1
|
Hudson WH, Wieland A. Technology meets TILs: Deciphering T cell function in the -omics era. Cancer Cell 2023; 41:41-57. [PMID: 36206755 PMCID: PMC9839604 DOI: 10.1016/j.ccell.2022.09.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/15/2022] [Accepted: 09/15/2022] [Indexed: 01/17/2023]
Abstract
T cells are at the center of cancer immunology because of their ability to recognize mutations in tumor cells and directly mediate cancer cell killing. Immunotherapies to rejuvenate exhausted T cell responses have transformed the clinical management of several malignancies. In parallel, the development of novel multidimensional analysis platforms, such as single-cell RNA sequencing and high-dimensional flow cytometry, has yielded unprecedented insights into immune cell biology. This convergence has revealed substantial heterogeneity of tumor-infiltrating immune cells in single tumors, across tumor types, and among individuals with cancer. Here we discuss the opportunities and challenges of studying the complex tumor microenvironment with -omics technologies that generate vast amounts of data, highlighting the opportunities and limitations of these technologies with a particular focus on interpreting high-dimensional studies of CD8+ T cells in the tumor microenvironment.
Collapse
Affiliation(s)
- William H Hudson
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Andreas Wieland
- Department of Otolaryngology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
2
|
Özcan A, Collado-Diaz V, Egholm C, Tomura M, Gunzer M, Halin C, Kolios AGA, Boyman O. CCR7-guided neutrophil redirection to skin-draining lymph nodes regulates cutaneous inflammation and infection. Sci Immunol 2022; 7:eabi9126. [PMID: 35119939 DOI: 10.1126/sciimmunol.abi9126] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neutrophils are the first nonresident effector immune cells that migrate to a site of infection or inflammation; however, improper control of neutrophil responses can cause considerable tissue damage. Here, we found that neutrophil responses in inflamed or infected skin were regulated by CCR7-dependent migration and phagocytosis of neutrophils in draining lymph nodes (dLNs). In mouse models of Toll-like receptor-induced skin inflammation and cutaneous Staphylococcus aureus infection, neutrophils migrated from the skin to the dLNs via lymphatic vessels in a CCR7-mediated manner. In the dLNs, these neutrophils were phagocytosed by lymph node-resident type 1 and type 2 conventional dendritic cells. CCR7 up-regulation on neutrophils was a conserved mechanism across different tissues and was induced by a broad range of microbial stimuli. In the context of cutaneous immune responses, disruption of CCR7 interactions by selective CCR7 deficiency of neutrophils resulted in increased antistaphylococcal immunity and aggravated skin inflammation. Thus, neutrophil homing to and clearance in skin-dLNs affects cutaneous immunity versus pathology.
Collapse
Affiliation(s)
- A Özcan
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland
| | - V Collado-Diaz
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - C Egholm
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland
| | - M Tomura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka 584-8540, Japan
| | - M Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Essen, Germany.,Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - C Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - A G A Kolios
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland
| | - O Boyman
- Department of Immunology, University Hospital Zurich, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Cuesta-Mateos C, Terrón F, Herling M. CCR7 in Blood Cancers - Review of Its Pathophysiological Roles and the Potential as a Therapeutic Target. Front Oncol 2021; 11:736758. [PMID: 34778050 PMCID: PMC8589249 DOI: 10.3389/fonc.2021.736758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/12/2021] [Indexed: 11/23/2022] Open
Abstract
According to the classical paradigm, CCR7 is a homing chemokine receptor that grants normal lymphocytes access to secondary lymphoid tissues such as lymph nodes or spleen. As such, in most lymphoproliferative disorders, CCR7 expression correlates with nodal or spleen involvement. Nonetheless, recent evidence suggests that CCR7 is more than a facilitator of lymphatic spread of tumor cells. Here, we review published data to catalogue CCR7 expression across blood cancers and appraise which classical and novel roles are attributed to this receptor in the pathogenesis of specific hematologic neoplasms. We outline why novel therapeutic strategies targeting CCR7 might provide clinical benefits to patients with CCR7-positive hematopoietic tumors.
Collapse
Affiliation(s)
- Carlos Cuesta-Mateos
- Immunology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria- Instituto la Princesa (IIS-IP), Madrid, Spain.,Immunological and Medicinal Products (IMMED S.L.), Madrid, Spain.,Catapult Therapeutics BV, Lelystad, Netherlands
| | - Fernando Terrón
- Immunological and Medicinal Products (IMMED S.L.), Madrid, Spain.,Catapult Therapeutics BV, Lelystad, Netherlands
| | - Marco Herling
- Clinic of Hematology and Cellular Therapy, University of Leipzig, Leipzig, Germany
| |
Collapse
|
4
|
Collado-Diaz V, Medina-Sanchez JD, Gkountidi AO, Halin C. Imaging leukocyte migration through afferent lymphatics. Immunol Rev 2021; 306:43-57. [PMID: 34708414 PMCID: PMC9298274 DOI: 10.1111/imr.13030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/11/2022]
Abstract
Afferent lymphatics mediate the transport of antigen and leukocytes, especially of dendritic cells (DCs) and T cells, from peripheral tissues to draining lymph nodes (dLNs). As such they play important roles in the induction and regulation of adaptive immunity. Over the past 15 years, great advances in our understanding of leukocyte trafficking through afferent lymphatics have been made through time‐lapse imaging studies performed in tissue explants and in vivo, allowing to visualize this process with cellular resolution. Intravital imaging has revealed that intralymphatic leukocytes continue to actively migrate once they have entered into lymphatic capillaries, as a consequence of the low flow conditions present in this compartment. In fact, leukocytes spend considerable time migrating, patrolling and interacting with the lymphatic endothelium or with other intralymphatic leukocytes within lymphatic capillaries. Cells typically only start to detach once they arrive in downstream‐located collecting vessels, where vessel contractions contribute to enhanced lymph flow. In this review, we will introduce the biology of afferent lymphatic vessels and report on the presumed significance of DC and T cell migration via this route. We will specifically highlight how time‐lapse imaging has contributed to the current model of lymphatic trafficking and the emerging notion that ‐ besides transport – lymphatic capillaries exert additional roles in immune modulation.
Collapse
Affiliation(s)
| | | | | | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Structure and Immune Function of Afferent Lymphatics and Their Mechanistic Contribution to Dendritic Cell and T Cell Trafficking. Cells 2021; 10:cells10051269. [PMID: 34065513 PMCID: PMC8161367 DOI: 10.3390/cells10051269] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/18/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Afferent lymphatic vessels (LVs) mediate the transport of antigen and leukocytes to draining lymph nodes (dLNs), thereby serving as immunologic communication highways between peripheral tissues and LNs. The main cell types migrating via this route are antigen-presenting dendritic cells (DCs) and antigen-experienced T cells. While DC migration is important for maintenance of tolerance and for induction of protective immunity, T cell migration through afferent LVs contributes to immune surveillance. In recent years, great progress has been made in elucidating the mechanisms of lymphatic migration. Specifically, time-lapse imaging has revealed that, upon entry into capillaries, both DCs and T cells are not simply flushed away with the lymph flow, but actively crawl and patrol and even interact with each other in this compartment. Detachment and passive transport to the dLN only takes place once the cells have reached the downstream, contracting collecting vessel segments. In this review, we describe how the anatomy of the lymphatic network supports leukocyte trafficking and provide updated knowledge regarding the cellular and molecular mechanisms responsible for lymphatic migration of DCs and T cells. In addition, we discuss the relevance of DC and T cell migration through afferent LVs and its presumed implications on immunity.
Collapse
|
6
|
Steele MM, Lund AW. Afferent Lymphatic Transport and Peripheral Tissue Immunity. THE JOURNAL OF IMMUNOLOGY 2021; 206:264-272. [PMID: 33397740 DOI: 10.4049/jimmunol.2001060] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/11/2020] [Indexed: 12/30/2022]
Abstract
Lymphatic vessels provide an anatomical framework for immune surveillance and adaptive immune responses. Although appreciated as the route for Ag and dendritic cell transport, peripheral lymphatic vessels are often not considered active players in immune surveillance. Lymphatic vessels, however, integrate contextual cues that directly regulate transport, including changes in intrinsic pumping and capillary remodeling, and express a dynamic repertoire of inflammatory chemokines and adhesion molecules that facilitates leukocyte egress out of inflamed tissue. These mechanisms together contribute to the course of peripheral tissue immunity. In this review, we focus on context-dependent mechanisms that regulate fluid and cellular transport out of peripheral nonlymphoid tissues to provide a framework for understanding the effects of afferent lymphatic transport on immune surveillance, peripheral tissue inflammation, and adaptive immunity.
