1
|
Ryu S, Kim KA, Kim J, Lee DH, Bae YS, Lee H, Kim BC, Kim HY. The protective roles of integrin α4β7 and Amphiregulin-expressing innate lymphoid cells in lupus nephritis. Cell Mol Immunol 2024; 21:723-737. [PMID: 38806623 PMCID: PMC11214630 DOI: 10.1038/s41423-024-01178-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 04/27/2024] [Indexed: 05/30/2024] Open
Abstract
Type 2 innate lymphoid cells (ILC2s) have emerged as key regulators of the immune response in renal inflammatory diseases such as lupus nephritis. However, the mechanisms underlying ILC2 adhesion and migration in the kidney remain poorly understood. Here, we revealed the critical role of integrin α4β7 in mediating renal ILC2 adhesion and function. We found that integrin α4β7 enables the retention of ILC2s in the kidney by binding to VCAM-1, E-cadherin, or fibronectin on structural cells. Moreover, integrin α4β7 knockdown reduced the production of the reparative cytokine amphiregulin (Areg) by ILC2s. In lupus nephritis, TLR7/9 signaling within the kidney microenvironment downregulates integrin α4β7 expression, leading to decreased Areg production and promoting the egress of ILC2s. Notably, IL-33 treatment upregulated integrin α4β7 and Areg expression in ILC2s, thereby enhancing survival and reducing inflammation in lupus nephritis. Together, these findings highlight the potential of targeting ILC2 adhesion as a therapeutic strategy for autoimmune kidney diseases.
Collapse
Affiliation(s)
- Seungwon Ryu
- Department of Microbiology, Gachon University College of Medicine, Incheon, 21999, South Korea
| | - Kyung Ah Kim
- Department of Nano-Bioengineering, Incheon National University, Incheon, 22012, South Korea
| | - Jinwoo Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, 03080, South Korea
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, 03080, South Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, 03080, South Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, SRC Center for Immune Research on Non-lymphoid Organs, Sungkyunkwan University, Suwon, 16419, South Korea
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Hajeong Lee
- Division of Nephrology, Department of Internal Medicine, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Byoung Choul Kim
- Department of Nano-Bioengineering, Incheon National University, Incheon, 22012, South Korea
| | - Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea.
- Department of Biological Sciences, SRC Center for Immune Research on Non-lymphoid Organs, Sungkyunkwan University, Suwon, 16419, South Korea.
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, 03080, South Korea.
| |
Collapse
|
2
|
Rodger B, Stagg AJ, Lindsay JO. The role of circulating T cells with a tissue resident phenotype (ex-T RM) in health and disease. Front Immunol 2024; 15:1415914. [PMID: 38817613 PMCID: PMC11137204 DOI: 10.3389/fimmu.2024.1415914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 04/26/2024] [Indexed: 06/01/2024] Open
Abstract
Tissue-resident memory T cells (TRM) are long-lived memory lymphocytes that persist in non-lymphoid tissues and provide the first line of defence against invading pathogens. They adapt to their environment in a tissue-specific manner, exerting effective pathogen control through a diverse T cell receptor (TCR) repertoire and the expression of proinflammatory cytokines and cytolytic proteins. More recently, several studies have indicated that TRM can egress from the tissue into the blood as so-called "ex-TRM", or "circulating cells with a TRM phenotype". The numerically small ex-TRM population can re-differentiate in the circulation, giving rise to new memory and effector T cells. Following their egress, ex-TRM in the blood and secondary lymphoid organs can be identified based on their continued expression of the residency marker CD103, alongside other TRM-like features. Currently, it is unclear whether exit is a stochastic process, or is actively triggered in response to unknown factors. Also, it is not known whether a subset or all TRM are able to egress. Ex-TRM may be beneficial in health, as mobilisation of specialised TRM and their recruitment to both their site of origin as well as distant tissues results in an efficient distribution of the immune response. However, there is emerging evidence of a pathogenic role for ex-TRM, with a suggestion that they may perpetuate both local and distant tissue inflammation. Here, we review the evidence for the existence of ex-TRM and examine their potential involvement in disease pathogenesis.
Collapse
Affiliation(s)
- Beverley Rodger
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Andrew J. Stagg
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - James O. Lindsay
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Department of Gastroenterology, Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| |
Collapse
|
3
|
Haugh A, Daud AI. Therapeutic Strategies in BRAF V600 Wild-Type Cutaneous Melanoma. Am J Clin Dermatol 2024; 25:407-419. [PMID: 38329690 DOI: 10.1007/s40257-023-00841-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 02/09/2024]
Abstract
There have been many recent advances in melanoma therapy. While 50% of melanomas have a BRAF mutation and are a target for BRAF inhibitors, the remaining 50% are BRAF wild-type. Immune checkpoint inhibitors targeting PD-1, cytotoxic T-lymphocyte-associated protein 4 (CTLA4) and lymphocyte activated gene-3 (Lag-3) are all approved for the treatment of patients with advanced BRAF wild-type melanoma; however, treatment of this patient population following initial immune checkpoint blockade is a current therapeutic challenge given the lack of other efficacious options. Here, we briefly review available US FDA-approved therapies for BRAF wild-type melanoma and focus on developing treatment avenues for this heterogeneous group of patients. We review the basics of genomic features of both BRAF mutant and BRAF wild-type melanoma as well as efforts underway to develop new targeted therapies involving the mitogen-activated protein kinase (MAPK) pathway for patients with BRAF wild-type tumors. We then focus on novel immunotherapies, including developing checkpoint inhibitors and agonists, cytokine therapies, oncolytic viruses and tumor-infiltrating lymphocytes, all of which represent potential therapeutic avenues for patients with BRAF wild-type melanoma who progress on currently approved immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Alexandra Haugh
- Department of Medicine, University of California San Francisco, 550 16th Street, 6809, San Francisco, CA, 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Adil I Daud
- Department of Medicine, University of California San Francisco, 550 16th Street, 6809, San Francisco, CA, 94158, USA.
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
4
|
Lobão B, Lourenço D, Giga A, Mendes-Bastos P. From PsO to PsA: the role of T RM and Tregs in psoriatic disease, a systematic review of the literature. Front Med (Lausanne) 2024; 11:1346757. [PMID: 38405187 PMCID: PMC10884248 DOI: 10.3389/fmed.2024.1346757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/25/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction Psoriasis (PsO) is a chronic skin condition driven by immune mediators like TNFα, INFγ, IL-17, and IL-23. Psoriatic arthritis (PsA) can develop in PsO patients. Although psoriatic lesions may apparently resolve with therapy, subclinical cutaneous inflammation may persist. The role of tissue-resident memory T-cells (TRM), and regulatory T cells (Tregs) that also contribute to chronic inflammation are being explored in this context. This systematic review explores TRM and Tregs in psoriatic disease (PsD) and its progression. Methods A systematic review, following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, was performed using Pubmed® and Web of Science™ databases on June 3rd 2023, using patient/population, intervention, comparison, and outcomes (PICO) criteria limited to the English language. Results A total of 62 reports were identified and included. In PsO, chronic inflammation is driven by cytokines including IL-17 and IL-23, and cellular mediators such as CD8+ and CD4+ T cells. TRM contributes to local inflammation, while Tregs may be dysfunctional in psoriatic skin lesions. Secukinumab and guselkumab, which target IL-17A and the IL-23p19 subunit, respectively, have different effects on CD8+ TRM and Tregs during PsO treatment. Inhibition of IL-23 may provide better long-term results due to its impact on the Treg to CD8+ TRM ratio. IL-23 may contribute to inflammation persisting even after treatment. In PsA, subclinical enthesitis is perceived as an early occurence, and Th17 cells are involved in this pathogenic process. Recent EULAR guidelines highlight the importance of early diagnosis and treatment to intercept PsA. In PsA, CD8+ TRM cells are present in synovial fluid and Tregs are reduced in peripheral blood. The progression from PsO to PsA is marked by a shift in immune profiles, with specific T-cells subsets playing key roles in perpetuating inflammation. Early intervention targeting TRM cells may hold promising, but clinical studies are limited. Ongoing studies such as IVEPSA and PAMPA aim to improve our knowledge regarding PsA interception in high-risk PsO patients, emphasizing the need for further research in this area. Conclusion Early intervention is crucial for PsO patients at high risk of PsA; T cells, particularly type 17 helper T cells, and CD8+ cells are key in the progression from PsO-to-PsA. Early targeting of TRM in PsD shows promise but more research is needed.
Collapse
Affiliation(s)
- Bárbara Lobão
- Instituto Português de Reumatologia, Lisboa, Portugal
- Centro Hospitalar de Setúbal, Setúbal, Portugal
| | | | - Ana Giga
- Janssen Portugal, Oeiras, Portugal
| | | |
Collapse
|
5
|
Elliott Williams M, Hardnett FP, Sheth AN, Wein AN, Li ZRT, Radzio-Basu J, Dinh C, Haddad LB, Collins EMB, Ofotokun I, Antia R, Scharer CD, Garcia-Lerma JG, Kohlmeier JE, Swaims-Kohlmeier A. The menstrual cycle regulates migratory CD4 T-cell surveillance in the female reproductive tract via CCR5 signaling. Mucosal Immunol 2024; 17:41-53. [PMID: 37866719 PMCID: PMC10990418 DOI: 10.1016/j.mucimm.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023]
Abstract
Despite their importance for immunity against sexually transmitted infections, the composition of female reproductive tract (FRT) memory T-cell populations in response to changes within the local tissue environment under the regulation of the menstrual cycle remains poorly defined. Here, we show that in humans and pig-tailed macaques, the cycle determines distinct clusters of differentiation 4 T-cell surveillance behaviors by subsets corresponding to migratory memory (TMM) and resident memory T cells. TMM displays tissue-itinerant trafficking characteristics, restricted distribution within the FRT microenvironment, and distinct effector responses to infection. Gene pathway analysis by RNA sequencing identified TMM-specific enrichment of genes involved in hormonal regulation and inflammatory responses. FRT T-cell subset fluctuations were discovered that synchronized to cycle-driven CCR5 signaling. Notably, oral administration of a CCR5 antagonist drug blocked TMM trafficking. Taken together, this study provides novel insights into the dynamic nature of FRT memory CD4 T cells and identifies the menstrual cycle as a key regulator of immune surveillance at the site of STI pathogen exposure.
Collapse
Affiliation(s)
- M Elliott Williams
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Felica P Hardnett
- Division of HIV Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Anandi N Sheth
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine and Grady Health System, Atlanta, GA, USA
| | - Alexander N Wein
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Zheng-Rong Tiger Li
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jessica Radzio-Basu
- Division of HIV Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Chuong Dinh
- Division of HIV Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Lisa B Haddad
- Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Elizabeth M B Collins
- Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Igho Ofotokun
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine and Grady Health System, Atlanta, GA, USA
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Christopher D Scharer
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - J Gerardo Garcia-Lerma
- Division of HIV Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jacob E Kohlmeier
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Alison Swaims-Kohlmeier
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA, USA; Division of HIV Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA; Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
6
|
Ruef N, Martínez Magdaleno J, Ficht X, Purvanov V, Palayret M, Wissmann S, Pfenninger P, Stolp B, Thelen F, Barreto de Albuquerque J, Germann P, Sharpe J, Abe J, Legler DF, Stein JV. Exocrine gland-resident memory CD8 + T cells use mechanosensing for tissue surveillance. Sci Immunol 2023; 8:eadd5724. [PMID: 38134242 DOI: 10.1126/sciimmunol.add5724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 11/09/2023] [Indexed: 12/24/2023]
Abstract
Tissue-resident CD8+ T cells (TRM) continuously scan peptide-MHC (pMHC) complexes in their organ of residence to intercept microbial invaders. Recent data showed that TRM lodged in exocrine glands scan tissue in the absence of any chemoattractant or adhesion receptor signaling, thus bypassing the requirement for canonical migration-promoting factors. The signals eliciting this noncanonical motility and its relevance for organ surveillance have remained unknown. Using mouse models of viral infections, we report that exocrine gland TRM autonomously generated front-to-back F-actin flow for locomotion, accompanied by high cortical actomyosin contractility, and leading-edge bleb formation. The distinctive mode of exocrine gland TRM locomotion was triggered by sensing physical confinement and was closely correlated with nuclear deformation, which acts as a mechanosensor via an arachidonic acid and Ca2+ signaling pathway. By contrast, naïve CD8+ T cells or TRM surveilling microbe-exposed epithelial barriers did not show mechanosensing capacity. Inhibition of nuclear mechanosensing disrupted exocrine gland TRM scanning and impaired their ability to intercept target cells. These findings indicate that confinement is sufficient to elicit autonomous T cell surveillance in glands with restricted chemokine expression and constitutes a scanning strategy that complements chemosensing-dependent migration.
