1
|
Ryu S, Liu X, Guo T, Guo Z, Zhang J, Cao YQ. Peripheral CCL2-CCR2 signalling contributes to chronic headache-related sensitization. Brain 2023; 146:4274-4291. [PMID: 37284790 PMCID: PMC10545624 DOI: 10.1093/brain/awad191] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/19/2023] [Accepted: 05/01/2023] [Indexed: 06/08/2023] Open
Abstract
Migraine, especially chronic migraine, is highly debilitating and still lacks effective treatment. The persistent headache arises from activation and sensitization of primary afferent neurons in the trigeminovascular pathway, but the underlying mechanisms remain incompletely understood. Animal studies indicate that signalling through chemokine C-C motif ligand 2 (CCL2) and C-C motif chemokine receptor 2 (CCR2) mediates the development of chronic pain after tissue or nerve injury. Some migraine patients had elevated CCL2 levels in CSF or cranial periosteum. However, whether the CCL2-CCR2 signalling pathway contributes to chronic migraine is not clear. Here, we modelled chronic headache with repeated administration of nitroglycerin (NTG, a reliable migraine trigger in migraineurs) and found that both Ccl2 and Ccr2 mRNA were upregulated in dura and trigeminal ganglion (TG) tissues that are implicated in migraine pathophysiology. In Ccl2 and Ccr2 global knockout mice, repeated NTG administration did not evoke acute or persistent facial skin hypersensitivity as in wild-type mice. Intraperitoneal injection of CCL2 neutralizing antibodies inhibited chronic headache-related behaviours induced by repeated NTG administration and repetitive restraint stress, suggesting that the peripheral CCL2-CCR2 signalling mediates headache chronification. We found that CCL2 was mainly expressed in TG neurons and cells associated with dura blood vessels, whereas CCR2 was expressed in subsets of macrophages and T cells in TG and dura but not in TG neurons under both control and disease states. Deletion of Ccr2 gene in primary afferent neurons did not alter NTG-induced sensitization, but eliminating CCR2 expression in either T cells or myeloid cells abolished NTG-induced behaviours, indicating that both CCL2-CCR2 signalling in T cells and macrophages are required to establish chronic headache-related sensitization. At cellular level, repeated NTG administration increased the number of TG neurons that responded to calcitonin-gene-related peptide (CGRP) and pituitary adenylate cyclase activating polypeptide (PACAP) as well as the production of CGRP in wild-type but not Ccr2 global knockout mice. Lastly, co-administration of CCL2 and CGRP neutralizing antibodies was more effective in reversing NTG-induced behaviours than individual antibodies. Taken together, these results suggest that migraine triggers activate CCL2-CCR2 signalling in macrophages and T cells. This consequently enhances both CGRP and PACAP signalling in TG neurons, ultimately leading to persistent neuronal sensitization underlying chronic headache. Our work not only identifies the peripheral CCL2 and CCR2 as potential targets for chronic migraine therapy, but also provides proof-of-concept that inhibition of both peripheral CGRP and CCL2-CCR2 signalling is more effective than targeting either pathway alone.
Collapse
Affiliation(s)
- Sun Ryu
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, Campus Box MSC 8054-86-05, St. Louis, MO 63110, USA
| | - Xuemei Liu
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, Campus Box MSC 8054-86-05, St. Louis, MO 63110, USA
| | - Tingting Guo
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, Campus Box MSC 8054-86-05, St. Louis, MO 63110, USA
| | - Zhaohua Guo
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, Campus Box MSC 8054-86-05, St. Louis, MO 63110, USA
| | - Jintao Zhang
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, Campus Box MSC 8054-86-05, St. Louis, MO 63110, USA
| | - Yu-Qing Cao
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, Campus Box MSC 8054-86-05, St. Louis, MO 63110, USA
| |
Collapse
|
2
|
Parween F, Singh SP, Zhang HH, Kathuria N, Otaizo-Carrasquero FA, Shamsaddini A, Gardina PJ, Ganesan S, Kabat J, Lorenzi HA, Myers TG, Farber JM. Chemokine positioning determines mutually exclusive roles for their receptors in extravasation of pathogenic human T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.25.525561. [PMID: 36789428 PMCID: PMC9928044 DOI: 10.1101/2023.01.25.525561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Pro-inflammatory T cells co-express multiple chemokine receptors, but the distinct functions of individual receptors on these cells are largely unknown. Human Th17 cells uniformly express the chemokine receptor CCR6, and we discovered that the subgroup of CD4+CCR6+ cells that co-express CCR2 possess a pathogenic Th17 signature, can produce inflammatory cytokines independent of TCR activation, and are unusually efficient at transendothelial migration (TEM). The ligand for CCR6, CCL20, was capable of binding to activated endothelial cells (ECs) and inducing firm arrest of CCR6+CCR2+ cells under conditions of flow - but CCR6 could not mediate TEM. By contrast, CCL2 and other ligands for CCR2, despite being secreted from both luminal and basal sides of ECs, failed to bind to the EC surfaces - and CCR2 could not mediate arrest. Nonetheless, CCR2 was required for TEM. To understand if CCR2's inability to mediate arrest was due solely to an absence of EC-bound ligands, we generated a CCL2-CXCL9 chimeric chemokine that could bind to the EC surface. Although display of CCL2 on the ECs did indeed lead to CCR2-mediated arrest of CCR6+CCR2+ cells, activating CCR2 with surface-bound CCL2 blocked TEM. We conclude that mediating arrest and TEM are mutually exclusive activities of chemokine receptors and/or their ligands that depend, respectively, on chemokines that bind to the EC luminal surfaces versus non-binding chemokines that form transendothelial gradients under conditions of flow. Our findings provide fundamental insights into mechanisms of lymphocyte extravasation and may lead to novel strategies to block or enhance their migration into tissue.
Collapse
Affiliation(s)
- Farhat Parween
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Satya P. Singh
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Hongwei H Zhang
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Nausheen Kathuria
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Francisco A. Otaizo-Carrasquero
- Research Technologies Branch, Genomic Technologies, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Amirhossein Shamsaddini
- Research Technologies Branch, Genomic Technologies, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Paul J. Gardina
- Research Technologies Branch, Genomic Technologies, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Sundar Ganesan
- Research Technologies Branch, Biological Imaging, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Juraj Kabat
- Research Technologies Branch, Biological Imaging, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Hernan A. Lorenzi
- Bioinformatics and Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Timothy G. Myers
- Research Technologies Branch, Genomic Technologies, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| | - Joshua M. Farber
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda MD 20892, USA
| |
Collapse
|
3
|
Johnson CS, Cook LM. Osteoid cell-derived chemokines drive bone-metastatic prostate cancer. Front Oncol 2023; 13:1100585. [PMID: 37025604 PMCID: PMC10070788 DOI: 10.3389/fonc.2023.1100585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
One of the greatest challenges in improving prostate cancer (PCa) survival is in designing new therapies to effectively target bone metastases. PCa regulation of the bone environment has been well characterized; however, bone-targeted therapies have little impact on patient survival, demonstrating a need for understanding the complexities of the tumor-bone environment. Many factors contribute to creating a favorable microenvironment for prostate tumors in bone, including cell signaling proteins produced by osteoid cells. Specifically, there has been extensive evidence from both past and recent studies that emphasize the importance of chemokine signaling in promoting PCa progression in the bone environment. Chemokine-focused strategies present promising therapeutic options for treating bone metastasis. These signaling pathways are complex, with many being produced by (and exerting effects on) a plethora of different cell types, including stromal and tumor cells of the prostate tumor-bone microenvironment. This review highlights an underappreciated molecular family that should be interrogated for treatment of bone metastatic prostate cancer (BM-PCa).
Collapse
Affiliation(s)
- Catherine S. Johnson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, NE, United States
| | - Leah M. Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Leah M. Cook,
| |
Collapse
|
4
|
Packard TA, Schwarzer R, Herzig E, Rao D, Luo X, Egedal JH, Hsiao F, Widera M, Hultquist JF, Grimmett ZW, Messer RJ, Krogan NJ, Deeks SG, Roan NR, Dittmer U, Hasenkrug KJ, Greene WC. CCL2: a Chemokine Potentially Promoting Early Seeding of the Latent HIV Reservoir. mBio 2022; 13:e0189122. [PMID: 36073812 PMCID: PMC9600577 DOI: 10.1128/mbio.01891-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 08/16/2022] [Indexed: 11/20/2022] Open
Abstract
HIV infects long-lived CD4 memory T cells, establishing a latent viral reservoir that necessitates lifelong antiretroviral therapy (ART). How this reservoir is formed so quickly after infection remains unclear. We now show the innate inflammatory response to HIV infection results in CCL2 chemokine release, leading to recruitment of cells expressing the CCR2 receptor, including a subset of central memory CD4 T cells. Supporting a role for the CCL2/CCR2 axis in rapid reservoir formation, we find (i) treatment of humanized mice with anti-CCL2 antibodies during early HIV infection decreases reservoir seeding and preserves CCR2/5+ cells and (ii) CCR2/5+ cells from the blood of HIV-infected individuals on long-term ART contain significantly more integrated provirus than CCR2/5-negative memory or naive cells. Together, these studies support a model where the host's innate inflammatory response to HIV infection, including CCL2 production, leads to the recruitment of CCR2/5+ central memory CD4 T cells to zones of virus-associated inflammation, likely contributing to rapid formation of the latent HIV reservoir. IMPORTANCE There are currently over 35 million people living with HIV worldwide, and we still have no vaccine or scalable cure. One of the difficulties with HIV is its ability to rapidly establish a viral reservoir in lymphoid tissues that allows it to elude antivirals and the immune system. Thus, it is important to understand how HIV accomplishes this so we can develop preventive strategies. Our current results show that an early inflammatory response to HIV infection includes production of the chemokine CCL2, which recruits a unique subset of CCR2/5+ CD4+ T cells that become infected and form a significant reservoir for latent infection. Furthermore, we show that blockade of CCL2 in humanized mice significantly reduces persistent HIV infection. This information is relevant to the development of therapeutics to prevent and/or treat chronic HIV infections.