Collapse
Affiliation(s)
- Maria M Steele
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY 10016
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY 10016; .,Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016; and.,Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
7
|
Abstract
BACKGROUND Chemokine (C-C motif) ligand 19 (CCL19) is a leukocyte chemoattractant that plays a crucial role in cell trafficking and leukocyte activation. Dysfunctional CD8+ T cells play a crucial role in persistent HBV infection. However, whether HBV can be cleared by CCL19-activated immunity remains unclear. METHODS We assessed the effects of CCL19 on the activation of PBMCs in patients with HBV infection. We also examined how CCL19 influences HBV clearance and modulates HBV-responsive T cells in a mouse model of chronic hepatitis B (CHB). In addition, C-C chemokine-receptor type 7 (CCR7) knockdown mice were used to elucidate the underlying mechanism of CCL19/CCR7 axis-induced immune activation. RESULTS From in vitro experiments, we found that CCL19 enhanced the frequencies of Ag-responsive IFN-γ+ CD8+ T cells from patients by approximately twofold, while CCR7 knockdown (LV-shCCR7) and LY294002 partially suppressed IFN-γ secretion. In mice, CCL19 overexpression led to rapid clearance of intrahepatic HBV likely through increased intrahepatic CD8+ T-cell proportion, decreased frequency of PD-1+ CD8+ T cells in blood and compromised suppression of hepatic APCs, with lymphocytes producing a significantly high level of Ag-responsive TNF-α and IFN-γ from CD8+ T cells. In both CCL19 over expressing and CCR7 knockdown (AAV-shCCR7) CHB mice, the frequency of CD8+ T-cell activation-induced cell death (AICD) increased, and a high level of Ag-responsive TNF-α and low levels of CD8+ regulatory T (Treg) cells were observed. CONCLUSIONS Findings in this study provide insights into how CCL19/CCR7 axis modulates the host immune system, which may promote the development of immunotherapeutic strategies for HBV treatment by overcoming T-cell tolerance.
Collapse
|
8
|
Yan Y, Chen R, Wang X, Hu K, Huang L, Lu M, Hu Q. CCL19 and CCR7 Expression, Signaling Pathways, and Adjuvant Functions in Viral Infection and Prevention. Front Cell Dev Biol 2019; 7:212. [PMID: 31632965 PMCID: PMC6781769 DOI: 10.3389/fcell.2019.00212] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/18/2019] [Indexed: 12/11/2022] Open
Abstract
Chemokine (C–C motif) ligand 19 (CCL19) is a critical regulator of the induction of T cell activation, immune tolerance, and inflammatory responses during continuous immune surveillance, homeostasis, and development. Migration of CC-chemokine receptor 7 (CCR7)-expressing cells to secondary lymphoid organs is a crucial step in the onset of adaptive immunity, which is initiated by a complex interaction between CCR7 and its cognate ligands. Recent advances in knowledge regarding the response of the CCL19-CCR7 axis to viral infections have elucidated the complex network of interplay among the invading virus, target cells and host immune responses. Viruses use various strategies to evade or delay the cytokine response, gaining additional time to replicate in the host. In this review, we summarize the impacts of CCL19 and CCR7 expression on the regulation of viral pathogenesis with an emphasis on the corresponding signaling pathways and adjuvant mechanisms. We present and discuss the expression, signaling adaptor proteins and effects of CCL19 and CCR7 as these molecules differentially impact different viral infections and viral life cycles in host homeostatic strategies. The underlying mechanisms discussed in this review may assist in the design of novel agents to modulate chemokine activity for viral prevention.
Collapse
Affiliation(s)
- Yan Yan
- Center of Clinical Laboratory, The Fifth People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, China.,The International Joint Research Laboratory for Infection and Immunity (China-Germany), Jiangnan University, Wuxi, China
| | - Renfang Chen
- The International Joint Research Laboratory for Infection and Immunity (China-Germany), Jiangnan University, Wuxi, China.,Hepatology Institute of Wuxi, The Fifth People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xu Wang
- Center of Clinical Laboratory, The Fifth People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Kai Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Lihua Huang
- The International Joint Research Laboratory for Infection and Immunity (China-Germany), Jiangnan University, Wuxi, China.,Hepatology Institute of Wuxi, The Fifth People's Hospital of Wuxi, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Mengji Lu
- The International Joint Research Laboratory for Infection and Immunity (China-Germany), Jiangnan University, Wuxi, China.,Institute of Virology, University Hospital of Essen, University Duisburg-Essen, Essen, Germany
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Institute for Infection and Immunity, St. George's, University of London, London, United Kingdom
| |
Collapse
|
9
|
Kang NJ, Han SC, Yoon SH, Sim JY, Maeng YH, Kang HK, Yoo ES. Cinnamomum camphora Leaves Alleviate Allergic Skin Inflammatory Responses In Vitro and In Vivo. Toxicol Res 2019; 35:279-285. [PMID: 31341557 PMCID: PMC6629446 DOI: 10.5487/tr.2019.35.3.279] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/24/2018] [Accepted: 01/16/2019] [Indexed: 11/23/2022] Open
Abstract
In this study, we investigated the therapeutic potential of Cinnamomum camphora leaves on allergic skin inflammation such as atopic dermatitis. We evaluated the effects of C. camphora leaves on human adult low-calcium high-temperature keratinocytes and atopic dermatitis mice. C. camphora leaves inhibited Macrophage-derived chemokine (an inflammatory chemokine) production in interferon-γ (10 ng/mL) stimulated Human adult low-calcium high-temperature keratinocytes in a dose dependent manner. C. camphora leaves suppressed the phosphorylation of janus kinase signal transducer and activator of transcription 1. C. camphora leaves also suppressed the phosphorylation of extracellular signal-regulated kinase 1/2, a central signaling molecule in the inflammation process. These results suggest that C. camphora leaves exhibits anti-inflammatory effect via the phosphorylation of signal transducer and activator of transcription 1 and extracellular signal-regulated kinase 1/2. To study the advanced effects of C. camphora leaves on atopic dermatitis, we induced experimental atopic dermatitis in mice by applying 2,4-dinitrochlorobenzene. The group treated with C. camphora leaves (100 mg/kg) showed remarkable improvement of atopic dermatitis symptoms: reduced serum immunoglobulin E levels, smaller lymph nodes with reduced thickness and length, decreased ear edema, and reduced levels of inflammatory cell infiltration in the ears. Interestingly, the effects of C. camphora leaves on atopic dermatitis symptoms were stronger than those of hydrocort cream, a positive control. Taken together, C. camphora leaves showed alleviating effects on the inflammatory chemokine production in vitro and atopic dermatitis symptoms in vivo. These results suggest that C. camphora leaves help in the treatment of allergic inflammation such as atopic dermatitis.
Collapse
Affiliation(s)
- Na-Jin Kang
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Sang-Chul Han
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Seok-Hyun Yoon
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Jae-Yeop Sim
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Young Hee Maeng
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Hee-Kyoung Kang
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Eun-Sook Yoo
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| |
Collapse
|
10
|
Han SC, Kang NJ, Yoon WJ, Kim S, Na MC, Koh YS, Hyun JW, Lee NH, Ko MH, Kang HK, Yoo ES. External Application of Apo-9'-fucoxanthinone, Isolated from Sargassum muticum, Suppresses Inflammatory Responses in a Mouse Model of Atopic Dermatitis. Toxicol Res 2016; 32:109-14. [PMID: 27123161 PMCID: PMC4843979 DOI: 10.5487/tr.2016.32.2.109] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/15/2015] [Accepted: 01/27/2016] [Indexed: 11/20/2022] Open
Abstract
Allergic skin inflammation such as atopic dermatitis is characterized by skin barrier dysfunction, edema, and infiltration with various inflammatory cells. The anti-inflammatory effects of Apo-9′-fucoxanthinone, isolated from Sargassum muticum, have been described in many diseases, but the mechanism by which it modulates the immune system is poorly understood. In this study, the ability of Apo-9′-fucoxanthinone to suppress allergic reactions was investigated using a mouse model of atopic dermatitis. The Apo-9′-fucoxanthinone-treated group showed significantly decreased immunoglobulin E in serum. Also, Apo-9′-fucoxanthinone treatment resulted in a smaller lymph node size with reduced the thickness and length compared to the induction group. In addition, Apo-9′-fucoxanthinone inhibited the expression of interleukin-4, interferon-gamma and tumor necrosis factor-alpha by phorbol 12-myristate 13-acetate and ionomycin-stimulated lymphocytes. These results suggest that Apo-9′-fucoxanthinone may be a useful therapeutic strategy for treating chronic inflammatory diseases.