Collapse
Affiliation(s)
- Nora Ruef
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Jose Martínez Magdaleno
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Xenia Ficht
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 22, 4058 Basel, Switzerland
| | - Vladimir Purvanov
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, 8280 Kreuzlingen, Switzerland
| | - Matthieu Palayret
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Stefanie Wissmann
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Petra Pfenninger
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Bettina Stolp
- Department for Infectious Diseases, Integrative Virology, Center for Integrative Infectious Disease Research, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Flavian Thelen
- Department of Medical Oncology and Hematology, University of Zürich and University Hospital Zürich, 8091 Zürich, Switzerland
| | | | - Philipp Germann
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08003 Barcelona, Spain
| | - James Sharpe
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08003 Barcelona, Spain
- European Molecular Biology Laboratory (EMBL) Barcelona, 08003 Barcelona, Spain
- Institucio' Catalana de Recerca i Estudis Avancats (ICREA), 08010 Barcelona, Spain
| | - Jun Abe
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, 8280 Kreuzlingen, Switzerland
- Faculty of Biology, University of Konstanz, 78464 Konstanz, Germany
- Theodor Kocher Institute, University of Bern, 3011 Bern, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| |
Collapse
|
7
|
Mittra S, Harding SM, Kaech SM. Memory T Cells in the Immunoprevention of Cancer: A Switch from Therapeutic to Prophylactic Approaches. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:907-916. [PMID: 37669503 PMCID: PMC10491418 DOI: 10.4049/jimmunol.2300049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/24/2023] [Indexed: 09/07/2023]
Abstract
Cancer immunoprevention, the engagement of the immune system to prevent cancer, is largely overshadowed by therapeutic approaches to treating cancer after detection. Vaccines or, alternatively, the utilization of genetically engineered memory T cells could be methods of engaging and creating cancer-specific T cells with superb memory, lenient activation requirements, potent antitumor cytotoxicity, tumor surveillance, and resilience against immunosuppressive factors in the tumor microenvironment. In this review we analyze memory T cell subtypes based on their potential utility in cancer immunoprevention with regard to longevity, localization, activation requirements, and efficacy in fighting cancers. A particular focus is on how both tissue-resident memory T cells and stem memory T cells could be promising subtypes for engaging in immunoprevention.
Collapse
Affiliation(s)
- Siddhesh Mittra
- University of Toronto Schools, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Shane M. Harding
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Departments of Radiation Oncology and Immunology, University of Toronto; Toronto, Canada
| | - Susan M. Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
8
|
Povoleri GAM, Durham LE, Gray EH, Lalnunhlimi S, Kannambath S, Pitcher MJ, Dhami P, Leeuw T, Ryan SE, Steel KJA, Kirkham BW, Taams LS. Psoriatic and rheumatoid arthritis joints differ in the composition of CD8+ tissue-resident memory T cell subsets. Cell Rep 2023; 42:112514. [PMID: 37195862 PMCID: PMC10790246 DOI: 10.1016/j.celrep.2023.112514] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/21/2023] [Accepted: 05/01/2023] [Indexed: 05/19/2023] Open
Abstract
CD69+CD103+ tissue-resident memory T (TRM) cells are important drivers of inflammation. To decipher their role in inflammatory arthritis, we apply single-cell, high-dimensional profiling to T cells from the joints of patients with psoriatic arthritis (PsA) or rheumatoid arthritis (RA). We identify three groups of synovial CD8+CD69+CD103+ TRM cells: cytotoxic and regulatory T (Treg)-like TRM cells are present in both PsA and RA, while CD161+CCR6+ type 17-like TRM cells with a pro-inflammatory cytokine profile (IL-17A+TNFα+IFNγ+) are specifically enriched in PsA. In contrast, only one population of CD4+CD69+CD103+ TRM cells is detected and at similarly low frequencies in both diseases. Type 17-like CD8+ TRM cells have a distinct transcriptomic signature and a polyclonal, but distinct, TCR repertoire. Type 17-like cells are also enriched in CD8+CD103- T cells in PsA compared with RA. These findings illustrate differences in the immunopathology of PsA and RA, with a particular enrichment for type 17 CD8+ T cells in the PsA joint.
Collapse
Affiliation(s)
- Giovanni A M Povoleri
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Lucy E Durham
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Elizabeth H Gray
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Sylvine Lalnunhlimi
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Shichina Kannambath
- BRC Genomics Core, NIHR Biomedical Research Center, Guy's and St Thomas' NHS Foundation Trust and King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Michael J Pitcher
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK
| | - Pawan Dhami
- BRC Genomics Core, NIHR Biomedical Research Center, Guy's and St Thomas' NHS Foundation Trust and King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Thomas Leeuw
- Immunology & Inflammation Research TA, Sanofi-Aventis Deutschland GmbH, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Sarah E Ryan
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Kathryn J A Steel
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK
| | - Bruce W Kirkham
- Rheumatology Department, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Leonie S Taams
- Centre for Inflammation Biology and Cancer Immunology (CIBCI), Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London SE1 1UL, UK.
| |
Collapse
|
9
|
Rosato PC, Lotfi-Emran S, Joag V, Wijeyesinghe S, Quarnstrom CF, Degefu HN, Nedellec R, Schenkel JM, Beura LK, Hangartner L, Burton DR, Masopust D. Tissue-resident memory T cells trigger rapid exudation and local antibody accumulation. Mucosal Immunol 2023; 16:17-26. [PMID: 36657662 PMCID: PMC10338064 DOI: 10.1016/j.mucimm.2022.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 01/18/2023]
Abstract
Adaptive immunity is didactically partitioned into humoral and cell-mediated effector mechanisms, which may imply that each arm is separate and does not function together. Here, we report that the activation of CD8+ resident memory T cells (TRM) in nonlymphoid tissues triggers vascular permeability, which facilitates rapid distribution of serum antibodies into local tissues. TRM reactivation was associated with transcriptional upregulation of antiviral signaling pathways as well as Fc receptors and components of the complement cascade. Effects were local, but evidence is presented that TRM in brain and reproductive mucosa are both competent to induce rapid antibody exudation. TRM reactivation in the mouse female genital tract increased local concentrations of virus-specific neutralizing antibodies, including anti-vesicular stomatitis virus, and passively transferred anti-HIV antibodies. We showed that this response was sufficient to increase the efficacy of ex vivo vesicular stomatitis virus neutralization. These results indicate that CD8+ TRM antigen recognition can enhance local humoral immunity.
Collapse
Affiliation(s)
- Pamela C Rosato
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA; Geisel School of Medicine at Dartmouth College, Dartmouth Cancer Center, Department of Microbiology and Immunology, Lebanon, NH, USA
| | - Sahar Lotfi-Emran
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA
| | - Vineet Joag
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA
| | - Sathi Wijeyesinghe
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA
| | - Clare F Quarnstrom
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA
| | - Hanna N Degefu
- Geisel School of Medicine at Dartmouth College, Dartmouth Cancer Center, Department of Microbiology and Immunology, Lebanon, NH, USA
| | - Rebecca Nedellec
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Jason M Schenkel
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA
| | - Lalit K Beura
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA; Brown University, Department of Molecular Microbiology and Immunology, Providence, RI, USA
| | - Lars Hangartner
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Dennis R Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - David Masopust
- University of Minnesota, Center for Immunology, Department of Microbiology and Immunology, Minneapolis, MN, USA.
| |
Collapse
|
10
|
Yang H, Xie Y, Li C. Understanding the mechanisms for COVID-19 vaccine's protection against infection and severe disease. Expert Rev Vaccines 2023; 22:186-192. [PMID: 36715150 DOI: 10.1080/14760584.2023.2174529] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Multiple COVID-19 vaccines have been approved and employed in the fight against the pandemic. However, these vaccines have limited long-term effectiveness against severe cases and a decreased ability to prevent mild disease. AREAS COVERED This review discusses the relevant factors influencing the efficacy of the vaccines against mild and severe infection, analyzes the possible underlying mechanisms contributing to the different outcomes in terms of vaccine function and disease progression, and proposes improvements for the next generation of vaccines. EXPERT OPINION The reduced efficacy of the COVID-19 vaccine in the prevention of viral infection is closely related to the emergence of novel SARS-CoV-2 variants and their rapid transmission ability. Fundamentally, the immune responses induced by COVID-19 vaccines cannot effectively halt virus replication in the upper respiratory tract because only a limited number of specific antibodies reach these areas and decrease in concentration over time. However, the established immune response can provide sufficient protection against severe diseases by blocking viral infection of the lower respiratory tract or lung owing to sufficient antibody repertoires and memory responses. Considering this situation, future COVID-19 vaccines should have the potential to replenish the mucosal immune response in the respiratory tract to prevent viral infection.
Collapse
Affiliation(s)
- Huijie Yang
- Divsion of respiratory virus vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Ying Xie
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Changgui Li
- Divsion of respiratory virus vaccines, National Institutes for Food and Drug Control, Beijing, China
| |
Collapse
|
11
|
[Advances in the Study of Tissue-resident Memory T Cells in Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:862-869. [PMID: 36617472 PMCID: PMC9845087 DOI: 10.3779/j.issn.1009-3419.2022.102.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have been widely used in the treatment of lung cancer, but the benefit population is limited and there is a lack of effective predictive markers of efficacy. Tissue-resident memory T cells (TRM) reside in tissues and exert anti-tumor effects by expressing the integrins CD103, CD49a or C-type lectin CD69 and immune checkpoint receptors. TRM expressing programmed cell death 1 (PD-1) is enriched with transcriptional products associated with cytotoxicity and enhances T cell (antigen) receptor (TCR)-mediated cytotoxicity. TRM is a promising biomarker for predicting the efficacy and prognosis of immunotherapy in lung cancer patients. This review will describe the progress of TRM research in lung cancer.
.