Collapse
Affiliation(s)
| | - Roland Schwarzer
- J. David Gladstone Institutes, San Francisco, California, USA
- Institute for Translational HIV Research, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Eytan Herzig
- J. David Gladstone Institutes, San Francisco, California, USA
| | - Deepashri Rao
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Xiaoyu Luo
- J. David Gladstone Institutes, San Francisco, California, USA
| | | | - Feng Hsiao
- J. David Gladstone Institutes, San Francisco, California, USA
| | - Marek Widera
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Judd F. Hultquist
- J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Biosciences Institute (QBI), University of California San Francisco, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, USA
| | | | - Ronald J. Messer
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Nevan J. Krogan
- J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Biosciences Institute (QBI), University of California San Francisco, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, USA
| | - Steven G. Deeks
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Nadia R. Roan
- J. David Gladstone Institutes, San Francisco, California, USA
- Department of Urology, University of California San Francisco, San Francisco, California, USA
| | - Ulf Dittmer
- Institute for Translational HIV Research, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kim J. Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Warner C. Greene
- J. David Gladstone Institutes, San Francisco, California, USA
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
5
|
Sex Differences in Coronary Artery Disease and Diabetes Revealed by scRNA-Seq and CITE-Seq of Human CD4+ T Cells. Int J Mol Sci 2022; 23:ijms23179875. [PMID: 36077273 PMCID: PMC9456056 DOI: 10.3390/ijms23179875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/20/2022] Open
Abstract
Despite the decades-old knowledge that males and people with diabetes mellitus (DM) are at increased risk for coronary artery disease (CAD), the reasons for this association are only partially understood. Among the immune cells involved, recent evidence supports a critical role of T cells as drivers and modifiers of CAD. CD4+ T cells are commonly found in atherosclerotic plaques. We aimed to understand the relationship of CAD with sex and DM by single-cell RNA (scRNA-Seq) and antibody sequencing (CITE-Seq) of CD4+ T cells. Peripheral blood mononuclear cells (PBMCs) of 61 men and women who underwent cardiac catheterization were interrogated by scRNA-Seq combined with 49 surface markers (CITE-Seq). CAD severity was quantified using Gensini scores, with scores above 30 considered CAD+ and below 6 considered CAD-. Four pairs of groups were matched for clinical and demographic parameters. To test how sex and DM changed cell proportions and gene expression, we compared matched groups of men and women, as well as diabetic and non-diabetic subjects. We analyzed 41,782 single CD4+ T cell transcriptomes for sex differences in 16 women and 45 men with and without coronary artery disease and with and without DM. We identified 16 clusters in CD4+ T cells. The proportion of cells in CD4+ effector memory cluster 8 (CD4T8, CCR2+ Em) was significantly decreased in CAD+, especially among DM+ participants. This same cluster, CD4T8, was significantly decreased in female participants, along with two other CD4+ T cell clusters. In CD4+ T cells, 31 genes showed significant and coordinated upregulation in both CAD and DM. The DM gene signature was partially additive to the CAD gene signature. We conclude that (1) CAD and DM are clearly reflected in PBMC transcriptomes, and (2) significant differences exist between women and men and (3) between subjects with DM and non-DM.
Collapse
|
6
|
Ben-Yehuda H, Arad M, Peralta Ramos JM, Sharon E, Castellani G, Ferrera S, Cahalon L, Colaiuta SP, Salame TM, Schwartz M. Key role of the CCR2-CCL2 axis in disease modification in a mouse model of tauopathy. Mol Neurodegener 2021; 16:39. [PMID: 34172073 PMCID: PMC8234631 DOI: 10.1186/s13024-021-00458-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 05/26/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND For decades, dementia has been characterized by accumulation of waste in the brain and low-grade inflammation. Over the years, emerging studies highlighted the involvement of the immune system in neurodegenerative disease emergence and severity. Numerous studies in animal models of amyloidosis demonstrated the beneficial role of monocyte-derived macrophages in mitigating the disease, though less is known regarding tauopathy. Boosting the immune system in animal models of both amyloidosis and tauopathy, resulted in improved cognitive performance and in a reduction of pathological manifestations. However, a full understanding of the chain of events that is involved, starting from the activation of the immune system, and leading to disease mitigation, remained elusive. Here, we hypothesized that the brain-immune communication pathway that is needed to be activated to combat tauopathy involves monocyte mobilization via the C-C chemokine receptor 2 (CCR2)/CCL2 axis, and additional immune cells, such as CD4+ T cells, including FOXP3+ regulatory CD4+ T cells. METHODS We used DM-hTAU transgenic mice, a mouse model of tauopathy, and applied an approach that boosts the immune system, via blocking the inhibitory Programmed cell death protein-1 (PD-1)/PD-L1 pathway, a manipulation previously shown to alleviate disease symptoms and pathology. An anti-CCR2 monoclonal antibody (αCCR2), was used to block the CCR2 axis in a protocol that partially eliminates monocytes from the circulation at the time of anti-PD-L1 antibody (αPD-L1) injection, and for the critical period of their recruitment into the brain following treatment. RESULTS Performance of DM-hTAU mice in short-term and working memory tasks, revealed that the beneficial effect of αPD-L1, assessed 1 month after a single injection, was abrogated following blockade of CCR2. This was accompanied by the loss of the beneficial effect on disease pathology, assessed by measurement of cortical aggregated human tau load using Homogeneous Time Resolved Fluorescence-based immunoassay, and by evaluation of hippocampal neuronal survival. Using both multiparametric flow cytometry, and Cytometry by Time Of Flight, we further demonstrated the accumulation of FOXP3+ regulatory CD4+ T cells in the brain, 12 days following the treatment, which was absent subsequent to CCR2 blockade. In addition, measurement of hippocampal levels of the T-cell chemoattractant, C-X-C motif chemokine ligand 12 (Cxcl12), and of inflammatory cytokines, revealed that αPD-L1 treatment reduced their expression, while blocking CCR2 reversed this effect. CONCLUSIONS The CCR2/CCL2 axis is required to modify pathology using PD-L1 blockade in a mouse model of tauopathy. This modification involves, in addition to monocytes, the accumulation of FOXP3+ regulatory CD4+ T cells in the brain, and the T-cell chemoattractant, Cxcl12.
Collapse
Affiliation(s)
- Hila Ben-Yehuda
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Arad
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Efrat Sharon
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Giulia Castellani
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Shir Ferrera
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Liora Cahalon
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Tomer-Meir Salame
- Flow Cytometry Unit, Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
7
|
Leal JM, Huang JY, Kohli K, Stoltzfus C, Lyons-Cohen MR, Olin BE, Gale M, Gerner MY. Innate cell microenvironments in lymph nodes shape the generation of T cell responses during type I inflammation. Sci Immunol 2021; 6:eabb9435. [PMID: 33579750 PMCID: PMC8274717 DOI: 10.1126/sciimmunol.abb9435] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/21/2020] [Accepted: 01/14/2021] [Indexed: 12/11/2022]
Abstract
Microanatomical organization of innate immune cells within lymph nodes (LNs) is critical for the generation of adaptive responses. In particular, steady-state LN-resident dendritic cells (Res cDCs) are strategically localized to intercept lymph-draining antigens. Whether myeloid cell organization changes during inflammation and how that might affect the generation of immune responses are unknown. Here, we report that during type I, but not type II, inflammation after adjuvant immunization or viral infection, antigen-presenting Res cDCs undergo CCR7-dependent intranodal repositioning from the LN periphery into the T cell zone (TZ) to elicit T cell priming. Concurrently, inflammatory monocytes infiltrate the LNs via local blood vessels, enter the TZ, and cooperate with Res cDCs by providing polarizing cytokines to optimize T cell effector differentiation. Monocyte infiltration is nonuniform across LNs, generating distinct microenvironments with varied local innate cell composition. These spatial microdomains are associated with divergent early T cell effector programming, indicating that innate microenvironments within LNs play a critical role in regulating the quality and heterogeneity of T cell responses. Together, our findings reveal that dynamic modulation of innate cell microenvironments during type I inflammation leads to optimized generation of adaptive immune responses to vaccines and infections.