Collapse
Affiliation(s)
- Sang-Chul Han
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Na-Jin Kang
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Weon-Jong Yoon
- Jeju Biodiversity Research Institute (JBRI), Jeju Technopark (JTP), Jeju, Korea
| | - Sejin Kim
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Min-Chull Na
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Young-Sang Koh
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Jin-Won Hyun
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Nam-Ho Lee
- Department of Chemistry, College of Natural Science, Jeju National University, Jeju, Korea
| | - Mi-Hee Ko
- Jeju Biodiversity Research Institute (JBRI), Jeju Technopark (JTP), Jeju, Korea
| | - Hee-Kyoung Kang
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Eun-Sook Yoo
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| |
Collapse
|
11
|
Pretorius A, Faber F, van Kleef M. Immune gene expression profiling of PBMC isolated from horses vaccinated with attenuated African horsesickness virus serotype 4. Immunobiology 2016; 221:236-44. [DOI: 10.1016/j.imbio.2015.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/01/2015] [Indexed: 12/24/2022]
|
12
|
The Salivary Gland Acts as a Sink for Tissue-Resident Memory CD8(+) T Cells, Facilitating Protection from Local Cytomegalovirus Infection. Cell Rep 2015; 13:1125-1136. [PMID: 26526997 DOI: 10.1016/j.celrep.2015.09.082] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 09/14/2015] [Accepted: 09/25/2015] [Indexed: 11/20/2022] Open
Abstract
Tissue-resident memory T cells (TRM) reside in barrier tissues and provide local immediate protective immunity. Here, we show that the salivary gland (SG) most-effectively induces CD8(+) and CD4(+) TRM cells against murine cytomegalovirus (MCMV), which persists in and spreads from this organ. TRM generation depended on local antigen for CD4(+), but not CD8(+), TRM cells, highlighting major differences in T cell subset-specific demands for TRM development. CMV-specific CD8(+) T cells fail to control virus replication upon primary infection in the SG due to CMV-induced MHC I downregulation in glandular epithelial cells. Using intraglandular infection, we challenge this notion and demonstrate that memory CD8(+) T cells confer immediate protection against locally introduced MCMV despite active viral immune evasion, owing to early viral tropism to cells that largely withstand MHC I downregulation. Thus, we unravel a yet-unappreciated role for memory CD8(+) T cells in protecting mucosal tissues against CMV infection.
Collapse
|
13
|
Kang NJ, Han SC, Kang HJ, Ko G, Yoon WJ, Kang HK, Yoo ES. Anti-Inflammatory Effect of 3-Bromo-4,5-Dihydroxybenzaldehyde, a Component of Polysiphonia morrowii, In Vivo and In Vitro. Toxicol Res 2015; 33:325-332. [PMID: 29071017 PMCID: PMC5654201 DOI: 10.5487/tr.2017.33.4.325] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/21/2017] [Accepted: 09/14/2017] [Indexed: 12/18/2022] Open
Abstract
3-Bromo-4,5-dihydroxybenzaldehyde (BDB) is a natural bromophenol compound that is most commonly isolated from red algae. The present study was designed to investigate the anti-inflammatory properties of BDB on atopic dermatitis (AD) in mice induced by 2,4-dinitrochlorobenzene (DNCB) and on lipopolysaccharide (LPS)-stimulated murine macrophages. BDB treatment (100 mg/kg) resulted in suppression of the development of AD symptoms compared with the control treatment (induction-only), as demonstrated by reduced immunoglobulin E levels in serum, smaller lymph nodes with reduced thickness and length, a decrease in ear edema, and reduced levels of inflammatory cell infiltration in the ears. In RAW 264.7 murine macrophages, BDB (12.5, 25, 50, and 100 μM) suppressed the production of interleukin-6, a proinflammatory cytokine, in a dose-dependent manner. BDB also had an inhibitory effect on the phosphorylation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and signal transducer and activator of transcription 1 (STAT1; Tyr 701), two major signaling molecules involved in cellular inflammation. Taken together, the results show that BDB treatment alleviates inflammatory responses in an atopic dermatitis mouse model and RAW 264.7 macrophages. These results suggest that BDB may be a useful therapeutic strategy for treating conditions involving allergic inflammation such as atopic dermatitis.
Collapse
Affiliation(s)
- Na-Jin Kang
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Sang-Chul Han
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Hyun-Jae Kang
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Geum Ko
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Weon-Jong Yoon
- Jeju Biodiversity Research Institute (JBRI), Jeju Technopark (JTP), Jeju, Korea
| | - Hee-Kyoung Kang
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| | - Eun-Sook Yoo
- Department of Medicine, School of Medicine, Jeju National University, Jeju, Korea
| |
Collapse
|
14
|
Thome JJC, Farber DL. Emerging concepts in tissue-resident T cells: lessons from humans. Trends Immunol 2015; 36:428-35. [PMID: 26072286 DOI: 10.1016/j.it.2015.05.003] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 05/14/2015] [Accepted: 05/14/2015] [Indexed: 01/14/2023]
Abstract
Intensified efforts to promote protective T cell-based immunity in vaccines and immunotherapies have created a compelling need to expand our understanding of human T cell function and maintenance beyond its characterization in peripheral blood. Mouse studies of T cell immunity show that, in response to infection, T cells migrate to diverse sites and persist as tissue-resident memory T cells (TRM), which mediate rapid in situ protection on antigen recall. Here we discuss new approaches to probe human T cell immunity, including novel sampling, that indicate a broad distribution and high frequency of human TRM in multiple sites. These newer findings further implicate anatomic compartmentalization as a generalized mechanism for long-term maintenance of human T cells throughout life.
Collapse
Affiliation(s)
- Joseph J C Thome
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
| | - Donna L Farber
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Medical Center, New York, NY 10032, USA; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
15
|
Bielecki B, Jatczak-Pawlik I, Wolinski P, Bednarek A, Glabinski A. Central Nervous System and Peripheral Expression of CCL19, CCL21 and Their Receptor CCR7 in Experimental Model of Multiple Sclerosis. Arch Immunol Ther Exp (Warsz) 2015; 63:367-76. [PMID: 25957582 PMCID: PMC4572056 DOI: 10.1007/s00005-015-0339-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 04/16/2015] [Indexed: 11/25/2022]
Abstract
It is well documented that inflammatory chemokines play a significant role in the development of multiple sclerosis (MS) and its model, experimental autoimmune encephalomyelitis (EAE). Recently, the involvement of homeostatic (or lymphoid) chemokines in the pathogenesis of autoimmune diseases has become an object of intensive study. In this work, quantitative analysis of CCL19, CCL21 and CCR7 expression in the central nervous system (CNS), as well as in inflammatory mononuclear cells isolated from several organs during the first attack, remission and the second attack of chronic-relapsing EAE (ChREAE), was performed. Using real-time PCR, RNAse Protection Assay and immunohistochemistry, the expression of both chemokines, as well as of their common receptor CCR7, was analyzed in the brain, spleen, lymph nodes and peripheral blood mononuclear cells. Increased expression of CCL19 and CCL21 was observed mostly in mononuclear inflammatory cells isolated from the CNS during active ChREAE. At the same time the expression of CCR7 in blood mononuclear leukocytes was reduced. This observation extends our current knowledge about the possible role of chemokines CCL19, CCL21 and their receptor CCR7 in the pathogenesis of ChREAE and, by extension, MS.
Collapse
Affiliation(s)
- Bartosz Bielecki
- Department of Neurology and Stroke, Medical University of Lodz, Lodz, Poland.
- U1195 Inserm and University Paris-Sud, 80 General Leclerc st., 94276, Le Kremlin-Bicetre, France.
| | | | - Pawel Wolinski
- Department of Propedeutics of Neurology, Medical University of Lodz, Lodz, Poland
| | - Andrzej Bednarek
- Department of Molecular Cancerogenesis, Medical University of Lodz, Lodz, Poland
| | - Andrzej Glabinski
- Department of Neurology and Stroke, Medical University of Lodz, Lodz, Poland
- Department of Propedeutics of Neurology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
16
|
Kang NJ, Han SC, Kang GJ, Koo DH, Koh YS, Hyun JW, Lee NH, Ko MH, Kang HK, Yoo ES. Diphlorethohydroxycarmalol inhibits interleukin-6 production by regulating NF-κB, STAT5 and SOCS1 in lipopolysaccharide-stimulated RAW264.7 cells. Mar Drugs 2015; 13:2141-57. [PMID: 25871292 PMCID: PMC4413204 DOI: 10.3390/md13042141] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 03/09/2015] [Accepted: 03/30/2015] [Indexed: 01/01/2023] Open
Abstract
Diphlorethohydroxycarmalol (DPHC) is a phlorotannin compound isolated from Ishige okamuarae, a brown alga. This study was conducted to investigate the anti-inflammatory effect and action mechanism of DPHC in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. We found that DPHC strongly reduces the production of interleukin 6 (IL-6), but not that of tumor necrosis factor-alpha (TNF-α) induced by LPS. DPHC (12.5 and 100 μM) suppressed the phosphorylation and the nuclear translocation of NF-kappaB (NF-κB), a central signaling molecule in the inflammation process induced by LPS. The suppressor of cytokine signaling 1 (SOCS1) is a negative feedback regulator of Janus kinase (Jak)-signal transducer and activator of transcription (STAT) signaling. In this study, DPHC inhibited STAT5 expression and upregulated that of SOCS1 at a concentration of 100 μM. Furthermore, N-tosyl-l-phenylalanine chloromethyl ketone (TPCK) (a specific NF-κB inhibitor) and JI (a specific Jak2 inhibitor) reduced the production of IL-6, but not that of tumor necrosis factor-alpha (TNF-α) in LPS-stimulated RAW 264.7 macrophages. These findings demonstrate that DPHC inhibits IL-6 production via the downregulation of NF-κB and Jak2-STAT5 pathway and upregulation of SOCS1.