Collapse
|
12
|
Franklin M, Connolly E, Hussell T. Recruited and Tissue-Resident Natural Killer Cells in the Lung During Infection and Cancer. Front Immunol 2022; 13:887503. [PMID: 35844626 PMCID: PMC9284027 DOI: 10.3389/fimmu.2022.887503] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/17/2022] [Indexed: 11/23/2022] Open
Abstract
Natural killer (NK) cells are an important component of the innate immune system, and have a key role in host defense against infection and in tumor surveillance. Tumors and viruses employ remarkably similar strategies to avoid recognition and killing by NK cells and so much can be learnt by comparing NK cells in these disparate diseases. The lung is a unique tissue environment and immune cells in this organ, including NK cells, exist in a hypofunctional state to prevent activation against innocuous stimuli. Upon infection, rapid NK cell infiltration into the lung occurs, the amplitude of which is determined by the extent of inflammation and damage. Activated NK cells kill infected cells and produce pro-inflammatory cytokines and chemokines to recruit cells of the adaptive immune system. More recent evidence has shown that NK cells also play an additional role in resolution of inflammation. In lung cancer however, NK cell recruitment is impaired and those that are present have reduced functionality. The majority of lung NK cells are circulatory, however recently a small population of tissue-resident lung NK cells has been described. The specific role of this subset is yet to be determined, but they show similarity to resident memory T cell subsets. Whether resident or recruited, NK cells are important in the control of pulmonary infections, but equally, can drive excessive inflammation if not regulated. In this review we discuss how NK cells are recruited, controlled and retained in the specific environment of the lung in health and disease. Understanding these mechanisms in the context of infection may provide opportunities to promote NK cell recruitment and function in the lung tumor setting.
Collapse
|
13
|
Watanabe R, Hashimoto M. Vasculitogenic T Cells in Large Vessel Vasculitis. Front Immunol 2022; 13:923582. [PMID: 35784327 PMCID: PMC9240193 DOI: 10.3389/fimmu.2022.923582] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Vasculitis is an autoimmune disease of unknown etiology that causes inflammation of the blood vessels. Large vessel vasculitis is classified as either giant cell arteritis (GCA), which occurs exclusively in the elderly, or Takayasu arteritis (TAK), which mainly affects young women. Various cell types are involved in the pathogenesis of large vessel vasculitis. Among these, dendritic cells located between the adventitia and the media initiate the inflammatory cascade as antigen-presenting cells, followed by activation of macrophages and T cells contributing to vessel wall destruction. In both diseases, naive CD4+ T cells are polarized to differentiate into Th1 or Th17 cells, whereas differentiation into regulatory T cells, which suppress vascular inflammation, is inhibited. Skewed T cell differentiation is the result of aberrant intracellular signaling, such as the mechanistic target of rapamycin (mTOR) or the Janus kinase signal transducer and activator of transcription (JAK-STAT) pathways. It has also become clear that tissue niches in the vasculature fuel activated T cells and maintain tissue-resident memory T cells. In this review, we outline the most recent understanding of the pathophysiology of large vessel vasculitis. Then, we provide a summary of skewed T cell differentiation in the vasculature and peripheral blood. Finally, new therapeutic strategies for correcting skewed T cell differentiation as well as aberrant intracellular signaling are discussed.
Collapse
|
14
|
PLGA particle vaccination elicits resident memory CD8 T cells protecting from tumors and infection. Eur J Pharm Sci 2022; 175:106209. [DOI: 10.1016/j.ejps.2022.106209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/02/2022] [Accepted: 05/12/2022] [Indexed: 11/20/2022]
|
15
|
Mei X, Li H, Zhou X, Cheng M, Cui K. The Emerging Role of Tissue-Resident Memory CD8 + T Lymphocytes in Human Digestive Tract Cancers. Front Oncol 2022; 11:819505. [PMID: 35096624 PMCID: PMC8795735 DOI: 10.3389/fonc.2021.819505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/23/2021] [Indexed: 12/15/2022] Open
Abstract
Malignant digestive tract tumors are a great threat to human public health. In addition to surgery, immunotherapy brings hope for the treatment of these tumors. Tissue-resident memory CD8+ T (Trm) cells are a focus of tumor immunology research and treatment due to their powerful cytotoxic effects, ability to directly kill epithelial-derived tumor cells, and overall impact on maintaining mucosal homeostasis and antitumor function in the digestive tract. They are a group of noncirculating immune cells expressing adhesion and migration molecules such as CD69, CD103, and CD49a that primarily reside on the barrier epithelium of nonlymphoid organs and respond rapidly to both viral and bacterial infection and tumorigenesis. This review highlights new research exploring the role of CD8+ Trm cells in a variety of digestive tract malignant tumors, including esophageal cancer, gastric cancer, colorectal cancer, and hepatocellular carcinoma. A summary of CD8+ Trm cell phenotypes and characteristics, tissue distribution, and antitumor functions in different tumor environments is provided, illustrating how these cells may be used in immunotherapies against digestive tract tumors.
Collapse
Affiliation(s)
- Xinyu Mei
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huan Li
- Department of Thoracic Surgery, Anhui Provincial Hospital Affiliated With Anhui Medical University, Hefei, China
| | - Xinpeng Zhou
- Department of Thoracic Surgery, Anhui Provincial Hospital, Wannan Medical College, Hefei, China
| | - Min Cheng
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China.,Cancer Immunotherapy Center, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Kele Cui
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China.,Cancer Immunotherapy Center, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Department of Clinical Laboratory, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
16
|
Krüppel-like Factor 2 (KLF2) in Immune Cell Migration. Vaccines (Basel) 2021; 9:vaccines9101171. [PMID: 34696279 PMCID: PMC8539188 DOI: 10.3390/vaccines9101171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/27/2021] [Accepted: 10/06/2021] [Indexed: 01/30/2023] Open
Abstract
Krüppel-like factor 2 (KLF2), a transcription factor of the krüppel-like family, is a key regulator of activation, differentiation, and migration processes in various cell types. In this review, we focus on the functional relevance of KLF2 in immune cell migration and homing. We summarize the key functions of KLF2 in the regulation of chemokine receptors and adhesion molecules and discuss the relevance of the KLF2-mediated control of immune cell migration in the context of immune responses, infections, and diseases.
Collapse
|
17
|
Wu B, Zhang G, Guo Z, Wang G, Xu X, Li JL, Whitmire JK, Zheng J, Wan YY. The SKI proto-oncogene restrains the resident CD103 +CD8 + T cell response in viral clearance. Cell Mol Immunol 2021; 18:2410-2421. [PMID: 32612153 PMCID: PMC8484360 DOI: 10.1038/s41423-020-0495-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023] Open
Abstract
Acute viral infection causes illness and death. In addition, an infection often results in increased susceptibility to a secondary infection, but the mechanisms behind this susceptibility are poorly understood. Since its initial identification as a marker for resident memory CD8+ T cells in barrier tissues, the function and regulation of CD103 integrin (encoded by ITGAE gene) have been extensively investigated. Nonetheless, the function and regulation of the resident CD103+CD8+ T cell response to acute viral infection remain unclear. Although TGFβ signaling is essential for CD103 expression, the precise molecular mechanism behind this regulation is elusive. Here, we reveal a TGFβ-SKI-Smad4 pathway that critically and specifically directs resident CD103+CD8+ T cell generation for protective immunity against primary and secondary viral infection. We found that resident CD103+CD8+ T cells are abundant in both lymphoid and nonlymphoid tissues from uninfected mice. CD103 acts as a costimulation signal to produce an optimal antigenic CD8+ T cell response to acute viral infection. There is a reduction in resident CD103+CD8+ T cells following primary infection that results in increased susceptibility of the host to secondary infection. Intriguingly, CD103 expression inversely and specifically correlates with SKI proto-oncogene (SKI) expression but not R-Smad2/3 activation. Ectopic expression of SKI restricts CD103 expression in CD8+ T cells in vitro and in vivo to hamper viral clearance. Mechanistically, SKI is recruited to the Itgae loci to directly suppress CD103 transcription by regulating histone acetylation in a Smad4-dependent manner. Our study therefore reveals that resident CD103+CD8+ T cells dictate protective immunity during primary and secondary infection. Interfering with SKI function may amplify the resident CD103+CD8+ T cell response to promote protective immunity.
Collapse
Affiliation(s)
- Bing Wu
- grid.10698.360000000122483208Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ,grid.10698.360000000122483208Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Ge Zhang
- grid.10698.360000000122483208Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ,grid.10698.360000000122483208Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ,grid.411971.b0000 0000 9558 1426Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning 116044 China
| | - Zengli Guo
- grid.10698.360000000122483208Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ,grid.10698.360000000122483208Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Gang Wang
- grid.10698.360000000122483208Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ,grid.10698.360000000122483208Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ,grid.417303.20000 0000 9927 0537Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002 China
| | - Xiaojiang Xu
- grid.280664.e0000 0001 2110 5790Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, Chapel Hill, NC 27709 USA
| | - Jian-liang Li
- grid.280664.e0000 0001 2110 5790Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, Chapel Hill, NC 27709 USA
| | - Jason K. Whitmire
- grid.10698.360000000122483208Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ,grid.10698.360000000122483208Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ,grid.10698.360000000122483208Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Junnian Zheng
- grid.417303.20000 0000 9927 0537Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002 China
| | - Yisong Y. Wan
- grid.10698.360000000122483208Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ,grid.10698.360000000122483208Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| |
Collapse
|
18
|
Bacillus subtilis Spore-Trained Dendritic Cells Enhance the Generation of Memory T Cells via ICAM1. Cells 2021; 10:cells10092267. [PMID: 34571913 PMCID: PMC8469252 DOI: 10.3390/cells10092267] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 11/22/2022] Open
Abstract
Immunological memory is a cardinal feature of the immune system. The intestinal mucosa is the primary exposure and entry site of infectious organisms. For an effective and long-lasting safeguard, a robust immune memory system is required, especially by the mucosal immunity. It is well known that tissue-resident memory T cells (Trms) provide a first response against infections reencountered at mucosal tissues surfaces, where they accelerate pathogen clearance. However, their function in intestinal immunization remains to be investigated. Here, we report enhanced local mucosal and systemic immune responses through oral administration of H9N2 influenza whole inactivated virus (H9N2 WIV) plus Bacillus subtilis spores. Subsequently, H9N2 WIV plus spores led to the generation of CD103+ CD69+ Trms, which were independent of circulating T cells during the immune period. Meanwhile, we also found that Bacillus subtilis spores could stimulate Acrp30 expression in 3T3-L1 adipocytes. Moreover, spore-stimulated adipocyte supernatant also upregulated the expression of intercellular adhesion molecule-1 (ICAM1) in dendritic cells (DCs). Furthermore, the proportion of HA-tetramer+ cells was severely curtailed upon suppressed ICAM1 expression, which also depended on HA-loaded DCs. Taken together, our data demonstrated that spore-promoted H9N2 WIV induced an immune response by enhancing Trms populations, which were associated with the activation of ICAM1 in DCs.
Collapse
|
19
|
Ochsner SP, Li W, Rajendrakumar AM, Palaniyandi S, Acharya G, Liu X, Wang G, Krammer F, Shi M, Tuo W, Pauza CD, Zhu X. FcRn-Targeted Mucosal Vaccination against Influenza Virus Infection. THE JOURNAL OF IMMUNOLOGY 2021; 207:1310-1321. [PMID: 34380652 DOI: 10.4049/jimmunol.2100297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/02/2021] [Indexed: 11/19/2022]
Abstract
The respiratory tract is constantly exposed to various airborne pathogens. Most vaccines against respiratory infections are designed for the parenteral routes of administration; consequently, they provide relatively minimal protection in the respiratory tract. A vaccination strategy that aims to induce the protective mucosal immune responses in the airway is urgently needed. The FcRn mediates IgG Ab transport across the epithelial cells lining the respiratory tract. By mimicking this natural IgG transfer, we tested whether FcRn delivers vaccine Ags to induce a protective immunity to respiratory infections. In this study, we designed a monomeric IgG Fc fused to influenza virus hemagglutinin (HA) Ag with a trimerization domain. The soluble trimeric HA-Fc were characterized by their binding with conformation-dependent HA Abs or FcRn. In wild-type, but not FcRn knockout, mice, intranasal immunization with HA-Fc plus CpG adjuvant conferred significant protection against lethal intranasal challenge with influenza A/PR/8/34 virus. Further, mice immunized with a mutant HA-Fc lacking FcRn binding sites or HA alone succumbed to lethal infection. Protection was attributed to high levels of neutralizing Abs, robust and long-lasting B and T cell responses, the presence of lung-resident memory T cells and bone marrow plasma cells, and a remarkable reduction of virus-induced lung inflammation. Our results demonstrate for the first time, to our knowledge, that FcRn can effectively deliver a trimeric viral vaccine Ag in the respiratory tract and elicit potent protection against respiratory infection. This study further supports a view that FcRn-mediated mucosal immunization is a platform for vaccine delivery against common respiratory pathogens.