Collapse
Affiliation(s)
- Joseph M Leal
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Jessica Y Huang
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Karan Kohli
- Department of Surgery, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Caleb Stoltzfus
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Miranda R Lyons-Cohen
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Brandy E Olin
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Michael Gale
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Michael Y Gerner
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA.
| |
Collapse
|
8
|
Uchida N, Mori K, Fujita-Nakata M, Nakanishi M, Sanada M, Nagayama S, Sugiyama H, Matsui M. Systemic cellular immunity and neuroinflammation during acute flare-up in multiple sclerosis and neuromyelitis optica spectrum disorder patients. J Neuroimmunol 2021; 353:577500. [PMID: 33592574 DOI: 10.1016/j.jneuroim.2021.577500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/23/2021] [Accepted: 01/24/2021] [Indexed: 11/24/2022]
Abstract
Twenty-seven treatment-naïve patients with relapsing-remitting multiple sclerosis (MS) and 13 with neuromyelitis optica spectrum disorder (NMOSD) were enrolled during a time of acute flare-up. Common cerebrospinal fluid (CSF) features were increased CD29- and/or CD45RO-positive helper T cells capable of propagating inflammation in the central nervous system (CNS). B cell activation in the CSF was unique to MS, while an increase in CD4+CD192 (CCR2)+ cells in blood and breakdown of the blood-brain barrier (BBB) characterized NMOSD. Intravenous corticosteroid therapy suppressed neuroinflammation via modulation of cellular immunity in MS, as opposed to restoration of the BBB in NMOSD.
Collapse
Affiliation(s)
- Nobuaki Uchida
- Department of Neurology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Kentaro Mori
- Department of Neurology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Michiyo Fujita-Nakata
- Department of Neurology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Megumi Nakanishi
- Department of Neurology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Mitsuru Sanada
- Department of Neurology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Shigemi Nagayama
- Department of Neurology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan
| | - Hiroshi Sugiyama
- Department of Neurology, National Hospital Organization Utano National Hospital, 8 Narutaki-Ondoyama-cho, Ukyo-ku, Kyoto 616-8255, Japan
| | - Makoto Matsui
- Department of Neurology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku-gun, Ishikawa 920-0293, Japan.
| |
Collapse
|
9
|
Nelson MH, Knochelmann HM, Bailey SR, Huff LW, Bowers JS, Majchrzak-Kuligowska K, Wyatt MM, Rubinstein MP, Mehrotra S, Nishimura MI, Armeson KE, Giresi PG, Zilliox MJ, Broxmeyer HE, Paulos CM. Identification of human CD4 + T cell populations with distinct antitumor activity. SCIENCE ADVANCES 2020; 6:eaba7443. [PMID: 32937437 PMCID: PMC7458458 DOI: 10.1126/sciadv.aba7443] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 05/18/2020] [Indexed: 05/26/2023]
Abstract
How naturally arising human CD4+ T helper subsets affect cancer immunotherapy is unclear. We reported that human CD4+CD26high T cells elicit potent immunity against solid tumors. As CD26high T cells are often categorized as TH17 cells for their IL-17 production and high CD26 expression, we posited these populations would have similar molecular properties. Here, we reveal that CD26high T cells are epigenetically and transcriptionally distinct from TH17 cells. Of clinical importance, CD26high and TH17 cells engineered with a chimeric antigen receptor (CAR) regressed large human tumors to a greater extent than enriched TH1 or TH2 cells. Only human CD26high T cells mediated curative responses, even when redirected with a suboptimal CAR and without aid by CD8+ CAR T cells. CD26high T cells cosecreted effector cytokines, produced cytotoxic molecules, and persisted long term. Collectively, our work underscores the promise of CD4+ T cell populations to improve durability of solid tumor therapies.
Collapse
Affiliation(s)
- Michelle H Nelson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, SC, USA
| | - Hannah M Knochelmann
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, SC, USA
| | - Stefanie R Bailey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, SC, USA
| | - Logan W Huff
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, SC, USA
| | - Jacob S Bowers
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, SC, USA
| | - Kinga Majchrzak-Kuligowska
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, SC, USA
| | - Megan M Wyatt
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, SC, USA
| | - Mark P Rubinstein
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Shikhar Mehrotra
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Michael I Nishimura
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Kent E Armeson
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | | | - Michael J Zilliox
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, SC, USA
| |
Collapse
|
10
|
Eckert N, Permanyer M, Yu K, Werth K, Förster R. Chemokines and other mediators in the development and functional organization of lymph nodes. Immunol Rev 2020; 289:62-83. [PMID: 30977201 DOI: 10.1111/imr.12746] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 01/22/2019] [Indexed: 12/28/2022]
Abstract
Secondary lymphoid organs like lymph nodes (LNs) are the main inductive sites for adaptive immune responses. Lymphocytes are constantly entering LNs, scanning the environment for their cognate antigen and get replenished by incoming cells after a certain period of time. As only a minor percentage of lymphocytes recognizes cognate antigen, this mechanism of permanent recirculation ensures fast and effective immune responses when necessary. Thus, homing, positioning, and activation as well as egress require precise regulation within LNs. In this review we discuss the mediators, including chemokines, cytokines, growth factors, and others that are involved in the formation of the LN anlage and subsequent functional organization of LNs. We highlight very recent findings in the fields of LN development, steady-state migration in LNs, and the intranodal processes during an adaptive immune response.
Collapse
Affiliation(s)
- Nadine Eckert
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Marc Permanyer
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Kai Yu
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Kathrin Werth
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
11
|
Murphy PM. Chemokines and Chemokine Receptors. Clin Immunol 2019. [DOI: 10.1016/b978-0-7020-6896-6.00010-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
12
|
Ajina A, Maher J. Strategies to Address Chimeric Antigen Receptor Tonic Signaling. Mol Cancer Ther 2018; 17:1795-1815. [PMID: 30181329 PMCID: PMC6130819 DOI: 10.1158/1535-7163.mct-17-1097] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/19/2018] [Accepted: 06/18/2018] [Indexed: 12/18/2022]
Abstract
Adoptive cell transfer using chimeric antigen receptors (CAR) has emerged as one of the most promising new therapeutic modalities for patients with relapsed or refractory B-cell malignancies. Thus far, results in patients with advanced solid tumors have proven disappointing. Constitutive tonic signaling in the absence of ligand is an increasingly recognized complication when deploying these synthetic fusion receptors and can be a cause of poor antitumor efficacy, impaired survival, and reduced persistence in vivo In parallel, ligand-dependent tonic signaling can mediate toxicity and promote T-cell anergy, exhaustion, and activation-induced cell death. Here, we review the mechanisms underpinning CAR tonic signaling and highlight the wide variety of effects that can emerge after making subtle structural changes or altering the methodology of CAR transduction. We highlight strategies to prevent unconstrained tonic signaling and address its deleterious consequences. We also frame this phenomenon in the context of endogenous TCR tonic signaling, which has been shown to regulate peripheral tolerance, facilitate the targeting of foreign antigens, and suggest opportunities to coopt ligand-dependent CAR tonic signaling to facilitate in vivo persistence and efficacy. Mol Cancer Ther; 17(9); 1795-815. ©2018 AACR.
Collapse
MESH Headings
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Adam Ajina
- CAR Mechanics Group, King's College London, London, United Kingdom.
- School of Cancer and Pharmaceutical Studies, Guy's Hospital, London, United Kingdom
| | - John Maher
- CAR Mechanics Group, King's College London, London, United Kingdom
- School of Cancer and Pharmaceutical Studies, Guy's Hospital, London, United Kingdom
- Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, United Kingdom
- Department of Immunology, Eastbourne Hospital, East Sussex, United Kingdom
| |
Collapse
|
13
|
Okutani M, Tsukahara T, Kato Y, Fukuta K, Inoue R. Gene expression profiles of CD4/CD8 double-positive T cells in porcine peripheral blood. Anim Sci J 2018; 89:979-987. [PMID: 29740910 DOI: 10.1111/asj.13021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/27/2017] [Indexed: 12/01/2022]
Abstract
A characteristic subset of T cells, known as double positive T cells (DPTC) and expressing both cluster of differentiation 4 (CD4) and CD8, is observed in porcine peripheral blood. Previous studies suggested that DPTC might be memory cells. However, detailed phenotypes and functions of DPTC are yet to be fully elucidated and thus, the relatedness of DPTC with memory phenotypes remains unclear. In this study, DPTC gene expression profiles in peripheral blood were analyzed by DNA microarray in Experiment 1 and compared with those of CD4 single positive T cells (4SPTC) and CD8 single positive T cells (8SPTC). Expressions of IFNG, CCL5, NCK2, CCR2 and ITGB1 were higher than that of 4SPTC and 8SPTC. In contrast, expressions of CCR7 and SELL were lower than that of 4SPTC and 8SPTC. These results suggested that DPTC were either effector T cells or effector memory T cells (TEM ). Next, to determine whether DPTC were effector T cells or TEM , differences in the response of DPTC and 8SPTC against immunized/primed antigens were compared (Experiment 2). While DPTC showed quick elevation of IL2 and CD25 gene expressions against in vitro stimulation of primed/immunized antigens, 8SPTC did not. These results suggest that at least some DPTC likely belong to TEM .