Collapse
MESH Headings
- Administration, Cutaneous
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
- Anti-Inflammatory Agents, Non-Steroidal/isolation & purification
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Dermatitis, Atopic/immunology
- Dermatitis, Atopic/pathology
- Dermatitis, Atopic/prevention & control
- Dose-Response Relationship, Drug
- Female
- Gene Expression Regulation/drug effects
- Heterocyclic Compounds, 3-Ring/administration & dosage
- Heterocyclic Compounds, 3-Ring/isolation & purification
- Heterocyclic Compounds, 3-Ring/pharmacology
- Heterocyclic Compounds, 3-Ring/therapeutic use
- Interleukin-6/antagonists & inhibitors
- Interleukin-6/metabolism
- Lipopolysaccharides/antagonists & inhibitors
- Lipopolysaccharides/toxicity
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Mice
- Mice, Inbred BALB C
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/metabolism
- Pacific Ocean
- Phaeophyceae/chemistry
- Phaeophyceae/growth & development
- RAW 264.7 Cells
- Republic of Korea
- STAT5 Transcription Factor/antagonists & inhibitors
- STAT5 Transcription Factor/genetics
- STAT5 Transcription Factor/metabolism
- Skin/drug effects
- Skin/immunology
- Skin/pathology
- Specific Pathogen-Free Organisms
- Suppressor of Cytokine Signaling 1 Protein
- Suppressor of Cytokine Signaling Proteins/agonists
- Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors
- Suppressor of Cytokine Signaling Proteins/genetics
- Suppressor of Cytokine Signaling Proteins/metabolism
Collapse
Affiliation(s)
- Na-Jin Kang
- Department of Biomedicine & Drug Development, Jeju National University, Jeju 690-756, Korea.
| | - Sang-Chul Han
- Department of Medicine, Jeju National University School of Medicine, Jeju 690-756, Korea.
| | - Gyeoung-Jin Kang
- Department of Medicine, Jeju National University School of Medicine, Jeju 690-756, Korea.
| | - Dong-Hwan Koo
- Department of Biomedicine & Drug Development, Jeju National University, Jeju 690-756, Korea.
| | - Young-Sang Koh
- Department of Biomedicine & Drug Development, Jeju National University, Jeju 690-756, Korea.
- Department of Medicine, Jeju National University School of Medicine, Jeju 690-756, Korea.
| | - Jin-Won Hyun
- Department of Biomedicine & Drug Development, Jeju National University, Jeju 690-756, Korea.
- Department of Medicine, Jeju National University School of Medicine, Jeju 690-756, Korea.
| | - Nam-Ho Lee
- Department of Chemistry, School of Natural Science, Jeju National University, Jeju 690-756, Korea.
| | - Mi-Hee Ko
- Jeju Biodiversity Research Institute (JBRI), JejuTechnopark (JTP), Jeju 690-787, Korea.
| | - Hee-Kyoung Kang
- Department of Biomedicine & Drug Development, Jeju National University, Jeju 690-756, Korea.
- Department of Medicine, Jeju National University School of Medicine, Jeju 690-756, Korea.
| | - Eun-Sook Yoo
- Department of Biomedicine & Drug Development, Jeju National University, Jeju 690-756, Korea.
- Department of Medicine, Jeju National University School of Medicine, Jeju 690-756, Korea.
| |
Collapse
|
17
|
Han SC, Koo DH, Kang NJ, Yoon WJ, Kang GJ, Kang HK, Yoo ES. Docosahexaenoic Acid Alleviates Atopic Dermatitis by Generating Tregs and IL-10/TGF-β-Modified Macrophages via a TGF-β-Dependent Mechanism. J Invest Dermatol 2014; 135:1556-1564. [PMID: 25405323 DOI: 10.1038/jid.2014.488] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 10/19/2014] [Accepted: 10/27/2014] [Indexed: 11/09/2022]
Abstract
Regulatory T cells (Tregs) have key roles in the immune response by suppressing the differentiation and proliferation of various immune cells. The beneficial effects of docosahexaenoic acid (DHA) have been described for many diseases; however, the mechanism by which it modulates the immune system is poorly understood. Therefore, the aim of this study was to examine whether DHA suppresses allergic reactions and upregulates the generation of CD4(+)Foxp3(+) T cells. We also examined the effects of transfusing interleukin-10/transforming growth factor-β (TGF-β)-modified macrophages (M2 macrophages) treated with DHA into a mouse model of atopic dermatitis. Here, we show that administration of DHA upregulates the generation of TGF-β-dependent CD4(+) forkhead box protein 3 (Foxp3(+)) Tregs. DHA induced T-cell hypo-responsiveness and downregulated cytokines associated with T helper (Th)-1, Th2, and Th17 cells. The differentiation of Foxp3(+) Tregs into CD4(+) T cells was directly mediated by DHA-M2 macrophages, which deactivated effector macrophages and inhibited CD4(+) T-cell proliferation. DHA showed therapeutic effects in mice with experimental atopic dermatitis. These results show that DHA enhances the function of M2 macrophages and that the generation of Tregs effectively protects mice against an inflammatory immune disorder. Thus, DHA may be a useful therapeutic strategy for treating chronic inflammatory diseases.
Collapse
Affiliation(s)
- Sang-Chul Han
- Department of Pharmacology, School of Medicine, Jeju National University, Jeju, South Korea
| | - Dong-Hwan Koo
- Department of Biomedicine & Drug Development, Jeju National University, Jeju, South Korea
| | - Na-Jin Kang
- Department of Biomedicine & Drug Development, Jeju National University, Jeju, South Korea
| | - Weon-Jong Yoon
- Jeju Biodiversity Research Institute (JBRI), Jeju Technopark (JTP), Jeju, South Korea
| | - Gyeoung-Jin Kang
- Department of Pharmacology, College of Pharmacy, Dongguk University, Goyang, South Korea
| | - Hee-Kyoung Kang
- Department of Pharmacology, School of Medicine, Jeju National University, Jeju, South Korea; Department of Biomedicine & Drug Development, Jeju National University, Jeju, South Korea
| | - Eun-Sook Yoo
- Department of Pharmacology, School of Medicine, Jeju National University, Jeju, South Korea; Department of Biomedicine & Drug Development, Jeju National University, Jeju, South Korea.
| |
Collapse
|
18
|
Han SC, Kang GJ, Ko YJ, Kang HK, Moon SW, Ann YS, Yoo ES. External Application of Fermented Olive Flounder (Paralichthys olivaceus) Oil Alleviates Inflammatory Responses in 2,4-Dinitrochlorobenzeneinduced Atopic Dermatitis Mouse Model. Toxicol Res 2013; 28:159-64. [PMID: 24278605 PMCID: PMC3834415 DOI: 10.5487/tr.2012.28.3.159] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Revised: 08/23/2012] [Accepted: 08/24/2012] [Indexed: 11/23/2022] Open
Abstract
Allergic skin inflammation such as atopic dermatitis (AD) is characterized by edema and infiltration with various inflammatory cells such as mast cells, basophils, eosinophils and T cells. Thymic stromal lymphopoietin (TSLP) is produced mainly by epidermal keratinocytes, as well as dermal fibroblasts and mast cells in the skin lesions of AD. Omega-3 polyunsaturated fatty acids in fish oil can reduce inflammation in allergic patients. Fermentation has a tremendous capacity to transform chemical structures. The antiinflammatory effects of fish oil have been described in many diseases, but the beneficial effects by which fermented olive flounder oil (FOF) modulates the allergic response is poorly understood. In this study, we produced FOF and tested its ability to suppress the various allergic inflammatory responses. The ability of FOF to modulate the immune system was investigated using a mouse model of AD. The FOF-treated group showed significantly decreased immunoglobulin E (IgE) and histamine in serum. Also, the increased TSLP expression was significantly inhibited in the FOF group; the FOF-treated group was not appreciably different from the hydrocort cream treatment group. In addition, FOF treatment resulted in a smaller spleen size with reduced the thickness and length compared to the induction group. Splenocytes from mice treated with FOF produced significantly less IFN-γ, IL-4, T-box transcription factor (T-bet) and GATA binding protein 3 (GATA3) expression compared with the induction group. These results suggest that FOF may be effective in treating the allergic symptoms of AD. 5.