Collapse
Affiliation(s)
- Susan Park Ochsner
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD
| | - Weizhong Li
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD
| | - Arunraj Mekhemadhom Rajendrakumar
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD.,Animal Parasitic Diseases Laboratory, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD
| | - Senthilkumar Palaniyandi
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD
| | - Gyanada Acharya
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD
| | - Xiaoyang Liu
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD
| | - Gefei Wang
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY; and
| | - Meiqing Shi
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD
| | - Wenbin Tuo
- Animal Parasitic Diseases Laboratory, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD
| | | | - Xiaoping Zhu
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, MD;
| |
Collapse
|
20
|
The human memory T cell compartment changes across tissues of the female reproductive tract. Mucosal Immunol 2021; 14:862-872. [PMID: 33953338 PMCID: PMC8225572 DOI: 10.1038/s41385-021-00406-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 02/04/2023]
Abstract
Memory CD4 T cells in tissues fulfill numerous functions that are critical for local immune homeostasis and protection against pathogens. Previous studies have highlighted the phenotypic and functional heterogeneity of circulating and tissue-resident memory CD4 T cells across different human tissues such as skin, lung, liver, and colon. Comparatively little is known in regard to memory CD4 T cells across tissues of the female reproductive tract (FRT). We examined CD4 T cells in donor-matched vaginal, ecto- and endocervical tissues, which differ in mucosal structure and exposure to external environmental stimuli. We hypothesized that this could be reflected by tissue-specific differences in the memory CD4 T cell compartment. We found differences in CD4 subset distribution across these tissues. Specifically, CD69+CD103+ CD4 T cells were significantly more abundant in vaginal than cervical tissues. In contrast, the transcriptional profiles of CD4 subsets were fairly conserved across FRT tissues. CD69+CD103+ CD4 T cells showed a TH17 bias independent of tissue niche. Our data suggest that FRT tissues affect T cell subset distribution but have limited effects on the transcriptome of each subset. We discuss the implications for barrier immunity in the FRT.
Collapse
|
21
|
Gu HJ, Song S, Roh JY, Jung Y, Kim HJ. Expression pattern of tissue-resident memory T cells in cutaneous lupus erythematosus. Lupus 2021; 30:1427-1437. [PMID: 34013817 DOI: 10.1177/09612033211017218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Tissue resident memory T cells (TRMs) persist long-term in peripheral tissues without recirculation, triggering an immediate protective inflammatory state upon the re-recognition of the antigen. Despite evidence incriminating the dysregulation of TRMs in autoimmune diseases, few studies have examined their expression in cutaneous lupus erythematosus (CLE). OBJECTIVES We aimed to examine whether there are differences among TRM populations in CLE depending on different clinical conditions, such as the CLE subtype or association with systemic lupus erythematosus, and to determine the effect of type I interferon (IFN) on the development of TRMs in CLE. METHODS CLE disease activity was evaluated using the Cutaneous Lupus Erythematosus Disease Area and Severity Index. The expression of the TRM markers CD69 and CD103 in CLE lesions was evaluated by immunofluorescence. Flow cytometry was performed on peripheral blood mononuclear cells after IFNα treatment. RESULTS The number of TRMs expressing either CD69 or CD103 was significantly higher in CLE lesions than in control skin; however, it was not significantly different between discoid lupus erythematosus and subacute CLE, or dependent on the presence of concomitant systemic lupus. Lesional severity was not correlated with an increase in TRMs in CLE. IFNα treatment induced a conspicuous increase in CD69 expression in skin-homing T cells, more profoundly in CD4+ T cells than in CD8+ T cells. CONCLUSIONS Skin TRMs, either CD69 or CD103-positive cells, showed increased levels in the lesional skin of CLE, and IFNα increased the expression of CD69 in T cells.
Collapse
Affiliation(s)
- Hyeon-Jung Gu
- Department of Health Science and Technology, Gachon Advanced Institute for Health Science & Technology, Gachon University, Incheon, Korea
| | - Shinyoung Song
- Department of Dermatology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Joo Young Roh
- Department of Dermatology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - YunJae Jung
- Department of Health Science and Technology, Gachon Advanced Institute for Health Science & Technology, Gachon University, Incheon, Korea.,Department of Microbiology, Gachon University College of Medicine, Incheon, Korea
| | - Hee Joo Kim
- Department of Dermatology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| |
Collapse
|
22
|
Christian LS, Wang L, Lim B, Deng D, Wu H, Wang XF, Li QJ. Resident memory T cells in tumor-distant tissues fortify against metastasis formation. Cell Rep 2021; 35:109118. [PMID: 33979626 PMCID: PMC8204287 DOI: 10.1016/j.celrep.2021.109118] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 02/15/2021] [Accepted: 04/21/2021] [Indexed: 10/27/2022] Open
Abstract
As a critical machinery for rapid pathogen removal, resident memory T cells (TRMs) are locally generated after the initial encounter. However, their development accompanying tumorigenesis remains elusive. Using a murine breast cancer model, we show that TRMs develop in the tumor, the contralateral mammary mucosa, and the pre-metastatic lung. Single-cell RNA sequencing of TRMs reveals two phenotypically distinct populations representing their active versus quiescent phases. These TRMs in different tissue compartments share the same TCR clonotypes and transcriptomes with a subset of intratumoral effector/effector memory T cells (TEff/EMs), indicating their developmental ontogeny. Furthermore, CXCL16 is highly produced by tumor cells and CXCR6- TEff/EMs are the major subset preferentially egressing the tumor to form distant TRMs. Functionally, releasing CXCR6 retention in the primary tumor amplifies tumor-derived TRMs in the lung and leads to superior protection against metastases. This immunologic fortification suggests a potential strategy to prevent metastasis in clinical oncology.
Collapse
Affiliation(s)
- Laura S Christian
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Bryan Lim
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Dachuan Deng
- TCRCure (TianKeYa) Biopharma, Ltd., Durham, NC 27701, USA
| | - Haiyang Wu
- TCRCure (TianKeYa) Biopharma, Ltd., Durham, NC 27701, USA
| | - Xiao-Fan Wang
- Departments of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Qi-Jing Li
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
23
|
O’Neil TR, Hu K, Truong NR, Arshad S, Shacklett BL, Cunningham AL, Nasr N. The Role of Tissue Resident Memory CD4 T Cells in Herpes Simplex Viral and HIV Infection. Viruses 2021; 13:359. [PMID: 33668777 PMCID: PMC7996247 DOI: 10.3390/v13030359] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/05/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Tissue-resident memory T cells (TRM) were first described in 2009. While initially the major focus was on CD8+ TRM, there has recently been increased interest in defining the phenotype and the role of CD4+ TRM in diseases. Circulating CD4+ T cells seed CD4+ TRM, but there also appears to be an equilibrium between CD4+ TRM and blood CD4+ T cells. CD4+ TRM are more mobile than CD8+ TRM, usually localized deeper within the dermis/lamina propria and yet may exhibit synergy with CD8+ TRM in disease control. This has been demonstrated in herpes simplex infections in mice. In human recurrent herpes infections, both CD4+ and CD8+ TRM persisting between lesions may control asymptomatic shedding through interferon-gamma secretion, although this has been more clearly shown for CD8+ T cells. The exact role of the CD4+/CD8+ TRM axis in the trigeminal ganglia and/or cornea in controlling recurrent herpetic keratitis is unknown. In HIV, CD4+ TRM have now been shown to be a major target for productive and latent infection in the cervix. In HSV and HIV co-infections, CD4+ TRM persisting in the dermis support HIV replication. Further understanding of the role of CD4+ TRM and their induction by vaccines may help control sexual transmission by both viruses.
Collapse
Affiliation(s)
- Thomas R. O’Neil
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (T.R.O.); (K.H.); (N.R.T.); (S.A.)
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Kevin Hu
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (T.R.O.); (K.H.); (N.R.T.); (S.A.)
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Naomi R. Truong
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (T.R.O.); (K.H.); (N.R.T.); (S.A.)
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Sana Arshad
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (T.R.O.); (K.H.); (N.R.T.); (S.A.)
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Barbara L. Shacklett
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA 95616, USA;
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (T.R.O.); (K.H.); (N.R.T.); (S.A.)
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Najla Nasr
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (T.R.O.); (K.H.); (N.R.T.); (S.A.)
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2000, Australia
| |
Collapse
|
24
|
Dijkman K, Aguilo N, Boot C, Hofman SO, Sombroek CC, Vervenne RA, Kocken CH, Marinova D, Thole J, Rodríguez E, Vierboom MP, Haanstra KG, Puentes E, Martin C, Verreck FA. Pulmonary MTBVAC vaccination induces immune signatures previously correlated with prevention of tuberculosis infection. Cell Rep Med 2021; 2:100187. [PMID: 33521701 PMCID: PMC7817873 DOI: 10.1016/j.xcrm.2020.100187] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 10/23/2020] [Accepted: 12/17/2020] [Indexed: 11/29/2022]
Abstract
To fight tuberculosis, better vaccination strategies are needed. Live attenuated Mycobacterium tuberculosis-derived vaccine, MTBVAC, is a promising candidate in the pipeline, proven to be safe and immunogenic in humans so far. Independent studies have shown that pulmonary mucosal delivery of Bacillus Calmette-Guérin (BCG), the only tuberculosis (TB) vaccine available today, confers superior protection over standard intradermal immunization. Here we demonstrate that mucosal MTBVAC is well tolerated, eliciting polyfunctional T helper type 17 cells, interleukin-10, and immunoglobulins in the airway and yielding a broader antigenic profile than BCG in rhesus macaques. Beyond our previous work, we show that local immunoglobulins, induced by MTBVAC and BCG, bind to M. tuberculosis and enhance pathogen uptake. Furthermore, after pulmonary vaccination, but not M. tuberculosis infection, local T cells expressed high levels of mucosal homing and tissue residency markers. Our data show that pulmonary MTBVAC administration has the potential to enhance its efficacy and justifies further exploration of mucosal vaccination strategies in preclinical efficacy studies.