Collapse
Affiliation(s)
- Mie Okutani
- Laboratory of Animal Science, Department of Agricultural and Life Sciences, Kyoto Prefectural University, Sakyo, Kyoto, Japan
| | | | - Yoshihiro Kato
- Technical Center, Toyohashi Feed Mills, Shinshiro, Aichi, Japan
| | - Kikuto Fukuta
- Technical Center, Toyohashi Feed Mills, Shinshiro, Aichi, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Agricultural and Life Sciences, Kyoto Prefectural University, Sakyo, Kyoto, Japan
| |
Collapse
|
14
|
Lee CH, Zhang HH, Singh SP, Koo L, Kabat J, Tsang H, Singh TP, Farber JM. C/EBPδ drives interactions between human MAIT cells and endothelial cells that are important for extravasation. eLife 2018; 7:32532. [PMID: 29469805 PMCID: PMC5869018 DOI: 10.7554/elife.32532] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/21/2018] [Indexed: 12/14/2022] Open
Abstract
Many mediators and regulators of extravasation by bona fide human memory-phenotype T cells remain undefined. Mucosal-associated invariant T (MAIT) cells are innate-like, antibacterial cells that we found excelled at crossing inflamed endothelium. They displayed abundant selectin ligands, with high expression of FUT7 and ST3GAL4, and expressed CCR6, CCR5, and CCR2, which played non-redundant roles in trafficking on activated endothelial cells. MAIT cells selectively expressed CCAAT/enhancer-binding protein delta (C/EBPδ). Knockdown of C/EBPδ diminished expression of FUT7, ST3GAL4 and CCR6, decreasing MAIT cell rolling and arrest, and consequently the cells' ability to cross an endothelial monolayer in vitro and extravasate in mice. Nonetheless, knockdown of C/EBPδ did not affect CCR2, which was important for the step of transendothelial migration. Thus, MAIT cells demonstrate a program for extravasastion that includes, in part, C/EBPδ and C/EBPδ-regulated genes, and that could be used to enhance, or targeted to inhibit T cell recruitment into inflamed tissue.
Collapse
Affiliation(s)
- Chang Hoon Lee
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| | - Hongwei H Zhang
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| | - Satya P Singh
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| | - Lily Koo
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| | - Juraj Kabat
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| | - Hsinyi Tsang
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| | - Tej Pratap Singh
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| | - Joshua M Farber
- Inflammation Biology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, United States
| |
Collapse
|
15
|
Braud VM, Biton J, Becht E, Knockaert S, Mansuet-Lupo A, Cosson E, Damotte D, Alifano M, Validire P, Anjuère F, Cremer I, Girard N, Gossot D, Seguin-Givelet A, Dieu-Nosjean MC, Germain C. Expression of LLT1 and its receptor CD161 in lung cancer is associated with better clinical outcome. Oncoimmunology 2018; 7:e1423184. [PMID: 29721382 PMCID: PMC5927544 DOI: 10.1080/2162402x.2017.1423184] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/21/2017] [Accepted: 12/22/2017] [Indexed: 12/18/2022] Open
Abstract
Co-stimulatory and inhibitory receptors expressed by immune cells in the tumor microenvironment modulate the immune response and cancer progression. Their expression and regulation are still not fully characterized and a better understanding of these mechanisms is needed to improve current immunotherapies. Our previous work has identified a novel ligand/receptor pair, LLT1/CD161, that modulates immune responses. Here, we extensively characterize its expression in non-small cell lung cancer (NSCLC). We show that LLT1 expression is restricted to germinal center (GC) B cells within tertiary lymphoid structures (TLS), representing a new hallmark of the presence of active TLS in the tumor microenvironment. CD161-expressing immune cells are found at the vicinity of these structures, with a global enrichment of NSCLC tumors in CD161+ CD4+ and CD8+ T cells as compared to normal distant lung and peripheral blood. CD161+ CD4+ T cells are more activated and produce Th1-cytokines at a higher frequency than their matched CD161-negative counterparts. Interestingly, CD161+ CD4+ T cells highly express OX40 co-stimulatory receptor, less frequently 4-1BB, and display an activated but not completely exhausted PD-1-positive Tim-3-negative phenotype. Finally, a meta-analysis revealed a positive association of CLEC2D (coding for LLT1) and KLRB1 (coding for CD161) gene expression with favorable outcome in NSCLC, independently of the size of T and B cell infiltrates. These data are consistent with a positive impact of LLT1/CD161 on NSCLC patient survival, and make CD161-expressing CD4+ T cells ideal candidates for efficient anti-tumor recall responses.
Collapse
Affiliation(s)
- Véronique M. Braud
- Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| | - Jérôme Biton
- Laboratory “Immune Microenvironment and Tumors”, Department “Cancer, Immunology, Immunotherapy”, INSERM UMRS 1138, Cordeliers Research Center, Paris, France
- University Pierre and Marie Curie/Paris VI, Paris, France
- University Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| | - Etienne Becht
- Laboratory “Immune Microenvironment and Tumors”, Department “Cancer, Immunology, Immunotherapy”, INSERM UMRS 1138, Cordeliers Research Center, Paris, France
- University Pierre and Marie Curie/Paris VI, Paris, France
- University Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| | - Samantha Knockaert
- Laboratory “Immune Microenvironment and Tumors”, Department “Cancer, Immunology, Immunotherapy”, INSERM UMRS 1138, Cordeliers Research Center, Paris, France
- University Pierre and Marie Curie/Paris VI, Paris, France
- University Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| | - Audrey Mansuet-Lupo
- Laboratory “Immune Microenvironment and Tumors”, Department “Cancer, Immunology, Immunotherapy”, INSERM UMRS 1138, Cordeliers Research Center, Paris, France
- University Pierre and Marie Curie/Paris VI, Paris, France
- University Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Department of Pathology, Hôpitaux Universitaires Paris Centre, AP-HP, Paris, France
| | - Estelle Cosson
- Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| | - Diane Damotte
- Laboratory “Immune Microenvironment and Tumors”, Department “Cancer, Immunology, Immunotherapy”, INSERM UMRS 1138, Cordeliers Research Center, Paris, France
- University Pierre and Marie Curie/Paris VI, Paris, France
- University Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Department of Pathology, Hôpitaux Universitaires Paris Centre, AP-HP, Paris, France
| | - Marco Alifano
- University Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Department of Thoracic Surgery, Hôpitaux Universitaires Paris Centre, AP-HP, Paris, France
| | - Pierre Validire
- Laboratory “Immune Microenvironment and Tumors”, Department “Cancer, Immunology, Immunotherapy”, INSERM UMRS 1138, Cordeliers Research Center, Paris, France
- Department of Pathology, Institut Mutualiste Montsouris, Paris, France
| | - Fabienne Anjuère
- Université Côte d'Azur, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| | - Isabelle Cremer
- Laboratory “Immune Microenvironment and Tumors”, Department “Cancer, Immunology, Immunotherapy”, INSERM UMRS 1138, Cordeliers Research Center, Paris, France
- University Pierre and Marie Curie/Paris VI, Paris, France
- University Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| | - Nicolas Girard
- University of Lyon, University Lyon 1, Lyon, France
- Institut du Thorax Curie-Montsouris, Institut Curie, Paris, France
| | - Dominique Gossot
- Thoracic Department, Institut du Thorax Curie-Montsouris, Institut Mutualiste Montsouris, Paris, France
| | - Agathe Seguin-Givelet
- Thoracic Department, Institut du Thorax Curie-Montsouris, Institut Mutualiste Montsouris, Paris, France
- Paris 13 University, Sorbonne Paris Cité, Faculty of Medicine SMBH, Bobigny, France
| | - Marie-Caroline Dieu-Nosjean
- Laboratory “Immune Microenvironment and Tumors”, Department “Cancer, Immunology, Immunotherapy”, INSERM UMRS 1138, Cordeliers Research Center, Paris, France
- University Pierre and Marie Curie/Paris VI, Paris, France
- University Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| | - Claire Germain
- Laboratory “Immune Microenvironment and Tumors”, Department “Cancer, Immunology, Immunotherapy”, INSERM UMRS 1138, Cordeliers Research Center, Paris, France
- University Pierre and Marie Curie/Paris VI, Paris, France
- University Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
16
|
Löfroos AB, Kadivar M, Resic Lindehammer S, Marsal J. Colorectal cancer-infiltrating T lymphocytes display a distinct chemokine receptor expression profile. Eur J Med Res 2017; 22:40. [PMID: 29020986 PMCID: PMC5637168 DOI: 10.1186/s40001-017-0283-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 10/02/2017] [Indexed: 02/08/2023] Open
Abstract
Background T lymphocytes exert important homeostatic functions in the healthy intestinal mucosa, whereas in case of colorectal cancer (CRC), infiltration of T lymphocytes into the tumor is crucial for an effective anti-tumor immune response. In both situations, the recruitment mechanisms of T lymphocytes into the tissues are essential for the immunological functions deciding the outcome. The recruitment of T lymphocytes is largely dependent on their expression of various chemokine receptors. The aim of this study was to identify potential chemokine receptors involved in the recruitment of T lymphocytes to normal human colonic mucosa and to CRC tissue, respectively, by examining the expression of 16 different chemokine receptors on T lymphocytes isolated from these tissues. Methods Tissues were collected from patients undergoing bowel resection for CRC. Lymphocytes were isolated through enzymatic tissue degradation of CRC tissue and nearby located unaffected mucosa, respectively. The expression of a broad panel of chemokine receptors on the freshly isolated T lymphocytes was examined by flow cytometry. Results In the normal colonic mucosa, the frequencies of cells expressing CCR2, CCR4, CXCR3, and CXCR6 differed significantly between CD4+ and CD8+ T lymphocytes, suggesting that the molecular mechanisms mediating T lymphocyte recruitment to the gut differ between CD4+ and CD8+ T lymphocytes. In CRC, the frequencies of cells expressing CCR2 and CXCR5 were significantly lower in both the CD4+ and CD8+ T lymphocyte populations compared to unaffected colonic mucosa, and the frequency of CCR9+ cytotoxic T lymphocytes was significantly decreased in CRC tissue. Conclusions With regard to the normal gut mucosa, the results suggest that the molecular mechanisms mediating T lymphocyte recruitment differ between CD4+ and CD8+ T lymphocytes, which are important for understanding gut homeostasis. Importantly, T lymphocytes from CRC compared to normal colonic tissue displayed a distinct chemokine receptor expression profile, suggesting that mechanisms for recruitment of T lymphocytes to CRC tissue are skewed compared to normal colonic mucosa. Understanding these mechanisms could help in developing new strategies in cancer immunotherapy and to optimize already available alternatives such as immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Ann-Britt Löfroos
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Immunology Section, Lund University, Lund, Sweden
| | | | - Sabina Resic Lindehammer
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Immunology Section, Lund University, Lund, Sweden
| | - Jan Marsal
- Department of Clinical Sciences, Lund University, Lund, Sweden. .,Immunology Section, Lund University, Lund, Sweden. .,Department of Gastroenterology, Skane University Hospital, 22185, Lund, Sweden.
