response and Reduces Lymphatic Vessel Density. Front Immunol 2019; 10:1313. [PMID: 31244852 PMCID: PMC6579890 DOI: 10.3389/fimmu.2019.01313] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/23/2019] [Indexed: 12/18/2022] Open
Abstract
Post-partum breast cancer patients, or breast cancer patients diagnosed within 10 years of last childbirth, are ~3-5 times more likely to develop metastasis in comparison to non-post-partum, or nulliparous, patients. Additionally, post-partum patients have increased tumor-associated lymphatic vessels and LN involvement, including when controlled for size of the primary tumor. In pre-clinical, immune-competent, mouse mammary tumor models of post-partum breast cancer (PPBC), tumor growth and lymphogenous tumor cell spread occur more rapidly in post-partum hosts. Here we report on PD-L1 expression by lymphatic endothelial cells and CD11b+ cells in the microenvironment of post-partum tumors, which is accompanied by an increase in PD-1 expression by T cells. Additionally, we observed increases in PD-L1 and PD-1 in whole mammary tissues during post-partum mammary gland involution; a known driver of post-partum tumor growth, invasion, and metastasis in pre-clinical models. Importantly, implantation of murine mammary tumor cells during post-partum mammary gland involution elicits a CD8+ T cell population that expresses both the co-inhibitory receptors PD-1 and Lag-3. However, upon anti-PD-1 treatment, during post-partum mammary gland involution, the involution-initiated promotional effects on tumor growth are reversed and the PD-1, Lag-3 double positive population disappears. Consequently, we observed an expansion of poly-functional CD8+ T cells that produced both IFNγ and TNFα. Finally, lymphatic vessel frequency decreased significantly following anti-PD-1 suggesting that anti-PD-1/PD-L1 targeted therapies may have efficacy in reducing tumor growth and dissemination in post-partum breast cancer patients.
Collapse
Affiliation(s)
- Beth A Jirón Tamburini
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States.,Department of Immunology and Microbiology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States
| | - Alan M Elder
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States.,Young Women's' Breast Cancer Translational Program and University of Colorado Cancer Center, Aurora, CO, United States
| | - Jeffrey M Finlon
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States
| | - Andrew B Winter
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States
| | - Veronica M Wessells
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States.,Young Women's' Breast Cancer Translational Program and University of Colorado Cancer Center, Aurora, CO, United States
| | - Virginia F Borges
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States.,Young Women's' Breast Cancer Translational Program and University of Colorado Cancer Center, Aurora, CO, United States
| | - Traci R Lyons
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Denver, CO, United States.,Young Women's' Breast Cancer Translational Program and University of Colorado Cancer Center, Aurora, CO, United States
| |
Collapse
|
107
|
Abstract
Lymphatic vessels collect interstitial fluid that has extravasated from blood vessels and return it to the circulatory system. Another important function of the lymphatic network is to facilitate immune cell migration and antigen transport from the periphery to draining lymph nodes. This migration plays a crucial role in immune surveillance, initiation of immune responses and tolerance. Here we discuss the significance and mechanisms of lymphatic migration of innate and adaptive immune cells in homeostasis, inflammation and cancer.
Collapse
Affiliation(s)
| | - Tatyana Chtanova
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia
- Faculty of Medicine, St. Vincent's Clinical School, University of New South Wales Sydney, Kensington, NSW, Australia
| |
Collapse
|
108
|
Garnier L, Gkountidi AO, Hugues S. Tumor-Associated Lymphatic Vessel Features and Immunomodulatory Functions. Front Immunol 2019; 10:720. [PMID: 31024552 PMCID: PMC6465591 DOI: 10.3389/fimmu.2019.00720] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/18/2019] [Indexed: 12/21/2022] Open
Abstract
The lymphatic system comprises a network of lymphoid tissues and vessels that drains the extracellular compartment of most tissues. During tumor development, lymphatic endothelial cells (LECs) substantially expand in response to VEGFR-3 engagement by VEGF-C produced in the tumor microenvironment, a process known as tumor-associated lymphangiogenesis. Lymphatic drainage from the tumor to the draining lymph nodes consequently increases, powering interstitial flow in the tumor stroma. The ability of a tumor to induce and activate lymphatic growth has been positively correlated with metastasis. Much effort has been made to identify genes responsible for tumor-associated lymphangiogenesis. Inhibition of lymphangiogenesis with soluble VEGFR-3 or with specific monoclonal antibodies decreases tumor spread to LNs in rodent models. Importantly, tumor-associated lymphatics do not only operate as tumor cell transporters but also play critical roles in anti-tumor immunity. Therefore, metastatic as well as primary tumor progression can be affected by manipulating tumor-associated lymphatic remodeling or function. Here, we review and discuss our current knowledge on the contribution of LECs immersed in the tumor microenvironment as immunoregulators, as well as a possible functional remodeling of LECs subsets depending on the organ microenvironment.