Collapse
Affiliation(s)
- Karin Dijkman
- Biomedical Primate Research Centre (BPRC), Rijswijk, the Netherlands
| | - Nacho Aguilo
- Department of Microbiology, Faculty of Medicine, IIS Aragon, University of Zaragoza, Zaragoza, Spain
- CIBERES, Instituto de Salud Carlos III, Madrid, Spain
| | - Charelle Boot
- Biomedical Primate Research Centre (BPRC), Rijswijk, the Netherlands
| | - Sam O. Hofman
- Biomedical Primate Research Centre (BPRC), Rijswijk, the Netherlands
| | | | | | | | - Dessislava Marinova
- Department of Microbiology, Faculty of Medicine, IIS Aragon, University of Zaragoza, Zaragoza, Spain
- CIBERES, Instituto de Salud Carlos III, Madrid, Spain
| | - Jelle Thole
- TuBerculosis Vaccine Initiative (TBVI), Lelystad, the Netherlands
| | | | | | | | | | - Carlos Martin
- Department of Microbiology, Faculty of Medicine, IIS Aragon, University of Zaragoza, Zaragoza, Spain
- CIBERES, Instituto de Salud Carlos III, Madrid, Spain
| | | |
Collapse
|
25
|
Middelburg J, Kemper K, Engelberts P, Labrijn AF, Schuurman J, van Hall T. Overcoming Challenges for CD3-Bispecific Antibody Therapy in Solid Tumors. Cancers (Basel) 2021; 13:287. [PMID: 33466732 PMCID: PMC7829968 DOI: 10.3390/cancers13020287] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy of cancer with CD3-bispecific antibodies is an approved therapeutic option for some hematological malignancies and is under clinical investigation for solid cancers. However, the treatment of solid tumors faces more pronounced hurdles, such as increased on-target off-tumor toxicities, sparse T-cell infiltration and impaired T-cell quality due to the presence of an immunosuppressive tumor microenvironment, which affect the safety and limit efficacy of CD3-bispecific antibody therapy. In this review, we provide a brief status update of the CD3-bispecific antibody therapy field and identify intrinsic hurdles in solid cancers. Furthermore, we describe potential combinatorial approaches to overcome these challenges in order to generate selective and more effective responses.
Collapse
Affiliation(s)
- Jim Middelburg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Kristel Kemper
- Genmab, 3584 CT Utrecht, The Netherlands; (K.K.); (P.E.); (A.F.L.); (J.S.)
| | - Patrick Engelberts
- Genmab, 3584 CT Utrecht, The Netherlands; (K.K.); (P.E.); (A.F.L.); (J.S.)
| | - Aran F. Labrijn
- Genmab, 3584 CT Utrecht, The Netherlands; (K.K.); (P.E.); (A.F.L.); (J.S.)
| | - Janine Schuurman
- Genmab, 3584 CT Utrecht, The Netherlands; (K.K.); (P.E.); (A.F.L.); (J.S.)
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| |
Collapse
|
26
|
Sanjabi S, Lear S. New cytometry tools for immune monitoring during cancer immunotherapy. CYTOMETRY PART B-CLINICAL CYTOMETRY 2021; 100:10-18. [PMID: 33432667 DOI: 10.1002/cyto.b.21984] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/23/2022]
Abstract
The success of cancer immunotherapy (CIT) in the past decade has brought renewed excitement and the need to better understand how the human immune system functions during health and disease. Advances in single cell technologies have also inspired the creation of a Human Cell Atlas to identify and describe every cell in the human body with the intention of elucidating how to "fix" the ones that fail normal function. For example, treatment of cancer patients with immune checkpoint blockade (ICB) antibodies can reinvigorate their T cells and produce durable clinical benefit in a subset of patients, but a number of resistance mechanisms exist that prohibit full benefit to all treated patients. Early detection of biomarkers of response and mechanisms of resistance are needed to identify the patients who can benefit most from ICB. A noninvasive approach to predict treatment outcomes early after immunotherapies is a longitudinal analysis of peripheral blood immune cells using flow cytometry. Here we review some of the advances in our understanding of how ICB antibodies can re-invigorate tumor-specific T cells and also highlight the recent advances in high complexity flow cytometry, including spectral cytometers, that allow longitudinal sampling and deep immune phenotyping in clinical settings. We encourage the scientific community to utilize advanced cytometry platforms and analyses for immune monitoring in order to optimize CIT treatments for maximum clinical benefit.
Collapse
Affiliation(s)
- Shomyseh Sanjabi
- Department of Oncology Biomarker Development, Genentech Developmental Sciences, South San Francisco, California, USA
| | - Sean Lear
- Department of OMNI Biomarker Development, Genentech Developmental Sciences, South San Francisco, California, USA
| |
Collapse
|
27
|
Liao W, Liu Y, Ma C, Wang L, Li G, Mishra S, Srinivasan S, Fan KKH, Wu H, Li Q, Zhao M, Liu X, Demel EL, Zhang X, Qiu Y, Lu Q, Zhang N. The downregulation of IL-18R defines bona fide kidney-resident CD8 + T cells. iScience 2021; 24:101975. [PMID: 33474536 PMCID: PMC7803637 DOI: 10.1016/j.isci.2020.101975] [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: 07/24/2020] [Revised: 12/01/2020] [Accepted: 12/17/2020] [Indexed: 11/05/2022] Open
Abstract
Stepwise induction of CD69 and CD103 marks distinct differentiation stages of mucosal Trms. But the majority of non-mucosal Trm lacks CD103 expression. The expression of CD69 alone cannot faithfully define Trm cells in heavily vascularized non-mucosal tissues, such as the kidney. Here, we found that a subset of kidney Trms downregulated IL-18 receptor during differentiation. Via global transcriptional analysis and parabiosis experiments, we have discovered that the downregulation of interleukin-18 receptor (IL-18R) is associated with the establishment of tissue residency. Together with the expression of CD69, IL-18Rlo exclusively identify tissue-resident cells whereas IL-18Rhi population contains both tissue-resident and migratory ones. Local cytokines including transforming growth factor β (TGF-β) and interferon α (IFN-α)/β as well as TGF-β-dependent suppression of transcription factor Tcf-1 are essential for IL-18R downregulation during kidney Trm differentiation. Together, we identified a convenient surface marker to distinguish bona fide kidney-resident CD8+ T cells as well as underlying molecular mechanisms controlling this differentiation process. CD8+ Trm cells downregulate IL-18 receptor during differentiation IL-18Rhi population is composed of both migratory and resident subsets IL-18Rlo population is exclusively tissue-resident TGF-β promotes, whereas IFN-α/β inhibits, IL-18R downregulation
Collapse
Affiliation(s)
- Wei Liao
- Department of Microbiology, Immunology and Molecular Genetics, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.,Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yong Liu
- Department of Microbiology, Immunology and Molecular Genetics, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.,Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Liwen Wang
- Department of Microbiology, Immunology and Molecular Genetics, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.,Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Guo Li
- Department of Microbiology, Immunology and Molecular Genetics, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.,Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Shruti Mishra
- Department of Microbiology, Immunology and Molecular Genetics, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Saranya Srinivasan
- Department of Microbiology, Immunology and Molecular Genetics, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Kenneth Ka-Ho Fan
- Department of Microbiology, Immunology and Molecular Genetics, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qianwen Li
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xun Liu
- Department of Microbiology, Immunology and Molecular Genetics, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Erika L Demel
- Department of Microbiology, Immunology and Molecular Genetics, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Xin Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Yuanzheng Qiu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China.,Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, Joe R. & Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
28
|
Nakamura Y, Zhenjie Z, Oya K, Tanaka R, Ishitsuka Y, Okiyama N, Watanabe R, Fujisawa Y. Poor Lymphocyte Infiltration to Primary Tumors in Acral Lentiginous Melanoma and Mucosal Melanoma Compared to Cutaneous Melanoma. Front Oncol 2020; 10:524700. [PMID: 33392063 PMCID: PMC7773936 DOI: 10.3389/fonc.2020.524700] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Recent clinical trials have demonstrated the efficacy of immune checkpoint inhibitors (ICIs) for treating melanoma. However, these previous studies comprised mainly Caucasian populations, in which cutaneous melanoma (CM) is the major clinical type. In contrast, Asian populations have a distinct profile of melanoma and show much higher frequencies of acral lentiginous melanoma (ALM) and mucosal melanoma (MCM). Compared with CM, ALM and MCM show poorer response to ICIs, but the mechanisms have not been fully understood. To evaluate the immune status in each melanoma subtype, we examined the number of total tumor-infiltrating lymphocytes (TILs), CD4+ TILs, CD8+ TILs, and tumor-infiltrating FoxP3+ regulatory T cells (Tregs) to evaluate the immune status in each melanoma subtype using data from 137 patients with melanoma. Total TIL numbers in ALM and MCM were significantly lower than that in CM. CD4+ TIL number in MCM was also lower than CM although CD4+ TIL number in ALM was comparable with CM. In contrast, CD8+ TIL numbers in both ALM and MCM were significantly lower than that in CM. Although number of tumor-infiltrating Tregs was comparable among the 3 subtypes, the proportion of tumor-infiltrating Tregs in CD4+ T cells in MCM was significantly higher than in CM and ALM. Multivariate regression analysis revealed that ALM and MCM were significantly associated with a lower total TIL number, but only MCM was significantly associated with a lower CD4+ TIL number. Multivariate regression analysis also revealed that both ALM and MCM were significantly associated with a lower CD8+ TIL number. Our results suggest that both ALM and MCM are independent factors of lower total TIL number, which may be associated with poorer responses to ICIs in ALM and MCM.
Collapse
Affiliation(s)
- Yoshiyuki Nakamura
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Zhu Zhenjie
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kazumasa Oya
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Ryota Tanaka
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yosuke Ishitsuka
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Naoko Okiyama
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Rei Watanabe
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yasuhiro Fujisawa
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
29
|
He Q, Jiang L, Cao K, Zhang L, Xie X, Zhang S, Ding X, He Y, Zhang M, Qiu T, Jin X, Zhao C, Zhang X, Xu J. A Systemic Prime-Intrarectal Pull Strategy Raises Rectum-Resident CD8+ T Cells for Effective Protection in a Murine Model of LM-OVA Infection. Front Immunol 2020; 11:571248. [PMID: 33072113 PMCID: PMC7541937 DOI: 10.3389/fimmu.2020.571248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/18/2020] [Indexed: 01/01/2023] Open
Abstract
As the entry sites of many pathogens such as human immunodeficiency virus (HIV), mucosal sites are defended by rapidly reacting resident memory T cells (TRM). TRMs represent a special subpopulation of memory T cells that persist long term in non-lymphoid sites without entering the circulation and provide the “sensing and alarming” role in the first-line defense against infection. The rectum and vagina are the two primary mucosal portals for HIV entry. However, compared to vaginal TRM, rectal TRM is poorly understood. Herein, we investigated the optimal vaccination strategy to induce rectal TRM. We identified an intranasal prime–intrarectal boost (pull) strategy that is effective in engaging rectal TRM alongside circulating memory T cells and demonstrated its protective efficacy in mice against infection of Listeria monocytogenes. On the contrary, the same vaccine delivered via either intranasal or intrarectal route failed to raise rectal TRM, setting it apart from vaginal TRM, which can be induced by both intranasal and intrarectal immunizations. Moreover, intramuscular prime was also effective in inducing rectal TRM in combination with intrarectal pull, highlighting the need of a primed systemic T cell response. A comparison of different pull modalities led to the identification that raising rectal TRM is mainly driven by local antigen presence. We further demonstrated the interval between prime and boost steps to be critical for the induction of rectal TRM, revealing circulating recently activated CD8+ T cells as the likely primary pullable precursor of rectal TRM. Altogether, our studies lay a new framework for harnessing rectal TRM in vaccine development.
Collapse
Affiliation(s)
- Qian He
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lang Jiang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kangli Cao
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Linxia Zhang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinci Xie
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuye Zhang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiangqing Ding
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yongquan He
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Miaomiao Zhang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tianyi Qiu
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xuanxuan Jin
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Zhao
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianqing Xu
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Pappalardo JL, Zhang L, Pecsok MK, Perlman K, Zografou C, Raddassi K, Abulaban A, Krishnaswamy S, Antel J, van Dijk D, Hafler DA. Transcriptomic and clonal characterization of T cells in the human central nervous system. Sci Immunol 2020; 5:eabb8786. [PMID: 32948672 PMCID: PMC8567322 DOI: 10.1126/sciimmunol.abb8786] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/26/2020] [Indexed: 08/04/2023]
Abstract
T cells provide critical immune surveillance to the central nervous system (CNS), and the cerebrospinal fluid (CSF) is thought to be a main route for their entry. Further characterization of the state of T cells in the CSF in healthy individuals is important for understanding how T cells provide protective immune surveillance without damaging the delicate environment of the CNS and providing tissue-specific context for understanding immune dysfunction in neuroinflammatory disease. Here, we have profiled T cells in the CSF of healthy human donors and have identified signatures related to cytotoxic capacity and tissue adaptation that are further exemplified in clonally expanded CSF T cells. By comparing profiles of clonally expanded T cells obtained from the CSF of patients with multiple sclerosis (MS) and healthy donors, we report that clonally expanded T cells from the CSF of patients with MS have heightened expression of genes related to T cell activation and cytotoxicity.