Collapse
Affiliation(s)
- Sang-Chul Han
- Department of Pharmacology, School of Medicine, Jeju National University, , Jeju 690-756, Korea
| | | | | | | | | | | | | |
Collapse
|
19
|
The developmental pathway for CD103(+)CD8+ tissue-resident memory T cells of skin. Nat Immunol 2013; 14:1294-301. [PMID: 24162776 DOI: 10.1038/ni.2744] [Citation(s) in RCA: 997] [Impact Index Per Article: 83.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 09/20/2013] [Indexed: 12/12/2022]
Abstract
Tissue-resident memory T cells (T(RM) cells) provide superior protection against infection in extralymphoid tissues. Here we found that CD103(+)CD8(+) T(RM) cells developed in the skin from epithelium-infiltrating precursor cells that lacked expression of the effector-cell marker KLRG1. A combination of entry into the epithelium plus local signaling by interleukin 15 (IL-15) and transforming growth factor-β (TGF-β) was required for the formation of these long-lived memory cells. Notably, differentiation into T(RM) cells resulted in the progressive acquisition of a unique transcriptional profile that differed from that of circulating memory cells and other types of T cells that permanently reside in skin epithelium. We provide a comprehensive molecular framework for the local differentiation of a distinct peripheral population of memory cells that forms a first-line immunological defense system in barrier tissues.
Collapse
|
20
|
Baaten BJG, Cooper AM, Swain SL, Bradley LM. Location, location, location: the impact of migratory heterogeneity on T cell function. Front Immunol 2013; 4:311. [PMID: 24115949 PMCID: PMC3792444 DOI: 10.3389/fimmu.2013.00311] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 09/16/2013] [Indexed: 01/13/2023] Open
Abstract
T cell migration is crucial for an effective adaptive immune response to invading pathogens. Naive and memory T cells encounter pathogen antigens, become activated, and differentiate into effector cells in secondary lymphoid tissues, and then migrate to the site(s) of infection where they exert effector activities that control and eliminate pathogens. To achieve activation, efficient effector function, and good memory formation, T cells must traffic between lymphoid and non-lymphoid tissues within the body. This complex process is facilitated by chemokine receptors, selectins, CD44, and integrins that mediate the interactions of T cells with the environment. The expression patterns of these migration receptors (MR) dictate the tissues into which the effector T cells migrate and enable them to occupy specific niches within the tissue. While MR have been considered primarily to facilitate cell movement, we highlight how the heterogeneity of signaling through these receptors influences the function and fate of T cells in situ. We explore what drives MR expression heterogeneity, how this affects migration, and how this impacts T cell effector function and memory formation.
Collapse
Affiliation(s)
- Bas J G Baaten
- Infectious and Inflammatory Diseases Center, Sanford-Burnham Medical Research Institute , La Jolla, CA , USA
| | | | | | | |
Collapse
|
21
|
Gebhardt T, Mackay LK. Local immunity by tissue-resident CD8(+) memory T cells. Front Immunol 2012; 3:340. [PMID: 23162555 PMCID: PMC3493987 DOI: 10.3389/fimmu.2012.00340] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 10/23/2012] [Indexed: 12/18/2022] Open
Abstract
Microbial infection primes a CD8+ cytotoxic T cell response that gives rise to a long-lived population of circulating memory cells able to provide protection against systemic reinfection. Despite this, effective CD8+ T cell surveillance of barrier tissues such as skin and mucosa typically wanes with time, resulting in limited T cell-mediated protection in these peripheral tissues. However, recent evidence suggests that a specialized subset of CD103+ memory T cells can permanently lodge and persist in peripheral tissues, and that these cells can compensate for the loss of peripheral immune surveillance by circulating memory T cells. Here, we review evolving concepts regarding the generation and long-term persistence of these tissue-resident memory T cells (TRM) in epithelial and neuronal tissues. We further discuss the role of TRM cells in local infection control and their contribution to localized immune phenomena, in both mice and humans.
Collapse
Affiliation(s)
- Thomas Gebhardt
- Department of Microbiology and Immunology, The University of Melbourne Melbourne, VIC, Australia
| | | |
Collapse
|
22
|
Peripheral tissue surveillance and residency by memory T cells. Trends Immunol 2012; 34:27-32. [PMID: 23036434 DOI: 10.1016/j.it.2012.08.008] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Revised: 08/21/2012] [Accepted: 08/21/2012] [Indexed: 11/24/2022]
Abstract
T cell immunity has long been described in terms of two circulating memory populations. Central memory T (T(CM)) cells migrate between the secondary lymphoid organs and are capable of mounting a recall proliferative response on pathogen re-encounter, whereas effector memory T (T(EM)) cells traffic between blood and extralymphoid compartments for effective peripheral immune surveillance. It is now clear that there exists a third category of memory cells that never returns to the circulation. These tissue-resident memory T (T(RM)) cells are phenotypically distinct from T(EM) cells, persist in elevated numbers in areas involved in prior infection and have been implicated in various immune phenomena, such as the control of persisting infections and immune disorders in fixed regions of the body.
Collapse
|
23
|
Fermented fish oil suppresses T helper 1/2 cell response in a mouse model of atopic dermatitis via generation of CD4+CD25+Foxp3+ T cells. BMC Immunol 2012; 13:44. [PMID: 22873180 PMCID: PMC3537649 DOI: 10.1186/1471-2172-13-44] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 07/26/2012] [Indexed: 11/21/2022] Open
Abstract
Background Allergic skin inflammation such as atopic dermatitis (AD), which is characterized by pruritus and inflammation, is regulated partly through the activity of regulatory T cells (Tregs). Tregs play key roles in the immune response by preventing or suppressing the differentiation, proliferation and function of various immune cells, including CD4+ T cells. Recent studies report that fermentation has a tremendous capacity to transform chemical structures or create new substances, and the omega-3 polyunsaturated fatty acids (n-3 PUFAs) in fish oil can reduce inflammation in allergic patients. The beneficial effects of natural fish oil (NFO) have been described in many diseases, but the mechanism by which fermented fish oil (FFO) modulates the immune system and the allergic response is poorly understood. In this study, we produced FFO and tested its ability to suppress the allergic inflammatory response and to activate CD4+CD25+Foxp3+ Tregs. Results The ability of FFO and NFO to modulate the immune system was investigated using a mouse model of AD. Administration of FFO or NFO in the drinking water alleviated the allergic inflammation in the skin, and FFO was more effective than NFO. FFO treatment did increase the expression of the immune-suppressive cytokines TGF-β and IL-10. In addition, ingestion of FFO increased Foxp3 expression and the number of CD4+CD25+Foxp3+ Tregs compared with NFO. Conclusions These results suggest that the anti-allergic effect of FFO is associated with enrichment of CD4+CD25+ Foxp3+ T cells at the inflamed sites and that FFO may be effective in treating the allergic symptoms of AD.
Collapse
|
24
|
Tissue exit: a novel control point in the accumulation of antigen-specific CD8 T cells in the influenza a virus-infected lung. J Virol 2012; 86:3436-45. [PMID: 22278253 DOI: 10.1128/jvi.07025-11] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Memory/effector T cells efficiently migrate into extralymphoid tissues and sites of infection, providing immunosurveillance and a first line of defense against invading pathogens. Even though it is a potential means to regulate the size, quality, and duration of a tissue infiltrate, T cell egress from infected tissues is poorly understood. Using a mouse model of influenza A virus infection, we found that CD8 effector T cells egressed from the infected lung in a CCR7-dependent manner. In contrast, following antigen recognition, effector CD8 T cell egress decreased and CCR7 function was reduced in vivo and in vitro, indicating that the exit of CD8 T cells from infected tissues is tightly regulated. Our data suggest that the regulation of T cell egress is a mechanism to retain antigen-specific effectors at the site of infection to promote viral clearance, while decreasing the numbers of bystander T cells and preventing overt inflammation.