| |
Collapse
|
17
|
Foxp3-independent mechanism by which TGF-β controls peripheral T cell tolerance. Proc Natl Acad Sci U S A 2017; 114:E7536-E7544. [PMID: 28827353 DOI: 10.1073/pnas.1706356114] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Peripheral T cell tolerance is promoted by the regulatory cytokine TGF-β and Foxp3-expressing Treg cells. However, whether TGF-β and Treg cells are part of the same regulatory module, or exist largely as distinct pathways to repress self-reactive T cells remains incompletely understood. Using a transgenic model of autoimmune diabetes, here we show that ablation of TGF-β receptor II (TβRII) in T cells, but not Foxp3 deficiency, resulted in early-onset diabetes with complete penetrance. The rampant autoimmune disease was associated with enhanced T cell priming and elevated T cell expression of the inflammatory cytokine GM-CSF, concomitant with pancreatic infiltration of inflammatory monocytes that triggered immunopathology. Ablation of the GM-CSF receptor alleviated the monocyte response and inhibited disease development. These findings reveal that TGF-β promotes T cell tolerance primarily via Foxp3-independent mechanisms and prevents autoimmunity in this model by repressing the cross talk between adaptive and innate immune systems.
Collapse
|
18
|
Fingolimod alters the transcriptome profile of circulating CD4+ cells in multiple sclerosis. Sci Rep 2017; 7:42087. [PMID: 28155899 PMCID: PMC5290459 DOI: 10.1038/srep42087] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 01/04/2017] [Indexed: 01/09/2023] Open
Abstract
Multiple sclerosis is a demyelinating disease affecting the central nervous system. T cells are known to contribute to this immune-mediated condition. Fingolimod modulates sphingosine-1-phosphate receptors, thereby preventing the egress of lymphocytes, especially CCR7-expressing CD8+ and CD4+ T cells, from lymphoid tissues. Using Affymetrix Human Transcriptome Arrays (HTA 2.0), we performed a transcriptome profiling analysis of CD4+ cells obtained from the peripheral blood of patients with highly active relapsing-remitting multiple sclerosis. The samples were drawn before the first administration of fingolimod as well as 24 hours and 3 months after the start of therapy. Three months after treatment initiation, 890 genes were found to be differentially expressed with fold-change >2.0 and t-test p-value < 0.001, among them several microRNA precursors. A subset of 272 genes were expressed at lower levels, including CCR7 as expected, while 618 genes showed an increase in expression, e.g., CCR2, CX3CR1, CD39, CD58 as well as LYN, PAK1 and TLR2. To conclude, we studied the gene expression of CD4+ cells to evaluate the effects of fingolimod treatment, and we identified 890 genes to be altered in expression after continuous drug administration. T helper cells circulating in the blood during fingolimod therapy present a distinct gene expression signature.
Collapse
|
19
|
Lavender N, Yang J, Chen SC, Sai J, Johnson CA, Owens P, Ayers GD, Richmond A. The Yin/Yan of CCL2: a minor role in neutrophil anti-tumor activity in vitro but a major role on the outgrowth of metastatic breast cancer lesions in the lung in vivo. BMC Cancer 2017; 17:88. [PMID: 28143493 PMCID: PMC5286656 DOI: 10.1186/s12885-017-3074-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 01/18/2017] [Indexed: 01/15/2023] Open
Abstract
Background The role of the chemokine CCL2 in breast cancer is controversial. While CCL2 recruits and activates pro-tumor macrophages, it is also reported to enhance neutrophil-mediated anti-tumor activity. Moreover, loss of CCL2 in early development enhances breast cancer progression. Methods To clarify these conflicting findings, we examined the ability of CCL2 to alter naïve and tumor entrained neutrophil production of ROS, release of granzyme-B, and killing of tumor cells in multiple mouse models of breast cancer. CCL2 was delivered intranasally in mice to elevate CCL2 levels in the lung and effects on seeding and growth of breast tumor cells were evaluated. The TCGA data base was queried for relationship between CCL2 expression and relapse free survival of breast cancer patients and compared to subsets of breast cancer patients. Results Even though each of the tumor cell lines studied produced approximately equal amounts of CCL2, exogenous delivery of CCL2 to co-cultures of breast tumor cells and neutrophils enhanced the ability of tumor-entrained neutrophils (TEN) to kill the less aggressive 67NR variant of 4T1 breast cancer cells. However, exogenous CCL2 did not enhance naïve or TEN neutrophil killing of more aggressive 4T1 or PyMT breast tumor cells. Moreover, this anti-tumor activity was not observed in vivo. Intranasal delivery of CCL2 to BALB/c mice markedly enhanced seeding and outgrowth of 67NR cells in the lung and increased the recruitment of CD4+ T cells and CD8+ central memory T cells into lungs of tumor bearing mice. There was no significant increase in the recruitment of CD19+ B cells, or F4/80+, Ly6G+ and CD11c + myeloid cells. CCL2 had an equal effect on CD206+ and MHCII+ populations of macrophages, thus balancing the pro- and anti-tumor macrophage cell population. Analysis of the relationship between CCL2 levels and relapse free survival in humans revealed that overall survival is not significantly different between high CCL2 expressing and low CCL2 expressing breast cancer patients grouped together. However, examination of the relationship between high CCL2 expressing basal-like, HER2+ and luminal B breast cancer patients revealed that higher CCL2 expressing tumors in these subgroups have a significantly higher probability of surviving longer than those expressing low CCL2. Conclusions While our in vitro data support a potential anti-tumor role for CCL2 in TEN neutrophil- mediated tumor killing in poorly aggressive tumors, intranasal delivery of CCL2 increased CD4+ T cell recruitment to the pre-metastatic niche of the lung and this correlated with enhanced seeding and growth of tumor cells. These data indicate that effects of CCL2/CCR2 antagonists on the intratumoral leukocyte content should be monitored in ongoing clinical trials using these agents. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3074-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nicole Lavender
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA.,Department of Cancer Biology, Vanderbilt University Medical Center, 432 Preston Research Building, 2220 Pierce Avenue, Nashville, TN, 37232, USA
| | - Jinming Yang
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA.,Department of Cancer Biology, Vanderbilt University Medical Center, 432 Preston Research Building, 2220 Pierce Avenue, Nashville, TN, 37232, USA
| | - Sheau-Chiann Chen
- Department of Cancer Biology, Vanderbilt University Medical Center, 432 Preston Research Building, 2220 Pierce Avenue, Nashville, TN, 37232, USA.,Division of Cancer Biostatistics, Department of Biostatistics, Center for Quantitative Sciences, Nashville, TN, USA
| | - Jiqing Sai
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA.,Department of Cancer Biology, Vanderbilt University Medical Center, 432 Preston Research Building, 2220 Pierce Avenue, Nashville, TN, 37232, USA
| | - C Andrew Johnson
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA.,Department of Cancer Biology, Vanderbilt University Medical Center, 432 Preston Research Building, 2220 Pierce Avenue, Nashville, TN, 37232, USA
| | - Philip Owens
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA.,Department of Cancer Biology, Vanderbilt University Medical Center, 432 Preston Research Building, 2220 Pierce Avenue, Nashville, TN, 37232, USA
| | - Gregory D Ayers
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA.,Division of Cancer Biostatistics, Department of Biostatistics, Center for Quantitative Sciences, Nashville, TN, USA
| | - Ann Richmond
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA. .,Department of Cancer Biology, Vanderbilt University Medical Center, 432 Preston Research Building, 2220 Pierce Avenue, Nashville, TN, 37232, USA.