Collapse
Affiliation(s)
- Jenna L Pappalardo
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Le Zhang
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Maggie K Pecsok
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Kelly Perlman
- Montreal Neurologic Institute, Montreal, Quebec, Canada
| | - Chrysoula Zografou
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Khadir Raddassi
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Ahmad Abulaban
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Smita Krishnaswamy
- Departments of Genetics and Computer Science, Yale School of Medicine, New Haven, CT 06511, USA
| | - Jack Antel
- Montreal Neurologic Institute, Montreal, Quebec, Canada
| | - David van Dijk
- Departments of Internal Medicine (Cardiology), Cardiovascular Research Center, and Computer Science, New Haven, CT 06511, USA.
| | - David A Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06511, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
31
|
Zottnick S, Voß AL, Riemer AB. Inducing Immunity Where It Matters: Orthotopic HPV Tumor Models and Therapeutic Vaccinations. Front Immunol 2020; 11:1750. [PMID: 32922389 PMCID: PMC7457000 DOI: 10.3389/fimmu.2020.01750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/30/2020] [Indexed: 12/24/2022] Open
Abstract
Anogenital and oropharyngeal cancers caused by human papillomavirus (HPV) infections account for 4.5% of all cancer cases worldwide. So far, only the initial infection with selected high-risk types can be prevented by prophylactic vaccination. Already existing persistent HPV infections, however, can currently only be treated by surgical removal of resulting lesions. Therapeutic HPV vaccination, promoting cell-based anti-HPV immunity, would be ideal to eliminate and protect against HPV-induced lesions and tumors. A multitude of vaccination approaches has been tested to date, many of which led to high amounts of HPV-specific T cells in vivo. However, growing evidence suggests that not the induction of systemic but of local immunity is paramount for tackling mucosal infections and tumors. Therefore, recent therapeutic vaccination studies have focused on how to induce tissue-resident T cells in the anogenital and oropharyngeal mucosa. These approaches include direct mucosal vaccinations and influencing the migration of systemic T cells toward the mucosa. The efficacy of these new vaccination approaches is best tested in vivo by utilizing orthotopic tumor models, i.e. HPV-positive tumors being located in the animal's mucosa. In line with this, we here review existing HPV tumor models and describe two novel tumorigenic cell lines for the MHC-humanized mouse model A2.DR1. These were used for the establishment of an HPV16 E6/E7-positive vaginal tumor model, suitable for testing therapeutic vaccines containing HLA-A2-restricted HPV16-derived epitopes. The newly developed MHC-humanized orthotopic HPV16-positive tumor model is likely to improve the translatability of in vivo findings to the clinical setting.
Collapse
Affiliation(s)
- Samantha Zottnick
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Alessa L Voß
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Angelika B Riemer
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| |
Collapse
|
32
|
Chu KL, Batista NV, Girard M, Watts TH. Monocyte-Derived Cells in Tissue-Resident Memory T Cell Formation. THE JOURNAL OF IMMUNOLOGY 2020; 204:477-485. [PMID: 31964721 DOI: 10.4049/jimmunol.1901046] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022]
Abstract
There is currently much interest in how different dendritic cell and macrophage populations contribute to T cell-mediated immunity. Although conventional dendritic cell subsets have received much attention for their role in T cell priming, there is emerging evidence for a role for monocyte-derived APC (MoAPC) in tissue-resident memory T cell (Trm) formation. Cells of the monocyte/macrophage lineage play a key role in providing chemokines and cytokines for the localization, differentiation, and survival of Trm and Trm precursors. In addition, inflammatory MoAPC are the key providers of TNF superfamily costimulatory signals, a signal we refer to as signal 4 for T cell activation. Recent evidence suggests that signal 4 from MoAPC occurs postpriming and substantially increases Trm formation. Key questions remain, such as the Ag dependence of signal 4 and the specific mechanisms by which MoAPC-Trm interactions affect the long-term maintenance of Trm.
Collapse
Affiliation(s)
- Kuan-Lun Chu
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Nathália V Batista
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Mélanie Girard
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tania H Watts
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
33
|
Borges da Silva H, Peng C, Wang H, Wanhainen KM, Ma C, Lopez S, Khoruts A, Zhang N, Jameson SC. Sensing of ATP via the Purinergic Receptor P2RX7 Promotes CD8 + Trm Cell Generation by Enhancing Their Sensitivity to the Cytokine TGF-β. Immunity 2020; 53:158-171.e6. [PMID: 32640257 DOI: 10.1016/j.immuni.2020.06.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 05/01/2020] [Accepted: 06/11/2020] [Indexed: 12/14/2022]
Abstract
Tissue-resident memory (Trm) CD8+ T cells mediate protective immunity in barrier tissues, but the cues promoting Trm cell generation are poorly understood. Sensing of extracellular adenosine triphosphate (eATP) by the purinergic receptor P2RX7 is needed for recirculating CD8+ T cell memory, but its role for Trm cells is unclear. Here we showed that P2RX7 supported Trm cell generation by enhancing CD8+ T cell sensing of TGF-β, which was necessary for tissue residency. P2RX7-deficient Trm cells progressively decayed in non-lymphoid tissues and expressed dysregulated Trm-specific markers. P2RX7 was required for efficient re-expression of the receptor TGF-βRII through calcineurin signaling. Forced Tgfbr2 expression rescued P2RX7-deficient Trm cell generation, and TGF-β sensitivity was dictated by P2RX7 agonists and antagonists. Forced Tgfbr2 also rescued P2RX7-deficient Trm cell mitochondrial function. Sustained P2RX7 signaling was required for long-term Trm cell maintenance, indicating that P2RX7 signaling drives induction and CD8+ T cell durability in barrier sites.
Collapse
Affiliation(s)
- Henrique Borges da Silva
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Changwei Peng
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Haiguang Wang
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kelsey M Wanhainen
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Sharon Lopez
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alexander Khoruts
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Stephen C Jameson
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
34
|
Liu L, Huang X, Xu C, Chen C, Zhao W, Li D, Li L, Wang L, Du M. Decidual CD8 +T cells exhibit both residency and tolerance signatures modulated by decidual stromal cells. J Transl Med 2020; 18:221. [PMID: 32487187 PMCID: PMC7268777 DOI: 10.1186/s12967-020-02371-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 05/12/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND During early pregnancy, tolerance of the semi-allogeneic fetus necessitates comprehensive modifications of the maternal immune system. How decidual CD8+T (CD8+dT) cells balance maternal tolerance of the fetus with defense from invading pathogens remains undefined. METHODS We investigated the distribution patterns of CD8+T cells and their heterogeneity in paired peripheral blood and decidual tissue in the first trimester of pregnancy using flow cytometry and mRNA-Seq. Gene Set Enrichment Analysis was utilized to determine the transcriptional features of CD8+dT cells. Moreover, we examined activation of T cells when they were cocultured with trophoblasts, in addition to the effect of the fetal-maternal environment on peripheral CD8+T (CD8+pT) cells. RESULTS We found that, compared with CD8+pT cells, CD8+dT cells consisted mainly of effector memory cells (TEM) and terminally differentiated effector memory cells (TEMRA). Both TEM and TEMRA subsets contained increased numbers of CD27+CD28- cells, which have been shown to possess only partial effector functions. In-depth analysis of the gene-expression profiles of CD8+dT cells revealed significant enrichment in T cell exhaustion-related genes and core tissue residency signature genes that have been found recently to be shared by tissue resident memory cells and tumor-infiltrating lymphocytes (TILs). In accordance with gene expression, protein levels of the exhaustion-related molecules PD-1 and CD39 and the tissue resident molecules CD103 and CXCR3 were increased significantly with almost no perforin secretion in CD8+dT cells compared with CD8+pT cells. However, the levels of granzyme B, IFN-γ, and IL-4 in CD8+dT cells were increased significantly compared with those in CD8+pT cells. Both CD8+dT and CD8+pT cells were not activated after being cocultured with autologous trophoblast cells. Moreover, the production of granzyme B in CD103+CD8+dT cells decreased significantly compared with that in their CD103- counterparts. Coculture with decidual stromal cells and trophoblasts upregulated CD103 expression significantly in CD8+pT cells. CONCLUSIONS Our findings indicate that the selective silencing of effector functions of resident CD8+dT cells may favor maternal-fetal tolerance and that the decidual microenvironment plays an important role in promoting the residency of CD8+T cells and their tolerance-defense balance.
Collapse
Affiliation(s)
- Lu Liu
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, People's Republic of China
| | - Xixi Huang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, People's Republic of China
| | - Chunfang Xu
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, People's Republic of China
| | - Chunqin Chen
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, People's Republic of China
| | - Weijie Zhao
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, People's Republic of China
| | - Dajin Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, People's Republic of China
| | - Liping Li
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, People's Republic of China.
| | - Li Wang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, People's Republic of China.
| | - Meirong Du
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, People's Republic of China.
| |
Collapse
|
35
|
Morris SE, Farber DL, Yates AJ. Tissue-Resident Memory T Cells in Mice and Humans: Towards a Quantitative Ecology. THE JOURNAL OF IMMUNOLOGY 2020; 203:2561-2569. [PMID: 31685700 DOI: 10.4049/jimmunol.1900767] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022]
Abstract
In recent years, tissue-resident memory T cells (TRM) have emerged as essential components of immunological memory. Following antigenic challenge, TRM remain in nonlymphoid tissues and defend against re-exposure. Although accumulating evidence suggests important roles for TRM in mediating protective immunity, fundamental aspects of the population biology of TRM remain poorly understood. In this article, we discuss how results from different systems shed light on the ecological dynamics of TRM in mice and humans. We highlight the importance of dissecting processes contributing to TRM maintenance, and how these might vary across phenotypically and spatially heterogeneous subsets. We also discuss how the diversity of TRM communities within specific tissues may evolve under competition and in response to antigenic perturbation. Throughout, we illustrate how mathematical models can clarify inferences obtained from experimental data and help elucidate the homeostatic mechanisms underpinning the ecology of TRM populations.