Collapse
Affiliation(s)
- Laure Garnier
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Anastasia-Olga Gkountidi
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Stephanie Hugues
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
109
|
Hunter MC, Teijeira A, Montecchi R, Russo E, Runge P, Kiefer F, Halin C. Dendritic Cells and T Cells Interact Within Murine Afferent Lymphatic Capillaries. Front Immunol 2019; 10:520. [PMID: 30967863 PMCID: PMC6440485 DOI: 10.3389/fimmu.2019.00520] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/26/2019] [Indexed: 11/13/2022] Open
Abstract
Afferent lymphatic vessels contribute to immunity by transporting antigen and leukocytes to draining lymph nodes (LNs) and are emerging as new players in the regulation of peripheral tolerance. Performing intravital microscopy in inflamed murine ear skin we found that migrating dendritic cells (DCs) and antigen-experienced effector T cells spend considerable time arresting or clustering within afferent lymphatic capillaries. We also observed that intralymphatic T cells frequently interacted with DCs. When imaging polyclonal T cells during an ongoing contact-hypersensitivity response, most intralymphatic DC-T cell interactions were short-lived. Conversely, during a delayed-type-hypersensitivity response, cognate antigen-bearing DCs engaged in long-lived MHCII-(I-A/I-E)-dependent interactions with antigen-specific T cells. Long-lived intralymphatic DC-T cell interactions reduced the speed of DC crawling but did not delay overall DC migration to draining LNs. While further consequences of these intralymphatic interactions still need to be explored, our findings suggest that lymphatic capillaries represent a unique compartment in which adaptive immune interaction and modulation occur.
Collapse
Affiliation(s)
| | - Alvaro Teijeira
- Institute of Pharmaceutical Sciences, ETH Zürich, Zurich, Switzerland
| | | | - Erica Russo
- Institute of Pharmaceutical Sciences, ETH Zürich, Zurich, Switzerland
| | - Peter Runge
- Institute of Pharmaceutical Sciences, ETH Zürich, Zurich, Switzerland
| | - Friedemann Kiefer
- Max Planck Institute for Molecular Biomedicine, Münster, Germany.,European Institute for Molecular Imaging - EIMI, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
110
|
|
111
|
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: 38] [Impact Index Per Article: 7.6] [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
|
112
|
|
113
|
Lane RS, Lund AW. Non-hematopoietic Control of Peripheral Tissue T Cell Responses: Implications for Solid Tumors. Front Immunol 2018; 9:2662. [PMID: 30498499 PMCID: PMC6249380 DOI: 10.3389/fimmu.2018.02662] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/29/2018] [Indexed: 12/16/2022] Open
Abstract
In response to pathological challenge, the host generates rapid, protective adaptive immune responses while simultaneously maintaining tolerance to self and limiting immune pathology. Peripheral tissues (e.g., skin, gut, lung) are simultaneously the first site of pathogen-encounter and also the location of effector function, and mounting evidence indicates that tissues act as scaffolds to facilitate initiation, maintenance, and resolution of local responses. Just as both effector and memory T cells must adapt to their new interstitial environment upon infiltration, tissues are also remodeled in the context of acute inflammation and disease. In this review, we present the biochemical and biophysical mechanisms by which non-hematopoietic stromal cells and extracellular matrix molecules collaborate to regulate T cell behavior in peripheral tissue. Finally, we discuss how tissue remodeling in the context of tumor microenvironments impairs T cell accumulation and function contributing to immune escape and tumor progression.
Collapse
Affiliation(s)
- Ryan S Lane
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, United States
| | - Amanda W Lund
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, United States.,Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, United States.,Department of Dermatology, Oregon Health and Science University, Portland, OR, United States.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|