| |
Collapse
|
20
|
Cytochrome P450 1B1 Contributes to the Development of Angiotensin II-Induced Aortic Aneurysm in Male Apoe(-/-) Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2204-2219. [PMID: 27301358 DOI: 10.1016/j.ajpath.2016.04.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/31/2016] [Accepted: 04/15/2016] [Indexed: 02/07/2023]
Abstract
Cytochrome P450 (CYP) 1B1 is implicated in vascular smooth muscle cell migration, proliferation, and hypertension. We assessed the contribution of CYP1B1 to angiotensin (Ang) II-induced abdominal aortic aneurysm (AAA). Male Apoe(-/-)/Cyp1b1(+/+) and Apoe(-/-)/Cyp1b1(-/-) mice were infused with Ang II or its vehicle for 4 weeks; another group of Apoe(-/-)/Cyp1b1(+/+) mice was coadministered the CYP1B1 inhibitor 2,3',4,5'-tetramethoxystilbene (TMS) every third day for 4 weeks. On day 28 of Ang II infusion, AAAs were analyzed by ultrasound and ex vivo by Vernier calipers, mice were euthanized, and tissues were harvested. Ang II produced AAAs in Apoe(-/-)/Cyp1b1(+/+) mice; mice treated with TMS or Apoe(-/-)/Cyp1b1(-/-) mice had reduced AAAs. Ang II enhanced infiltration of macrophages, T cells, and platelets and increased platelet-derived growth factor D, Pdgfrb, Itga2, and matrix metalloproteinases 2 and 9 expression in aortic lesions; these changes were inhibited in mice treated with TMS and in Apoe(-/-)/Cyp1b1(-/-) mice. Oxidative stress resulted in cyclooxygenase-2 expression in aortic lesions. These effects were minimized in Apoe(-/-)/Cyp1b1(+/+) mice treated with TMS and in Apoe(-/-)/Cyp1b1(-/-) mice and by concurrent treatment with the superoxide scavenger 4-hydroxyl-2,2,6,6-tetramethylpiperidine-1-oxyl. CYP1B1 contributed to the development of Ang II-induced AAA and associated pathogenic events in mice, likely by enhancing oxidative stress and associated signaling events. Thus, CYP1B1 may serve as a target for therapeutic agents for AAA in males.
Collapse
|
21
|
Corliss BA, Azimi MS, Munson J, Peirce SM, Murfee WL. Macrophages: An Inflammatory Link Between Angiogenesis and Lymphangiogenesis. Microcirculation 2016; 23:95-121. [PMID: 26614117 PMCID: PMC4744134 DOI: 10.1111/micc.12259] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/23/2015] [Indexed: 12/14/2022]
Abstract
Angiogenesis and lymphangiogenesis often occur in response to tissue injury or in the presence of pathology (e.g., cancer), and it is these types of environments in which macrophages are activated and increased in number. Moreover, the blood vascular microcirculation and the lymphatic circulation serve as the conduits for entry and exit for monocyte-derived macrophages in nearly every tissue and organ. Macrophages both affect and are affected by the vessels through which they travel. Therefore, it is not surprising that examination of macrophage behaviors in both angiogenesis and lymphangiogenesis has yielded interesting observations that suggest macrophages may be key regulators of these complex growth and remodeling processes. In this review, we will take a closer look at macrophages through the lens of angiogenesis and lymphangiogenesis, examining how their dynamic behaviors may regulate vessel sprouting and function. We present macrophages as a cellular link that spatially and temporally connects angiogenesis with lymphangiogenesis, in both physiological growth and in pathological adaptations, such as tumorigenesis. As such, attempts to therapeutically target macrophages in order to affect these processes may be particularly effective, and studying macrophages in both settings will accelerate the field's understanding of this important cell type in health and disease.
Collapse
Affiliation(s)
- Bruce A. Corliss
- Department of Biomedical Engineering, 415 Lane Road, University of Virginia, Charlottesville, VA 22908
| | - Mohammad S. Azimi
- Department of Biomedical Engineering, 500 Lindy Boggs Energy Center, Tulane University, New Orleans, LA 70118
| | - Jenny Munson
- Department of Biomedical Engineering, 415 Lane Road, University of Virginia, Charlottesville, VA 22908
| | - Shayn M. Peirce
- Department of Biomedical Engineering, 415 Lane Road, University of Virginia, Charlottesville, VA 22908
| | - Walter Lee Murfee
- Department of Biomedical Engineering, 500 Lindy Boggs Energy Center, Tulane University, New Orleans, LA 70118
| |
Collapse
|
22
|
Ragweed-allergic subjects have decreased serum levels of chemokines CCL2, CCL3, CCL4 and CCL5 out of the pollen season. Cent Eur J Immunol 2016; 40:442-6. [PMID: 26862308 PMCID: PMC4737740 DOI: 10.5114/ceji.2015.56965] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/19/2015] [Indexed: 11/17/2022] Open
Abstract
CC-chemokines are important mediators of the allergic responses and regulate the cell trafficking. The aim of this study was to examine the serum levels of CCL2/MCP-1, CCL3/MIP-1α, CCL4/MIP-1β and CCL5/RANTES, and to determine whether there are differences between ragweed-allergic subjects and healthy individuals out of the pollen season. Peripheral blood samples were collected from 24 subjects allergic to ragweed pollen and 12 healthy controls. Serum concentrations of chemokines/cytokines were measured by an enzyme-linked immunosorbent assay. We observed significantly decreased concentrations of CCL2/MCP-1, CCL3/MIP-1α, CCL4/MIP-1β and CCL5/RANTES in the sera of ragweed-allergic patients compared to the healthy individuals (32.2 vs. 106.4 pg/ml, 89.5 vs. 135.7 pg/ml, 63.4 vs. 119.2 pg/ml and 11.2 vs. 18.1 ng/ml, respectively, p < 0.01). In contrast to the CC-chemokines, the serum levels of IL-8/CXCL8 showed a significant increase (p < 0.05) in the allergic group compared to the non-allergic subjects. Interleukin 4 levels were similar in both groups. In the sera of allergic patients, we have also detected significantly elevated levels of ragweed-specific IgE and IgG. However, decreased serum concentrations of the four CC-chemokines and elevated levels of IL-8/CXCL8 can be used as biomarkers for more accurate evaluation of the allergic status of patients with pollen allergy out of the season, to study the mechanisms for activation/inhibition of the subclinical allergic responses and for development of therapeutic strategies.
Collapse
|
23
|
Lisboa LF, Egli A, Fairbanks J, O'Shea D, Manuel O, Husain S, Kumar D, Humar A. CCL8 and the Immune Control of Cytomegalovirus in Organ Transplant Recipients. Am J Transplant 2015; 15:1882-92. [PMID: 25764912 DOI: 10.1111/ajt.13207] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 12/03/2014] [Accepted: 12/24/2014] [Indexed: 02/06/2023]
Abstract
Monitoring of cytomegalovirus cell-mediated immunity is a promising tool for the refinement of preventative and therapeutic strategies posttransplantation. Typically, the interferon-γ response to T cell stimulation is measured. We evaluated a broad range of cytokine and chemokines to better characterize the ex vivo host-response to CMV peptide stimulation. In a cohort of CMV viremic organ transplant recipients, chemokine expression-specifically CCL8 (AUC 0.849 95% CI 0.721-0.978; p = 0.003) and CXCL10 (AUC 0.841, 95% CI 0.707-0.974; p = 0.004)-was associated with control of viral replication. In a second cohort of transplant recipients at high-risk for CMV, the presence of a polymorphism in the CCL8 promoter conferred an increased risk of viral replication after discontinuation of antiviral prophylaxis (logrank hazard ratio 3.6; 95% CI 2.077-51.88). Using cell-sorting experiments, we determined that the primary cell type producing CCL8 in response to CMV peptide stimulation was the monocyte fraction. Finally, in vitro experiments using standard immunosuppressive agents demonstrated a dose-dependent reduction in CCL8 production. Chemokines appear to be important elements of the cell-mediated response to CMV infection posttransplant, as here suggested for CCL8, and translation of this knowledge may allow for the tailoring and improvement of preventative strategies.
Collapse
Affiliation(s)
- L F Lisboa
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - A Egli
- Infection Biology Lab, Department Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - J Fairbanks
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - D O'Shea
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - O Manuel
- Infectious Diseases Service and Transplantation Center, University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - S Husain
- Department of Medicine and Multi-organ Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - D Kumar
- Department of Medicine and Multi-organ Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - A Humar
- Department of Medicine and Multi-organ Transplant Program, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
24
|
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
|
25
|
Wendt ER, Ferry H, Greaves DR, Keshav S. Ratiometric analysis of fura red by flow cytometry: a technique for monitoring intracellular calcium flux in primary cell subsets. PLoS One 2015; 10:e0119532. [PMID: 25835294 PMCID: PMC4383592 DOI: 10.1371/journal.pone.0119532] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 01/21/2015] [Indexed: 11/19/2022] Open
Abstract
Calcium flux is a rapid and sensitive measure of cell activation whose utility could be enhanced with better techniques for data extraction. We describe a technique to monitor calcium flux by flow cytometry, measuring Fura Red calcium dye by ratiometric analysis. This technique has several advantages: 1) using a single calcium dye provides an additional channel for surface marker characterization, 2) allows robust detection of calcium flux by minority cell populations within a heterogeneous population of primary T cells and monocytes 3) can measure total calcium flux and additionally, the proportion of responding cells, 4) can be applied to studying the effects of drug treatment, simultaneously stimulating and monitoring untreated and drug treated cells. Using chemokine receptor activation as an example, we highlight the utility of this assay, demonstrating that only cells expressing a specific chemokine receptor are activated by cognate chemokine ligand. Furthermore, we describe a technique for simultaneously stimulating and monitoring calcium flux in vehicle and drug treated cells, demonstrating the effects of the Gαi inhibitor, pertussis toxin (PTX), on chemokine stimulated calcium flux. The described real time calcium flux assay provides a robust platform for characterizing cell activation within primary cells, and offers a more accurate technique for studying the effect of drug treatment on receptor activation in a heterogeneous population of primary cells.