Collapse
Affiliation(s)
- Sinead E Morris
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
| | - Donna L Farber
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032.,Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032; and.,Department of Surgery, Columbia University Medical Center, New York, NY 10032
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032;
| |
Collapse
|
36
|
Huang X, Liu L, Xu C, Peng X, Li D, Wang L, Du M. Tissue-resident CD8 + T memory cells with unique properties are present in human decidua during early pregnancy. Am J Reprod Immunol 2020; 84:e13254. [PMID: 32329123 DOI: 10.1111/aji.13254] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/13/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022] Open
Abstract
PROBLEM Resident memory T (TRM ) cells reside in the uterus during pregnancy may play an important role in balancing maternal-fetal tolerance with anti-infectious immunity. Although CD8+ TRM and decidual CD8+ T cells have been extensively characterized, the properties of decidual CD8+ TRM (dTRM ) cells remain poorly defined. METHOD OF STUDY We investigated the heterogeneity, phenotypes, and functions of dTRM cells, and compared the proportion of dTRM cells between normal pregnancy and recurrent spontaneous abortion (RSA) using flow cytometry. Moreover, we cocultured peripheral CD8+ T (CD8+ pT) cells with trophoblast, or decidual stomal cells (DSCs) in the presence or absence of anti-TGF-β antibody or TGF-β type I receptor inhibitor to explore the effects of maternal-fetal environment on decidual CD8+ TRM cell formation. RESULTS We found that CD69+ CD103+ TRM cells were abundant in CD8+ dT cells but not in CD4+ dT cells with effector-memory (EM, CD45RA- CCR7- ) phenotypes. The percentage of dTRM cells from RSA patients was significantly higher than that from normal pregnancy. Furthermore, dTRM cells showed increased expressions of chemokine receptors, T-cell exhaustion-related molecules, and produced more anti-inflammatory cytokines and effector cytokines upon stimulation. Moreover, DSCs produced a considerable level of TGF-β and upregulated CD103 expression on CD69+ CD8+ pT cells, which can be significantly reversed by blocking TGF-β receptor. CONCLUSION Our findings demonstrate that TRM cells with unique properties are present in the decidua during human early pregnancy. They possess an enhanced capacity to produce effector cytokines and regulatory molecules, which might be important in the balance between maternal-fetal immune tolerance and the capacity to aggressively respond to infections.
Collapse
Affiliation(s)
- Xixi Huang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Lu Liu
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Chunfang Xu
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiandong Peng
- Shanghai Jiai Genetics & IVF Institute, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Dajin Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Li Wang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Meirong Du
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Department of Obstetrics and Gynecology, School of Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| |
Collapse
|
37
|
Stein JV, Ruef N. Regulation of global CD8 + T-cell positioning by the actomyosin cytoskeleton. Immunol Rev 2020; 289:232-249. [PMID: 30977193 DOI: 10.1111/imr.12759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/04/2019] [Accepted: 02/06/2019] [Indexed: 12/12/2022]
Abstract
CD8+ T cells have evolved as one of the most motile mammalian cell types, designed to continuously scan peptide-major histocompatibility complexes class I on the surfaces of other cells. Chemoattractants and adhesion molecules direct CD8+ T-cell homing to and migration within secondary lymphoid organs, where these cells colocalize with antigen-presenting dendritic cells in confined tissue volumes. CD8+ T-cell activation induces a switch to infiltration of non-lymphoid tissue (NLT), which differ in their topology and biophysical properties from lymphoid tissue. Here, we provide a short overview on regulation of organism-wide trafficking patterns during naive T-cell recirculation and their switch to non-lymphoid tissue homing during activation. The migratory lifestyle of CD8+ T cells is regulated by their actomyosin cytoskeleton, which translates chemical signals from surface receptors into mechanical work. We explore how properties of the actomyosin cytoskeleton and its regulators affect CD8+ T cell function in lymphoid and non-lymphoid tissue, combining recent findings in the field of cell migration and actin network regulation with tissue anatomy. Finally, we hypothesize that under certain conditions, intrinsic regulation of actomyosin dynamics may render NLT CD8+ T-cell populations less dependent on input from extrinsic signals during tissue scanning.
Collapse
Affiliation(s)
- Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Nora Ruef
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
38
|
A role for the CCR5-CCL5 interaction in the preferential migration of HSV-2-specific effector cells to the vaginal mucosa upon nasal immunization. Mucosal Immunol 2019; 12:1391-1403. [PMID: 31551493 DOI: 10.1038/s41385-019-0203-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 08/09/2019] [Accepted: 08/25/2019] [Indexed: 02/04/2023]
Abstract
Our current study focused on elucidating the role of specific chemokine-receptor interactions in antigen (Ag)-specific immune cell migration from nasal to genital mucosal tissues. This cellular migration is critical to induce effective Ag-specific immune responses against sexually transmitted genital infections. In this study, nasal immunization with live attenuated HSV-2 TK- induced the upregulation of CCR5 expression in effector immune cells, including CD4+ T cells, in Ag-priming sites and vaginal tissue. The CCR5 ligands CCL3, CCL4, and CCL5 all showed upregulated expression in vaginal tissue; in particular, CCL5 expression was highly enhanced in the stromal cells of vaginal tissue after nasal immunization. Intravaginal blockade of CCL5 by using neutralizing antibody diminished the number of HSV-2-specific effector cells in the vagina. Furthermore, loss of CCR5, a receptor for CCL5, impaired the migration of nasally primed Ag-specific effector cells from the airway to vagina. Effector cells adoptively transferred from CCR5-deficient mice failed to migrate into vaginal tissue, consequently increasing recipient mice's susceptibility to HSV-2 vaginal infection. These results indicate that the CCR5-CCL5 chemokine pathway is required for the migration and retention of nasally primed Ag-specific effector cells in vagina for providing protective immunity against HSV-2 infection.
Collapse
|
39
|
Lai JCY, Cheng WK, Hopkins PD, Komba M, Carlow DA, Dutz JP. Topical Adjuvant Application during Subcutaneous Vaccination Promotes Resident Memory T Cell Generation. THE JOURNAL OF IMMUNOLOGY 2019; 203:2443-2450. [PMID: 31578270 DOI: 10.4049/jimmunol.1900199] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 08/26/2019] [Indexed: 12/18/2022]
Abstract
Skin tissue resident memory T cells (TRM) provide superior protection to a second infection. In this study, we evaluated the use of topical CpG oligodeoxynucleotide (ODN) as adjuvant to generate skin TRM in mice. Topical or s.c. CpG ODN adjuvant administration at the time of a s.c. Ag injection led to an accumulation of CD103- CD8 T cells in the epidermis. However, only mice with CpG ODN administered topically had significant numbers of CD103+ Ag-specific CD8 T cells persisting in the local epidermal skin, enhanced circulating memory cells in the blood, and showed protection from intradermal challenge with melanoma cells. Generation of Ag-specific CD8 T cells was dependent on TLR9 expression on hematopoietic cells and partially dependent on receptor expression on stromal cells. Topical challenge of immunized mice at a distal site led to significant expansion of Ag-specific T cells in the blood and accumulation in the challenged skin. We demonstrate that local and systemic T cell memory can be generated with topical CpG ODN at the time of s.c. immunization, suggesting a new method of enhancing current vaccine formulations to generate tissue TRM.
Collapse
Affiliation(s)
- Jacqueline C Y Lai
- Department of Dermatology and Skin Science, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4E8, Canada; and.,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Wing Ki Cheng
- Department of Dermatology and Skin Science, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4E8, Canada; and.,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Patrick D Hopkins
- Department of Dermatology and Skin Science, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4E8, Canada; and.,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Mitsuhiro Komba
- BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Douglas A Carlow
- Department of Dermatology and Skin Science, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4E8, Canada; and.,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Jan P Dutz
- Department of Dermatology and Skin Science, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4E8, Canada; and .,BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| |
Collapse
|
40
|
Abstract
Spondyloarthritis (SpA) is a term that refers to a group of inflammatory diseases that includes psoriatic arthritis, axial SpA and nonradiographic axial SpA, reactive arthritis, enteropathic arthritis and undifferentiated SpA. The disease subtypes share clinical and immunological features, including joint inflammation (peripheral and axial skeleton); skin, gut and eye manifestations; and the absence of diagnostic autoantibodies (seronegative). The diseases also share genetic factors. The aetiology of SpA is still the subject of research by many groups worldwide. Evidence from genetic, experimental and clinical studies has accumulated to indicate a clear role for the IL-17 pathway in the pathogenesis of SpA. The IL-17 family consists of IL-17A, IL-17B, IL-17C, IL-17D, IL-17E and IL-17F, of which IL-17A is the best studied. IL-17A is a pro-inflammatory cytokine that also has the capacity to promote angiogenesis and osteoclastogenesis. Of the six family members, IL-17A has the strongest homology with IL-17F. In this Review, we discuss how IL-17A and IL-17F and their cellular sources might contribute to the immunopathology of SpA.
Collapse
|
41
|
Abstract
Tissue-resident memory T (TRM) cells have emerged as a major component of T cell biology. Recent investigations have greatly advanced our understanding of TRMs. Common features have been discovered to distinguish memory T cells residing in various mucosal and non-mucosal tissues from their circulating counterparts. Given that most organs and tissues contain a unique microenvironment, local signal-induced tissue-specific features are tightly associated with the differentiation, homeostasis, and protective functions of TRMs. Here, we discuss recent advances in the TRM field with a special emphasis on the interaction between local signals and TRMs in the context of individual tissue environment.
Collapse
Affiliation(s)
- Yong Liu
- Department of Microbiology, Immunology and Molecular Genetics, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229; Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South Univeristy, Changsha, Hunan 410008, China
| | - Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229; The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
42
|
Prognostic significance of resident CD103 +CD8 +T cells in human colorectal cancer tissues. Acta Histochem 2019; 121:657-663. [PMID: 31153587 DOI: 10.1016/j.acthis.2019.05.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 05/07/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023]
Abstract
The prognostic significance and clinical implications of resident CD103+CD8+T cells in human colorectal cancer tissues still remains largely unexplored. In our present study, we aimed to characterize the resident CD8+T cells in human colorectal cancer tissues by using double staining of CD103 and CD8, and further evaluated the prognostic significance of resident CD8+T cells in colorectal cancer. We found that the OS rate of the colorectal cancer patients with higher infiltration of CD8+T cells, or with higher numbers of resident CD103+CD8+T cells, or with higher ratio of CD103+CD8+T cells over total CD8+T cells in cancer tissues was significantly better than that of the patients with lower infiltration of CD8+T cells, or with lower numbers of resident CD103+CD8+T cells, or with higher ratio of CD103+CD8+T cells over total CD8+T cells in cancer tissues, respectively. Moreover, higher infiltration of CD8+T cells in colorectal cancer tissues was significantly and inversely correlated with advanced TNM stage. Higher numbers of resident CD103+CD8+T cells in colorectal cancer tissues were significantly and inversely correlated with distant metastasis status. Higher ratio of CD103+CD8+T cells over total CD8+T cells in colorectal cancer tissues was significantly and inversely correlated with age status. The COX model analysis demonstrated that higher infiltration of CD8+T cells, higher numbers of resident CD103+CD8+T cells, or higher ratio of CD103+CD8+T cells over total CD8+T cells in colorectal cancer tissues, could serve as independent prognostic predictors for colorectal cancer patients. Taken together, our present study demonstrated the density of tumor infiltrating CD8+T cells or the numbers of resident CD103+CD8+T cells in colorectal tissues could be used as an important prognostic predictor for this malignancy.
Collapse
|
43
|
Westerhof LM, McGuire K, MacLellan L, Flynn A, Gray JI, Thomas M, Goodyear CS, MacLeod MK. Multifunctional cytokine production reveals functional superiority of memory CD4 T cells. Eur J Immunol 2019; 49:2019-2029. [PMID: 31177549 PMCID: PMC6900100 DOI: 10.1002/eji.201848026] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 04/29/2019] [Accepted: 06/06/2019] [Indexed: 11/20/2022]
Abstract
T cell protective immunity is associated with multifunctional memory cells that produce several different cytokines. Currently, our understanding of when and how these cells are generated is limited. We have used an influenza virus mouse infection model to investigate whether the cytokine profile of memory T cells is reflective of primary responding cells or skewed toward a distinct profile. We found that, in comparison to primary cells, memory T cells tended to make multiple cytokines simultaneously. Analysis of the timings of release of cytokine by influenza virus‐specific T cells, demonstrated that primary responding CD4 T cells from lymphoid organs were unable to produce a sustained cytokine response. In contrast CD8 T cells, memory CD4 T cells, and primary responding CD4 T cells from the lung produced a sustained cytokine response throughout the restimulation period. Moreover, memory CD4 T cells were more resistant than primary responding CD4 T cells to inhibitors that suppress T cell receptor signaling. Together, these data suggest that memory CD4 T cells display superior cytokine responses compared to primary responding cells. These data are key to our ability to identify the cues that drive the generation of protective memory CD4 T cells following infection.