Collapse
|
25
|
Accumulation and local proliferation of antigen-specific CD4+ T cells in antigen-bearing tissue. Immunol Cell Biol 2010; 89:566-72. [PMID: 21060322 DOI: 10.1038/icb.2010.128] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although activation and subsequent expansion of naive CD4(+) T cells within lymph nodes is well characterized, the fate of T effector cells activated within peripheral tissues during secondary reactions is poorly defined. Therefore, we studied the recruitment, proliferation and egress of antigen-specific Th1 effector cells in comparison with nonspecific Th1 cells throughout a delayed-type hypersensitivity reaction (DTH). Although we observed a high turnover of Th1 effector cells with unspecific high-rate recruitment and CCR7-dependent egress from the inflamed tissue in the early, acute DTH phase, a strong, selective accumulation of antigen-specific T cells occurred during the chronic, late DTH phase. This was mainly based on local proliferation of CD4(+) effector cells within the DTH tissue and concomitant retention. Considering the strong CCR7-dependent Th cell egress found in this model, the reduced CCR7 expression on antigen-specific T cells isolated from late-phase DTH tissue most likely contributes to the retention of these cells within the tissue. Thus, peripheral tissues can support not only the proliferation of CD8(+) T cells, as recently shown, but also that of CD4(+) T effector cells, forming a pool of tissue-resident T cells.
Collapse
|
26
|
Abstract
The development of Th1 lymphocytes is essential for cell-mediated immunity and resistance against intracellular pathogens. However, if left unregulated, the same response can cause serious damage to host tissues and lead to mortality. A number of different paracrine regulatory mechanisms involving distinct myeloid and lymphoid subpopulations have been implicated in controlling excessive secretion of inflammatory cytokines by Th1 cells. Much of this work has focused on interleukin (IL)-10, a cytokine with broad anti-inflammatory properties, one of which is to counteract the function of Th1 lymphocytes. While studying the role of IL-10 in regulating immunopathology during infection with the intracellular parasite Toxoplasma gondii, we discovered that the host-protective IL-10 derives in an autocrine manner from conventional interferon-gamma (IFN-gamma)-producing T-bet(+) Foxp3(neg) Th1 cells. In the following review, we will discuss these findings that support the general concept that production of IL-10 is an important self-regulatory function of CD4(+) T lymphocytes.
Collapse
|
27
|
CCR7 deficiency leads to leukocyte activation and increased clearance in response to pulmonary Pseudomonas aeruginosa infection. Infect Immun 2010; 78:2099-107. [PMID: 20176793 DOI: 10.1128/iai.00962-09] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
CCR7 is a chemokine receptor expressed on the surfaces of T cells, B cells, and mature dendritic cells that controls cell migration in response to the cognate ligands CCL19 and CCL21. CCR7 is critical for the generation of an adaptive T cell response. However, the roles of CCR7 in the host defense against pulmonary infection and innate immunity are not well understood. We investigated the role of CCR7 in the host defense against acute pulmonary infection with Pseudomonas aeruginosa. We intranasally infected C57BL/6 mice with P. aeruginosa and characterized the expression of CCR7 ligands and the surface expression of CCR7 on pulmonary leukocytes. In response to infection, expression of CCL19 and expression of CCL21 were oppositely regulated, and myeloid dendritic cells upregulated CCR7 expression. We further examined the effects of CCR7 deficiency on the inflammatory response to P. aeruginosa infection. We infected Ccr7(-/-) and wild-type mice with P. aeruginosa and characterized the accumulation of pulmonary leukocytes, production of proinflammatory mediators, neutrophil activation, and bacterial clearance. CCR7 deficiency led to an accumulation of myeloid dendritic cells and T cells in the lung in response to infection. CCR7 deficiency resulted in higher expression of CD80 and CD86 on dendritic cells; increased production of interleukin-12/23p40 (IL-12/23p40), gamma interferon (IFN-gamma), and IL-1 alpha; increased neutrophil respiratory burst; and, ultimately, increased clearance of acute P. aeruginosa infection. In conclusion, our results suggest that CCR7 deficiency results in a heightened proinflammatory environment in response to acute pulmonary P. aeruginosa infection and contributes to more efficient clearance.
Collapse
|
28
|
Flórido M, McColl SR, Appelberg R. Delayed recruitment of lymphocytes into the lungs of CD30-deficient mice during aerogenic Mycobacterium avium infections. Immunobiology 2009; 214:643-52. [PMID: 19250702 DOI: 10.1016/j.imbio.2008.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 12/05/2008] [Accepted: 12/08/2008] [Indexed: 11/25/2022]
Abstract
CD30 is a member of the tumor necrosis factor-receptor superfamily, a group of receptors known to act as accessory molecules in the development of the immune response. Control and CD30-deficient mice were aerogenically infected with Mycobacterium avium. Although the mycobacterial loads in the lungs were similar in both strains of mice, CD30-deficient animals exhibited delayed structuring of pulmonary granulomas and reduced recruitment of lymphocytes throughout a 240 days period of infection. Discrete alterations in the chemokine network were detected in the CD30-deficient animals although they showed no clear relation to the deficient inflammatory response. Thus CD30/CD153 interactions are involved in lung immune-mediated inflammation.
Collapse
Affiliation(s)
- Manuela Flórido
- Laboratory of Microbiology and Immunology of Infection, Instituto de Biologia Molecular e Celular (IBMC), University of Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal
| | | | | |
Collapse
|
29
|
Verhoeven D, Teijaro JR, Farber DL. Heterogeneous memory T cells in antiviral immunity and immunopathology. Viral Immunol 2008; 21:99-113. [PMID: 18476772 DOI: 10.1089/vim.2008.0002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Memory T cells are generated following an initial viral infection, and have the potential for mediating robust protective immunity to viral re-challenge due to their rapid and enhanced functional responses. In recent years, it has become clear that the memory T cell response to most viruses is remarkably diverse in phenotype, function, and tissue distribution, and can undergo dynamic changes during its long-term maintenance in vivo. However, the role of this variegation and compartmentalizationof memory T cells in protective immunity to viruses remains unclear. In this review,we discuss the diverse features of memory T cells that can delineate different subsets, the characteristics of memory T cells thus far identified to promote protective immune responses, and how the heterogeneous nature of memory T cells may also promote immunopathology during antiviral responses. We propose that given the profound heterogeneity of memory T cells, regulation of memory T cells during secondary responses could focus the response to participation of specific subsets,and/or inhibit memory T-cell subsets and functions that can lead to immunopathology.
Collapse
Affiliation(s)
- David Verhoeven
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | |
Collapse
|
30
|
Stubbe M, Vanderheyde N, Pircher H, Goldman M, Marchant A. Characterization of a subset of antigen-specific human central memory CD4+ T lymphocytes producing effector cytokines. Eur J Immunol 2008; 38:273-82. [PMID: 18081039 DOI: 10.1002/eji.200737611] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CCR7(+ )central memory (T(CM)) CD4(+) T cells play a central role in long-term immunological memory. Recent reports indicate that a proportion of CD4(+) T(CM) is able to produce effector cytokines. The phenotype and the role of this subset remain unknown. We characterized cytokine-producing human CD4(+) T(CM) specific for cleared protein and persistent viral Ag. Our results demonstrate that the type of Ag stimulation is a major determinant of CD4(+) T(CM) differentiation. CMV-specific T(CM) were significantly more differentiated than protein Ag-specific T(CM) and included higher proportions of IFN-gamma-producing cells. The expression of killer cell lectin-like receptor G1 (KLRG1) by protein Ag- and CMV-specific T(CM) was associated with increased production of effector cytokines. KLRG1(+) T(CM) expressed high levels of CD127, suggesting that they can survive long term under the influence of IL-7. The induction of KLRG1(+) T(CM) may therefore represent an important target of vaccination against pathogens controlled by cellular immune responses.
Collapse
Affiliation(s)
- Muriel Stubbe
- Institute for Medical Immunology, Université Libre de Bruxelles, Charleroi, Belgium
| | | | | | | | | |
Collapse
|
31
|
Bielecki B, Mazurek A, Wolinski P, Glabinski A. Expression of chemokine receptors CCR7 and CCR8 in the CNS during ChREAE. Scand J Immunol 2007; 66:383-92. [PMID: 17850582 DOI: 10.1111/j.1365-3083.2007.01954.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Chemokines and their receptors are important players in organism homeostasis, development and immune response to inflammatory stimuli. It has been recently confirmed that they are also involved in the development of several autoimmune diseases. In this study, we analysed the expression of two recently identified CC chemokine receptors, CCR7 and CCR8, in the central nervous system (CNS) and in peripheral tissues during chronic relapsing experimental autoimmune encephalomyelitis (ChREAE) -- an animal model of the human demyelinating disease multiple sclerosis (MS). We observed upregulation of both chemokine receptors in the CNS during the first and second attacks of ChREAE, whereas disease remission was characterized by a lower expression of those receptors. An analysis of the kinetics of CCR7 and CCR8 expression in the CNS during the first attack of the disease showed a constant increase in the first few days after the onset of clinical signs. This expression correlated with the clinical severity of ChREAE. CCR7-positive mononuclear cells were detected mostly in perivascular inflammatory cuffs in the CNS. In peripheral tissues (the spleen and kidneys) expression of both receptors was not upregulated during active ChREAE. These findings suggest that CCR7 and CCR8 may play a significant role in the pathogenesis of EAE and probably MS.