Collapse
Affiliation(s)
- Emily R. Wendt
- Nuffield Department of Clinical Medicine, Experimental Medicine Division, Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom
| | - Helen Ferry
- Nuffield Department of Clinical Medicine, Experimental Medicine Division, Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom
| | - David R. Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Satish Keshav
- Nuffield Department of Clinical Medicine, Experimental Medicine Division, Translational Gastroenterology Unit, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
26
|
Arlehamn CL, Seumois G, Gerasimova A, Huang C, Fu Z, Yue X, Sette A, Vijayanand P, Peters B. Transcriptional profile of tuberculosis antigen-specific T cells reveals novel multifunctional features. THE JOURNAL OF IMMUNOLOGY 2014; 193:2931-40. [PMID: 25092889 DOI: 10.4049/jimmunol.1401151] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In latent tuberculosis infection (LTBI) spread of the bacteria is contained by a persistent immune response, which includes CD4(+) T cells as important contributors. In this study we show that TB-specific CD4(+) T cells have a characteristic chemokine expression signature (CCR6(+)CXCR3(+)CCR4(-)), and that the overall number of these cells is significantly increased in LTBI donors compared with healthy subjects. We have comprehensively characterized the transcriptional signature of CCR6(+)CXCR3(+)CCR4(-) cells and found significant differences to conventional Th1, Th17, and Th2 cells, but no major changes between healthy and LTBI donors. CCR6(+)CXCR3(+)CCR4(-) cells display lineage-specific signatures of both Th1 and Th17 cells, but also have a unique gene expression program, including genes associated with susceptibility to TB, enhanced T cell activation, enhanced cell survival, and induction of a cytotoxic program akin to CTL cells. Overall, the gene expression signature of CCR6(+)CXCR3(+)CCR4(-) cells reveals characteristics important for controlling latent TB infections.
Collapse
Affiliation(s)
| | - Gregory Seumois
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and
| | - Anna Gerasimova
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and
| | - Charlie Huang
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and
| | - Zheng Fu
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and
| | - Xiaojing Yue
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and
| | - Pandurangan Vijayanand
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and Clinical and Experimental Sciences, Southampton National Institute for Health Research Respiratory Biomedical Research Unit, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Bjoern Peters
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; and
| |
Collapse
|
27
|
Affiliation(s)
- Wakiro Sato
- Department of Immunology; National Institute of Neuroscience; National Center of Neurology and Psychiatry (NCNP); Tokyo Japan
| |
Collapse
|
28
|
The increased type-1 and type-2 chemokine levels in children with acute RSV infection alter the development of adaptive immune responses. BIOMED RESEARCH INTERNATIONAL 2014; 2014:750521. [PMID: 25013801 PMCID: PMC4071813 DOI: 10.1155/2014/750521] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/17/2014] [Accepted: 05/19/2014] [Indexed: 12/16/2022]
Abstract
Severe RSV infections and frequent recurrence could be related to the altered polarization of type-2/type-1 T cells. This increases the importance of determining distinctive chemokines and chemokine receptor profiles on memory T cells. We analyzed systemic adaptive T cell response in the acute (n = 17) and convalescent phase (n = 7) of RSV-infected children, in the acute (n = 11) and convalescent phase (n = 6) of children with other viral respiratory infections (adenovirus and influenza virus), and in healthy children (n = 18). Expression of CCR4 and CXCR3 on effector-memory (TEM) and central-memory (TCM) T cells was compared between tested groups. Serum concentrations of specific chemokines were determined. High CXCL10 levels were detected in acutely infected children regardless of virus pathogen, whereas increased CCL17 production was RSV-specific. Higher percentages of CCR4+ CD4 TEM cells in acute RSV infection were accompanied with higher percentages of CXCR3+ CD8 TEM cells, whereas the development of long-lived memory CXCR3+ CD4 and CD8 TCM cells seems to be compromised, as only children with other viral infections had higher percentages in the convalescent phase. Presence of type-2 and type-1 adaptive antiviral immune response, together with insufficient development of long-lived type-1 T cell memory, could play an important role in RSV pathogenesis and reinfection.
Collapse
|
29
|
Morandi F, Rouas-Freiss N, Pistoia V. The emerging role of soluble HLA-G in the control of chemotaxis. Cytokine Growth Factor Rev 2014; 25:327-35. [PMID: 24882150 DOI: 10.1016/j.cytogfr.2014.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/10/2014] [Accepted: 04/28/2014] [Indexed: 10/25/2022]
Abstract
HLA-G is an immunosuppressive molecule, that impairs the function of different immune cell populations, both in physiological and pathological conditions. Here, we have analyzed data obtained by our group and others regarding sHLA-G concentration in plasma from patients with different diseases. Next, we have summarized novel data regarding the impairment of chemotaxis of different immune effector cells mediated by sHLA-G. Finally, we have discussed the impact of this function on the immune response during cancer, viral infection, autoimmunity, and on B cell differentiation in secondary lymphoid organs. In conclusion, we have delineated a role of sHLA-G in the control of chemotaxis of immune effector cells, that may be relevant to modulate immune responses in different settings.
Collapse
Affiliation(s)
- Fabio Morandi
- Laboratory of Oncology, Istituto Giannina Gaslini, Via Gaslini 1, 16147 Genoa, Italy.
| | - Nathalie Rouas-Freiss
- CEA, Institut des Maladies Emergentes et des Therapies Innovantes (iMETI), Service de Recherche en Hemato-Immunologie (SRHI), Hopital Saint-Louis, Avenue Claude Vellefaux 1, 75010 Paris, France.
| | - Vito Pistoia
- Laboratory of Oncology, Istituto Giannina Gaslini, Via Gaslini 1, 16147 Genoa, Italy.
| |
Collapse
|
30
|
Farber DL, Yudanin NA, Restifo NP. Human memory T cells: generation, compartmentalization and homeostasis. Nat Rev Immunol 2014; 14:24-35. [PMID: 24336101 PMCID: PMC4032067 DOI: 10.1038/nri3567] [Citation(s) in RCA: 631] [Impact Index Per Article: 57.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Memory T cells constitute the most abundant lymphocyte population in the body for the majority of a person's lifetime; however, our understanding of memory T cell generation, function and maintenance mainly derives from mouse studies, which cannot recapitulate the exposure to multiple pathogens that occurs over many decades in humans. In this Review, we discuss studies focused on human memory T cells that reveal key properties of these cells, including subset heterogeneity and diverse tissue residence in multiple mucosal and lymphoid tissue sites. We also review how the function and the adaptability of human memory T cells depend on spatial and temporal compartmentalization.
Collapse
Affiliation(s)
- Donna L Farber
- 1] Columbia Center for Translational Immunology and Department of Microbiology and Immunology, Columbia University Medical Center, 650 West 168th Street, BB1501, New York, New York 10032, USA. [2] Department of Surgery, Columbia University Medical Center, 650 West 168th Street, BB1501, New York 10032, USA
| | - Naomi A Yudanin
- Columbia Center for Translational Immunology and Department of Microbiology and Immunology, Columbia University Medical Center, 650 West 168th Street, BB1501, New York, New York 10032, USA
| | - Nicholas P Restifo
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
31
|
Schreiber HA, Loschko J, Karssemeijer RA, Escolano A, Meredith MM, Mucida D, Guermonprez P, Nussenzweig MC. Intestinal monocytes and macrophages are required for T cell polarization in response to Citrobacter rodentium. ACTA ACUST UNITED AC 2013; 210:2025-39. [PMID: 24043764 PMCID: PMC3782042 DOI: 10.1084/jem.20130903] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Using a new mouse model, the specific deletion of monocytes and macrophages reveals that, although not required to initiate immunity to Citrobacter rodentium, they contribute to the adaptive response via IL-12 secretion to induced IFN-γ+ Th1 polarization. Dendritic cells (DCs), monocytes, and macrophages are closely related phagocytes that share many phenotypic features and, in some cases, a common developmental origin. Although the requirement for DCs in initiating adaptive immune responses is well appreciated, the role of monocytes and macrophages remains largely undefined, in part because of the lack of genetic tools enabling their specific depletion. Here, we describe a two-gene approach that requires overlapping expression of LysM and Csf1r to define and deplete monocytes and macrophages. The role of monocytes and macrophages in immunity to pathogens was tested by their selective depletion during infection with Citrobacter rodentium. Although neither cell type was required to initiate immunity, monocytes and macrophages contributed to the adaptive immune response by secreting IL-12, which induced Th1 polarization and IFN-γ secretion. Thus, whereas DCs are indispensable for priming naive CD4+ T cells, monocytes and macrophages participate in intestinal immunity by producing mediators that direct T cell polarization.