Collapse
MESH Headings
- Animals
- Brain/immunology
- Central Nervous System/immunology
- Chronic Disease
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Female
- Image Processing, Computer-Assisted
- Immunohistochemistry
- Mice
- Multiple Sclerosis/genetics
- Multiple Sclerosis/immunology
- Receptors, CCR7
- Receptors, CCR8
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/genetics
- Receptors, Chemokine/immunology
- Spinal Cord/immunology
- Statistics, Nonparametric
- Up-Regulation
Collapse
Affiliation(s)
- B Bielecki
- Department of Experimental and Clinical Neurology, Medical University of Lodz, Lodz, Poland
| | | | | | | |
Collapse
|
32
|
Menning A, Höpken UE, Siegmund K, Lipp M, Hamann A, Huehn J. Distinctive role of CCR7 in migration and functional activity of naive- and effector/memory-like Treg subsets. Eur J Immunol 2007; 37:1575-83. [PMID: 17474155 DOI: 10.1002/eji.200737201] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Foxp3+CD25+CD4+ Treg play a fundamental role in the maintenance of self tolerance and the control of inflammatory reactions. Previous data demonstrated a division of labor between naive- and effector/memory-like Treg subsets, which is largely based on their lymph node-recirculating and inflammation-seeking migration behavior, respectively. The chemokine receptor CCR7 is expressed on both types of Treg subsets, albeit at different levels. Whether it fulfills similar or distinct roles in these subsets has not been studied so far. We here show that the recirculation of naive-like Treg through LN and, to some extent, the gut is dependent on CCR7. Lack of CCR7 not only prevents recirculation, but also almost completely abolishes the ability of naive-like Treg to control the priming phase of an immune response. In contrast, CCR7 deficiency in effector/memory-like Treg promotes their accumulation in inflamed sites, compatible with a role of CCR7 for exit from the tissue. Local Treg accumulation was accompanied by an enhanced suppression of inflammation. Together, our findings provide conclusive evidence that CCR7 expression on Treg differentially controls in vivo function of the naive- and effector/memory-like subsets.
Collapse
MESH Headings
- Adoptive Transfer/methods
- Animals
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/transplantation
- Cell Movement/genetics
- Cell Movement/physiology
- Cell Proliferation
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Hypersensitivity, Delayed/immunology
- Immune Tolerance/immunology
- Lymphocyte Activation
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, Transgenic
- Ovalbumin/immunology
- Peptide Fragments/immunology
- Receptors, CCR7
- Receptors, Chemokine/genetics
- Receptors, Chemokine/physiology
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/transplantation
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/transplantation
- Th1 Cells/immunology
- Th1 Cells/transplantation
- Vaccination
Collapse
Affiliation(s)
- Astrid Menning
- Experimentelle Rheumatologie, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
33
|
Stubbe M, Vanderheyde N, Goldman M, Marchant A. Antigen-specific central memory CD4+ T lymphocytes produce multiple cytokines and proliferate in vivo in humans. THE JOURNAL OF IMMUNOLOGY 2007; 177:8185-90. [PMID: 17114495 DOI: 10.4049/jimmunol.177.11.8185] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The function of Ag-specific central (T(CM)) and effector (T(EM)) memory CD4+ T lymphocytes remains poorly characterized in vivo in humans. Using CD154 as a marker of Ag-specific CD4+T cells, we studied the differentiation of memory subsets following anti-hepatitis B immunization. Hepatitis B surface Ag (HBs)-specific memory CD4+T cells were heterogeneous and included T(CM) (CCR7+CD27+) and T(EM) (CCR7(-)CD27(+/-)). HBs-specific T(CM) and T(EM) shared the capacity to produce multiple cytokines, including IL-2 and IFN-gamma. Several years postimmunization, approximately 10% of HBs-specific memory CD4+ T cells were in cycle (Ki67+) and the proliferating cells were CCR7+. These results suggest that the model of functional specialization of T(CM) and T(EM) cannot be applied to protein vaccine Ags and support the concept that T(CM) are capable of self-renewal and contribute to maintain the pool of memory cells.
Collapse
Affiliation(s)
- Muriel Stubbe
- Institute for Medical Immunology, Université Libre de Bruxelles, Rue Adrienne Bolland 8, 6041 Charleroi, Belgium
| | | | | | | |
Collapse
|
34
|
Rosseau S, Hocke A, Mollenkopf H, Schmeck B, Suttorp N, Kaufmann SHE, Zerrahn J. Comparative transcriptional profiling of the lung reveals shared and distinct features of Streptococcus pneumoniae and influenza A virus infection. Immunology 2006; 120:380-91. [PMID: 17163962 PMCID: PMC2265881 DOI: 10.1111/j.1365-2567.2006.02514.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pneumonia is the most common cause of death from infectious disease in the western hemisphere. Pathophysiological and protective processes are initiated by pattern recognition of microbial structures. To provide the molecular framework for a better understanding of processes relevant to host defence in pneumonia, we performed pulmonary transcriptome analysis in mice infected with the major bacterial and viral agents of community-acquired pneumonia, Streptococcus pneumoniae and influenza A virus. We detected differential expression of 1300 genes after infection with either pathogen. Of these, approximately 36% or 30% were specific for pneumococcal or influenza infection, respectively, yielding pathogen-specific as well as shared inflammatory transcriptional signatures. These results not only reveal a differential response on the cytokine and chemokine levels but also emphasize the important role of genes implicated in regulation and fine tuning of inflammation. As one, albeit unexpected, key feature of pneumococcal pneumonia we discovered down-regulation of B-cell responses, probably reflecting a pneumococcal virulence strategy. The pathophysiological consequences of influenza A virus infection were reflected by the emerging protective T-cell response and differential induction of genes involved in tissue regeneration and proliferation. These data provide new insights into pathogenesis of the most common forms of pneumonia, highlighting the value of transcriptional profiling for the elucidation of underlying mechanisms.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/immunology
- Female
- Gene Expression Profiling/methods
- Gene Expression Regulation, Bacterial/immunology
- Gene Expression Regulation, Viral/immunology
- Genetic Predisposition to Disease
- Immunity, Cellular/genetics
- Influenza A virus
- Lung/immunology
- Mice
- Mice, Inbred C57BL
- Oligonucleotide Array Sequence Analysis/methods
- Orthomyxoviridae Infections/genetics
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/pathology
- Pneumonia, Pneumococcal/genetics
- Pneumonia, Pneumococcal/immunology
- Pneumonia, Pneumococcal/pathology
- Pneumonia, Viral/genetics
- Pneumonia, Viral/immunology
- Pneumonia, Viral/pathology
- Polymerase Chain Reaction/methods
- T-Lymphocytes/immunology
- Transcription, Genetic
Collapse
Affiliation(s)
- Simone Rosseau
- Department of Internal Medicine, Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
35
|
Höpken UE, Wengner AM, Loddenkemper C, Stein H, Heimesaat MM, Rehm A, Lipp M. CCR7 deficiency causes ectopic lymphoid neogenesis and disturbed mucosal tissue integrity. Blood 2006; 109:886-95. [PMID: 17018859 DOI: 10.1182/blood-2006-03-013532] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Homeostatic trafficking of lymphocytes through extralymphoid tissues has been recently observed, and a potential role in immune surveillance and the establishment of peripheral tolerance are considered. However, the mechanisms regulating lymphocyte recirculation through peripheral tissues under noninflammatory conditions are not well understood. Here, we demonstrate that the chemokine receptor CCR7 controls not only lymphocyte trafficking to and within secondary lymphoid organs but also homeostatic migration of T and B lymphocytes through nonlymphoid tissues. Lack of CCR7 results in a massive accumulation of lymphocytes in epithelial tissues. In particular, the gastrointestinal mucosal tissue of CCR7-/- mice is highly permissive for the formation of lymphoid aggregates, which develop into ectopic follicular structures with major topologic characteristics of lymph nodes. Flow cytometry analysis of CD4+ T cells derived from ectopic follicles revealed that CD44hiCD62Llo effector memory T cells predominate in the gastric lymphoid aggregates. In aged mice, lack of CCR7 induced age-dependent histomorphologic changes in the stomach with profound cystic hyperplasia and an increased rate of mucosal proliferation resembling Menetrier disease. Thus, CCR7 regulates the cellular organization of visceral tissue by governing life-long recirculation of naive and memory lymphocytes under homeostatic conditions.
Collapse
Affiliation(s)
- Uta E Höpken
- Department of Tumor Genetics and Immunogenetics, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
36
|
Yang LT, Peng H, Zhu ZL, Li G, Huang ZT, Zhao ZX, Koup RA, Bailer RT, Wu CY. Long-lived effector/central memory T-cell responses to severe acute respiratory syndrome coronavirus (SARS-CoV) S antigen in recovered SARS patients. Clin Immunol 2006; 120:171-8. [PMID: 16781892 PMCID: PMC7106132 DOI: 10.1016/j.clim.2006.05.002] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Revised: 04/21/2006] [Accepted: 05/01/2006] [Indexed: 12/14/2022]
Abstract
The role of cell-mediated immunity in human SARS-CoV infection is still not well understood. In this study, we found that memory T-cell responses against the spike (S) protein were persistent for more than 1 year after SARS-CoV infection by detecting the production of IFN-γ using ELISA and ELISpot assays. Flow cytometric analysis showed that both CD4+ and CD8+ T cells were involved in cellular responses against SARS-CoV infection. Interestingly, most of SARS-CoV S-specific memory CD4+ T cells were central memory cells expressing CD45RO+ CCR7+ CD62L−. However, the majority of memory CD8+ T cells revealed effector memory phenotype expressing CD45RO− CCR7− CD62L−. Thus, our study provides the evidence that SARS-CoV infection in humans can induce cellular immune response that is persistent for a long period of time. These data may have an important implication in the possibility of designing effective vaccine against SARS-CoV infection, specifically in defining T-cell populations that are implicated in protective immunity.
Collapse
Affiliation(s)
- Li-Tao Yang
- Department of Immunology, Zhongshan Medical School, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Hui Peng
- Department of Immunology, Zhongshan Medical School, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Zhao-Ling Zhu
- Department of Immunology, Zhongshan Medical School, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Gang Li
- Third affiliated hospital of the Sun Yat-sen University, Guangzhou 510630, China
| | - Zi-Tong Huang
- Second affiliated hospital of the Sun Yat-sen University, Guangzhou 510120, China
| | - Zhi-Xin Zhao
- Third affiliated hospital of the Sun Yat-sen University, Guangzhou 510630, China
| | | | | | - Chang-You Wu
- Department of Immunology, Zhongshan Medical School, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou 510080, China
- Corresponding author. Fax: +86 20 87331552.
| |
Collapse
|
37
|
Debes GF, Dahl ME, Mahiny AJ, Bonhagen K, Campbell DJ, Siegmund K, Erb KJ, Lewis DB, Kamradt T, Hamann A. Chemotactic responses of IL-4-, IL-10-, and IFN-gamma-producing CD4+ T cells depend on tissue origin and microbial stimulus. THE JOURNAL OF IMMUNOLOGY 2006; 176:557-66. [PMID: 16365450 DOI: 10.4049/jimmunol.176.1.557] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Th1- and Th2-polarized immune responses are crucial in the defense against pathogens but can also promote autoimmunity and allergy. The chemokine receptors CXCR3 and CCR4 have been implicated in differential trafficking of IFN-gamma- and IL-4-producing T cells, respectively, but also in tissue and inflammation-specific homing independent of cytokine responses. Here, we tested whether CD4+ T cells isolated from murine tissues under homeostatic or inflammatory conditions exhibit restricted patterns of chemotactic responses that correlate with their production of IFN-gamma, IL-4, or IL-10. In uninfected mice, IL-4-producing T cells preferentially migrated to the CCR4 ligand, CCL17, whereas IFN-gamma-expressing T cells as well as populations of IL-4+ or IL-10+ T cells migrated to the CXCR3 ligand, CXCL9. All cytokine-producing T cell subsets strongly migrated to the CXCR4 ligand, CXCL12. We assessed chemotaxis of T cells isolated from mice infected with influenza A virus or the nematode Nippostrongylus brasiliensis, which induce a strong Th1 or Th2 response in the lung, respectively. Unexpectedly, the chemotactic responses of IL-4+ T cells and T cells expressing the immunosuppressive cytokine IL-10 were influenced not only by the strongly Th1- or Th2-polarized environments but also by their anatomical localization, i.e., lung or spleen. In contrast, IFN-gamma+ T cells exhibited robust chemotaxis toward CXCL9 and had the most consistent migration pattern in both infection models. The results support a model in which the trafficking responses of many effector and regulatory T cells are regulated as a function of the infectious and tissue environments.
Collapse
Affiliation(s)
- Gudrun F Debes
- Experimental Rheumatology, Medical Clinic, Charité University Medicine Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Chemokines are a large family of small, generally secreted polypeptides which guide lymphocyte movement throughout the body by controlling integrin avidity and inducing migration. Here, we look at recent, exciting findings on chemokine function throughout lymphocyte development and co-ordinated T and B cell migration during immune responses. Finally, we will review data on the regional control of immunity by tissue-specific chemokine receptors on effector/memory lymphocytes.
Collapse
Affiliation(s)
- Jens V Stein
- Theodor Kocher Institute, University of Bern, Freiestr. 1, 3012 Bern, Switzerland.
| | | |
Collapse
|
39
|
Debes GF, Arnold CN, Young AJ, Krautwald S, Lipp M, Hay JB, Butcher EC. Chemokine receptor CCR7 required for T lymphocyte exit from peripheral tissues. Nat Immunol 2005; 6:889-94. [PMID: 16116468 PMCID: PMC2144916 DOI: 10.1038/ni1238] [Citation(s) in RCA: 385] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2005] [Accepted: 07/06/2005] [Indexed: 01/23/2023]
Abstract
Lymphocytes travel throughout the body to carry out immune surveillance and participate in inflammatory reactions. Their path takes them from blood through tissues into lymph and back to blood. Molecules that control lymphocyte recruitment into extralymphoid tissues are well characterized, but exit is assumed to be random. Here, we showed that lymphocyte emigration from the skin was regulated and was sensitive to pertussis toxin. CD4(+) lymphocytes emigrated more efficiently than CD8(+) or B lymphocytes. T lymphocytes in the afferent lymph expressed functional chemokine receptor CCR7, and CCR7 was required for T lymphocyte exit from the skin. The regulated expression of CCR7 by tissue T lymphocytes may control their exit, acting with recruitment mechanisms to regulate lymphocyte transit and accumulation during immune surveillance and inflammation.
Collapse
Affiliation(s)
- Gudrun F Debes
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, California 94305, USA.
| | | | | | | | | | | | | |
Collapse
|
40
|
Bromley SK, Thomas SY, Luster AD. Chemokine receptor CCR7 guides T cell exit from peripheral tissues and entry into afferent lymphatics. Nat Immunol 2005; 6:895-901. [PMID: 16116469 DOI: 10.1038/ni1240] [Citation(s) in RCA: 413] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2005] [Accepted: 07/08/2005] [Indexed: 11/08/2022]
Abstract
T cell circulation between peripheral tissues and the lymphoid compartment is critical for immunosurveillance and host defense. However, the factors that determine whether T cells remain in peripheral tissue or return to the circulation are undefined. Here we demonstrate that the chemokine receptor CCR7 is a critical signal that determines T cell exit from peripheral tissue. Both CCR7(-) and CCR7(+) effector T cells entered mouse asthmatic lung and while CCR7(-) T cells accumulated, CCR7(+) T cells continued to migrate into afferent lymph. Delivery of both CCR7(+) and CCR7(-) T cells directly into the airways showed that only CCR7(+) T cells exited the lung and entered draining lymph nodes. Our study establishes a molecular basis for T cell exit from peripheral tissues.
Collapse
Affiliation(s)
- Shannon K Bromley
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | |
Collapse
|
41
|
Abstract
Antigen-experienced T cells have been divided into CD62L+ CCR7+ central memory (TCM) and CD62L- CCR7- effector memory (TEM) cells. Here, we examined coexpression of CD62L and CCR7 in lymphocytic choriomeningitis virus-specific memory CD8 T cells from both lymphoid and nonlymphoid tissues. Three main points emerged: firstly, memory cells frequently expressed a mixed CD62L- CCR7+ phenotype that differed from the phenotypes of classical TEM and TCM cells; secondly, TCM cells were not restricted to lymphoid organs but were also present in significant numbers in nonlymphoid tissues; and thirdly, a major shift from a TCM to TEM phenotype was found in memory cells that had been stimulated repetitively with antigen.
Collapse
Affiliation(s)
- Heike Unsoeld
- Institute for Medical Microbiology and Hygiene, Department of Immunology, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|