Collapse
Affiliation(s)
- Heidi A Schreiber
- Laboratory of Molecular Immunology, 2 Mucosal Immunology and 3 Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065
| | | | | | | | | | | | | | | |
Collapse
|
32
|
|
33
|
Sato W, Tomita A, Ichikawa D, Lin Y, Kishida H, Miyake S, Ogawa M, Okamoto T, Murata M, Kuroiwa Y, Aranami T, Yamamura T. CCR2(+)CCR5(+) T cells produce matrix metalloproteinase-9 and osteopontin in the pathogenesis of multiple sclerosis. THE JOURNAL OF IMMUNOLOGY 2012; 189:5057-65. [PMID: 23071279 DOI: 10.4049/jimmunol.1202026] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the CNS that is presumably mediated by CD4(+) autoimmune T cells. Although both Th1 and Th17 cells have the potential to cause inflammatory CNS pathology in rodents, the identity of pathogenic T cells remains unclear in human MS. Given that each Th cell subset preferentially expresses specific chemokine receptors, we were interested to know whether T cells defined by a particular chemokine receptor profile play an active role in the pathogenesis of MS. In this article, we report that CCR2(+)CCR5(+) T cells constitute a unique population selectively enriched in the cerebrospinal fluid of MS patients during relapse but not in patients with other neurologic diseases. After polyclonal stimulation, the CCR2(+)CCR5(+) T cells exhibited a distinct ability to produce matrix metalloproteinase-9 and osteopontin, which are involved in the CNS pathology of MS. Furthermore, after TCR stimulation, the CCR2(+)CCR5(+) T cells showed a higher invasive potential across an in vitro blood-brain barrier model compared with other T cells. Of note, the CCR2(+)CCR5(+) T cells from MS patients in relapse are reactive to myelin basic protein, as assessed by production of IFN-γ. We also demonstrated that the CCR6(-), but not the CCR6(+), population within CCR2(+)CCR5(+) T cells was highly enriched in the cerebrospinal fluid during MS relapse (p < 0.0005) and expressed higher levels of IFN-γ and matrix metalloproteinase-9. Taken together, we propose that autoimmune CCR2(+)CCR5(+)CCR6(-) Th1 cells play a crucial role in the pathogenesis of MS.
Collapse
Affiliation(s)
- Wakiro Sato
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Aomatsu T, Imaeda H, Takahashi K, Fujimoto T, Kasumi E, Yoden A, Tamai H, Fujiyama Y, Andoh A. Tacrolimus (FK506) suppresses TNF-α-induced CCL2 (MCP-1) and CXCL10 (IP-10) expression via the inhibition of p38 MAP kinase activation in human colonic myofibroblasts. Int J Mol Med 2012; 30:1152-8. [PMID: 22895606 DOI: 10.3892/ijmm.2012.1094] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 07/09/2012] [Indexed: 11/06/2022] Open
Abstract
In order to investigate the molecular mechanisms underlying the immunosuppressive effects of tacrolimus (FK506) on intestinal inflammation, we examined whether FK506 effects cytokine/chemokine secretion in human colonic myofibroblasts. Human colonic myofibroblasts were isolated from normal human colonic tissue. The mRNA and protein expression for human CCL2 and CXCL10 were analyzed by real-time PCR and ELISA, respectively. p38 MAP kinase activation was evaluated by western blotting. Tacrolimus (1 µM) suppressed tumor necrosis factor (TNF)-α-induced CCL2 and CXCL10 mRNA expression, but did not modulate TNF-α-induced interleukin (IL)-6 or CXCL8 mRNA expression. Dose-dependent, inhibitory effects of tacrolimus on CCL2 and CXCL10 expression were observed at the mRNA and protein levels. Significant inhibitory effects of tacrolimus were observed at concentrations as low as 0.5 µM for CCL2 and 0.1 µM for CXCL10, respectively. TNF-α-induced CCL2 and CXCL10 expression depended on p38 MAP kinase activation, and tacrolimus strongly inhibited the TNF-α-induced phosphorylation of p38 MAP kinase. Tacrolimus did not affect interferon (IFN)-γ-induced signaling transducer and activator of transcription (STAT)-1 phosphorylation, nor did it modulate CXCL10 mRNA and protein expression. In conclusion, tacrolimus suppressed CCL2 and CXCL10 expression in human colonic myofibroblasts. These inhibitory effects of tacrolimus may play key roles in the therapeutic effects of colonic inflammation in inflammatory bowel disease (IBD) patients.
Collapse
Affiliation(s)
- Tomoki Aomatsu
- Department of Medicine, Shiga University of Medical Science, Seta Tsukinowa, Otsu 520-2192, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gitau EN, Tuju J, Stevenson L, Kimani E, Karanja H, Marsh K, Bull PC, Urban BC. T-cell responses to the DBLα-tag, a short semi-conserved region of the Plasmodium falciparum membrane erythrocyte protein 1. PLoS One 2012; 7:e30095. [PMID: 22272280 PMCID: PMC3260199 DOI: 10.1371/journal.pone.0030095] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 12/09/2011] [Indexed: 11/25/2022] Open
Abstract
The Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) is a variant surface antigen expressed on mature forms of infected erythrocytes. It is considered an important target of naturally acquired immunity. Despite its extreme sequence heterogeneity, variants of PfEMP1 can be stratified into distinct groups. Group A PfEMP1 have been independently associated with low host immunity and severe disease in several studies and are now of potential interest as vaccine candidates. Although antigen-specific antibodies are considered the main effector mechanism in immunity to malaria, the induction of efficient and long-lasting antibody responses requires CD4+ T-cell help. To date, very little is known about CD4+ T-cell responses to PfEMP1 expressed on clinical isolates. The DBLα-tag is a small region from the DBLα-domain of PfEMP1 that can be amplified with universal primers and is accessible in clinical parasite isolates. We identified the dominant expressed PfEMP1 in 41 individual clinical parasite isolates and expressed the corresponding DBLα-tag as recombinant antigen. Individual DBLα-tags were then used to activate CD4+ T-cells from acute and convalescent blood samples in children who were infected with the respective clinical parasite isolate. Here we show that CD4+ T-cell responses to the homologous DBLα-tag were induced in almost all children during acute malaria and maintained in some for 4 months. Children infected with parasites that dominantly expressed group A-like PfEMP1 were more likely to maintain antigen-specific IFNγ-producing CD4+ T-cells than children infected with parasites dominantly expressing other PfEMP1. These results suggest that group A-like PfEMP1 may induce long-lasting effector memory T-cells that might be able to provide rapid help to variant-specific B cells. Furthermore, a number of children induced CD4+ T-cell responses to heterologous DBLα-tags, suggesting that CD4+ T-cells may recognise shared epitopes between several DBLα-tags.
Collapse
Affiliation(s)
- Evelyn N. Gitau
- KEMRI-Wellcome Trust Collaborative Programme, Centre for Geographic Medicine Coast, Kilifi, Kenya
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - James Tuju
- KEMRI-Wellcome Trust Collaborative Programme, Centre for Geographic Medicine Coast, Kilifi, Kenya
| | - Liz Stevenson
- KEMRI-Wellcome Trust Collaborative Programme, Centre for Geographic Medicine Coast, Kilifi, Kenya
| | - Eva Kimani
- KEMRI-Wellcome Trust Collaborative Programme, Centre for Geographic Medicine Coast, Kilifi, Kenya
| | - Henry Karanja
- KEMRI-Wellcome Trust Collaborative Programme, Centre for Geographic Medicine Coast, Kilifi, Kenya
| | - Kevin Marsh
- KEMRI-Wellcome Trust Collaborative Programme, Centre for Geographic Medicine Coast, Kilifi, Kenya
- Centre for Tropical Medicine, Nuffield Department of Internal Medicine, Oxford University, Oxford, United Kingdom
| | - Peter C. Bull
- KEMRI-Wellcome Trust Collaborative Programme, Centre for Geographic Medicine Coast, Kilifi, Kenya
- Centre for Tropical Medicine, Nuffield Department of Internal Medicine, Oxford University, Oxford, United Kingdom
| | - Britta C. Urban
- KEMRI-Wellcome Trust Collaborative Programme, Centre for Geographic Medicine Coast, Kilifi, Kenya
- Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
Shulman Z, Cohen SJ, Roediger B, Kalchenko V, Jain R, Grabovsky V, Klein E, Shinder V, Stoler-Barak L, Feigelson SW, Meshel T, Nurmi SM, Goldstein I, Hartley O, Gahmberg CG, Etzioni A, Weninger W, Ben-Baruch A, Alon R. Transendothelial migration of lymphocytes mediated by intraendothelial vesicle stores rather than by extracellular chemokine depots. Nat Immunol 2011; 13:67-76. [PMID: 22138716 DOI: 10.1038/ni.2173] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 10/25/2011] [Indexed: 12/11/2022]
Abstract
Chemokines presented by the endothelium are critical for integrin-dependent adhesion and transendothelial migration of naive and memory lymphocytes. Here we found that effector lymphocytes of the type 1 helper T cell (T(H)1 cell) and type 1 cytotoxic T cell (T(C)1 cell) subtypes expressed adhesive integrins that bypassed chemokine signals and established firm arrests on variably inflamed endothelial barriers. Nevertheless, the transendothelial migration of these lymphocytes strictly depended on signals from guanine nucleotide-binding proteins of the G(i) type and was promoted by multiple endothelium-derived inflammatory chemokines, even without outer endothelial surface exposure. Instead, transendothelial migration-promoting endothelial chemokines were stored in vesicles docked on actin fibers beneath the plasma membranes and were locally released within tight lymphocyte-endothelial synapses. Thus, effector T lymphocytes can cross inflamed barriers through contact-guided consumption of intraendothelial chemokines without surface-deposited chemokines or extraendothelial chemokine gradients.
Collapse
Affiliation(s)
- Ziv Shulman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|