Collapse
Affiliation(s)
- Lotus M Westerhof
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, 120 University Place, University of Glasgow, Glasgow, UK.,GLAZgo Discovery Centre, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Kris McGuire
- GLAZgo Discovery Centre, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Lindsay MacLellan
- GLAZgo Discovery Centre, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Ashley Flynn
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, 120 University Place, University of Glasgow, Glasgow, UK
| | - Joshua I Gray
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, 120 University Place, University of Glasgow, Glasgow, UK
| | - Matthew Thomas
- Respiratory, Inflammation and Autoimmunity IMED, AstraZeneca, Gothenburg, Sweden
| | - Carl S Goodyear
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, 120 University Place, University of Glasgow, Glasgow, UK.,GLAZgo Discovery Centre, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Megan Kl MacLeod
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, 120 University Place, University of Glasgow, Glasgow, UK
| |
Collapse
|
44
|
Xu MM, Murphy PA, Vella AT. Activated T-effector seeds: cultivating atherosclerotic plaque through alternative activation. Am J Physiol Heart Circ Physiol 2019; 316:H1354-H1365. [PMID: 30925075 PMCID: PMC6620674 DOI: 10.1152/ajpheart.00148.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 03/21/2019] [Accepted: 03/25/2019] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a chronic inflammatory pathology that precipitates substantial morbidity and mortality. Although initiated by physiological patterns of low and disturbed flow that differentially prime endothelial cells at sites of vessel branch points and curvature, the chronic, smoldering inflammation of atherosclerosis is accelerated by comorbidities involving inappropriate activation of the adaptive immune system, such as autoimmunity. The innate contributions to atherosclerosis, especially in the transition of monocyte to lipid-laden macrophage, are well established, but the mechanisms underpinning the infiltration, persistence, and effector dynamics of CD8 T cells in particular are not well understood. Adaptive immunity is centered on a classical cascade of antigen recognition and activation, costimulation, and effector cytokine secretion upon recall of antigen. However, chronic inflammation can generate alternative cues that supplant this behavior pattern and promote the retention and activation of peripherally activated T cells. Furthermore, the atherogenic foci that activated immune cell infiltrate are unique lipid-laden environments that offer a diverse array of stimuli, including those of survival, antigen hyporesponsiveness, and inflammatory cytokine expression. This review will focus on how known cardiovascular comorbidities may be influencing CD8 T-cell activation and how, once infiltrated within atherogenic foci, these T cells face a multitude of cues that skew the classical cascade of T-cell behavior, highlighting alternative modes of activation that may help contextualize associations of autoimmunity, viral infection, and immunotherapy with cardiovascular morbidity.
Collapse
Affiliation(s)
- Maria M Xu
- Department of Immunology, School of Medicine, University of Connecticut Health School of Medicine , Farmington, Connecticut
| | - Patrick A Murphy
- Center for Vascular Biology, University of Connecticut Health School of Medicine , Farmington, Connecticut
| | - Anthony T Vella
- Department of Immunology, School of Medicine, University of Connecticut Health School of Medicine , Farmington, Connecticut
| |
Collapse
|
45
|
Killer-like receptors and GPR56 progressive expression defines cytokine production of human CD4 + memory T cells. Nat Commun 2019; 10:2263. [PMID: 31118448 PMCID: PMC6531457 DOI: 10.1038/s41467-019-10018-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 04/10/2019] [Indexed: 12/15/2022] Open
Abstract
All memory T cells mount an accelerated response on antigen reencounter, but significant functional heterogeneity is present within the respective memory T-cell subsets as defined by CCR7 and CD45RA expression, thereby warranting further stratification. Here we show that several surface markers, including KLRB1, KLRG1, GPR56, and KLRF1, help define low, high, or exhausted cytokine producers within human peripheral and intrahepatic CD4+ memory T-cell populations. Highest simultaneous production of TNF and IFN-γ is observed in KLRB1+KLRG1+GPR56+ CD4 T cells. By contrast, KLRF1 expression is associated with T-cell exhaustion and reduced TNF/IFN-γ production. Lastly, TCRβ repertoire analysis and in vitro differentiation support a regulated, progressive expression for these markers during CD4+ memory T-cell differentiation. Our results thus help refine the classification of human memory T cells to provide insights on inflammatory disease progression and immunotherapy development. Despite the current human CD4 memory T cell stratification by CD45RA/CCR7, functional heterogeneities still exist within the respective subsets. Here the authors show that several surface markers, including KLRB1, KLRG1, GPR56 and KLRF1, help to further refine the subsetting of human CD4 memory T cells and provide insights for their differentiation.
Collapse
|
46
|
Liu W, Zeng Z, Luo S, Hu C, Xu N, Huang A, Zheng L, Sundberg EJ, Xi T, Xing Y. Gastric Subserous Vaccination With Helicobacter pylori Vaccine: An Attempt to Establish Tissue-Resident CD4+ Memory T Cells and Induce Prolonged Protection. Front Immunol 2019; 10:1115. [PMID: 31156652 PMCID: PMC6533896 DOI: 10.3389/fimmu.2019.01115] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/01/2019] [Indexed: 12/22/2022] Open
Abstract
Tissue-resident memory T (Trm) cells are enriched at the sites of previous infection and required for enhanced protective immunity. However, the emergence of Trm cells and their roles in providing protection are unclear in the field of Helicobacter pylori (H. pylori) vaccinology. Here, our results suggest that conventional vaccine strategies are unable to establish a measurable antigen (Ag)-specific memory cell pool in stomach; in comparison, gastric subserous injection of mice with micro-dose of Alum-based H. pylori vaccine can induce a pool of local CD4+ Trm cells. Regional recruitment of Ag-specific CD4+ T cells depends on the engagement of Ag and adjuvant-induced inflammation. Prior subcutaneous vaccination enhanced this recruitment. A stable pool of Ag-specific CD4+ T cells can be detected for 240 days. Two weeks of FTY720 administration in immune mice suggests that these cells do not experience the recirculation. Immunohistochemistry results show that close to the vaccination site, abundant CD4+T cells locate on epithelial niches, independent of lymphocyte cluster. Paradigmatically, Ag-specific CD4+ T cells with a phenotype of CD69+CD103- are preferential on lymphocytes isolated from epithelium. Upon Helicobacter infection, CD4+ Trm cells orchestrate a swift recall response with the recruitment of circulating antigen-specific Th1/Th17 cells to trigger a tissue-wide pathogen clearance. This study investigates the vaccine-induced gastric CD4+ Trm cells in a mice model, and highlights the need for designing a vaccine strategy against H. pylori by establishing the protective CD4+ Trm cells.
Collapse
Affiliation(s)
- Wei Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Zhiqin Zeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Shuanghui Luo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Chupeng Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Ningyin Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - An Huang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Eric J Sundberg
- Institute of Human Virology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
47
|
Swaims-Kohlmeier A, Haddad LB, Li ZRT, Brookmeyer KA, Baker JM, Widom CS, Lamousin JC, Chi KH, Chen CY, Kersh EN, Johnson JA, Herbst-Kralovetz MM, Hogben M, Ofotokun I, Kohlmeier JE. Chronic immune barrier dysregulation among women with a history of violence victimization. JCI Insight 2019; 4:126097. [PMID: 31092736 DOI: 10.1172/jci.insight.126097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/11/2019] [Indexed: 11/17/2022] Open
Abstract
We explored the association between violence victimization and increased risk for acquiring sexually transmitted infections (STIs) in women by measuring cellular immune barrier properties from the female reproductive tract. STI-negative participants reporting repeated prior victimization occurrences through the lifetime trauma and victimization history (LTVH) instrument were more likely to exhibit alterations in barrier homeostasis and the composition of critical immune mediators irrespective of demographic parameters or presence of bacterial vaginosis. By combining cellular data with mixed-effect linear modeling, we uncovered differences in local T cells, MHCII+ antigen-presenting cells, and epithelial cells indicative of altered trafficking behavior, increased immunosuppressive function, and decreased barrier integrity at sites of STI exposure that correlate most strongly with LTVH score. These data evidence a biological link between a history of violence victimization and risk of STI acquisition through immune dysregulation in the female reproductive tract.
Collapse
Affiliation(s)
- Alison Swaims-Kohlmeier
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Zheng-Rong Tiger Li
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kathryn A Brookmeyer
- Social and Behavioral Research and Evaluation Branch, Division of STD Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - James M Baker
- Department of Basic Medical Sciences, Department of Obstetrics and Gynecology, The University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Cathy Spatz Widom
- Psychology Department, John Jay College of Criminal Justice, and Graduate Center, City University of New York, New York, New York, USA
| | - James C Lamousin
- Department of Mental Health, South Mississippi State Hospital, and Choices PLLC, Purvis, Mississippi, USA
| | - Kai-Hua Chi
- Laboratory Branch, Division of STD Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Cheng Y Chen
- Laboratory Branch, Division of STD Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Ellen N Kersh
- Laboratory Branch, Division of STD Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jeffrey A Johnson
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Melissa M Herbst-Kralovetz
- Department of Basic Medical Sciences, Department of Obstetrics and Gynecology, The University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Matthew Hogben
- Social and Behavioral Research and Evaluation Branch, Division of STD Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Igho Ofotokun
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jacob E Kohlmeier
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
48
|
Nguyen QP, Deng TZ, Witherden DA, Goldrath AW. Origins of CD4 + circulating and tissue-resident memory T-cells. Immunology 2019; 157:3-12. [PMID: 30897205 PMCID: PMC6459775 DOI: 10.1111/imm.13059] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/08/2019] [Accepted: 03/15/2019] [Indexed: 02/06/2023] Open
Abstract
In response to infection, naive CD4+ T-cells proliferate and differentiate into several possible effector subsets, including conventional T helper effector cells (TH 1, TH 2, TH 17), T regulatory cells (Treg ) and T follicular helper cells (TFH ). Once infection is cleared, a small population of long-lived memory cells remains that mediate immune defenses against reinfection. Memory T lymphocytes have classically been categorized into central memory cell (TCM ) and effector memory cell (TEM ) subsets, both of which circulate between blood, secondary lymphoid organs and in some cases non-lymphoid tissues. A third subset of memory cells, referred to as tissue-resident memory cells (TRM ), resides in tissues without recirculation, serving as 'first line' of defense at barrier sites, such as skin, lung and intestinal mucosa, and augmenting innate immunity in the earliest phases of reinfection and recruiting circulating CD4+ and CD8+ T-cells. The presence of multiple CD4+ T helper subsets has complicated studies of CD4+ memory T-cell differentiation, and the mediators required to support their function. In this review, we summarize recent investigations into the origins of CD4+ memory T-cell populations and discuss studies addressing CD4+ TRM differentiation in barrier tissues.
Collapse
Affiliation(s)
- Quynh P. Nguyen
- Division of Biological SciencesUniversity of California San DiegoLa JollaCAUSA
| | - Tianda Z. Deng
- Division of Biological SciencesUniversity of California San DiegoLa JollaCAUSA
| | | | - Ananda W. Goldrath
- Division of Biological SciencesUniversity of California San DiegoLa JollaCAUSA
| |
Collapse
|
49
|
Takamura S, Kohlmeier JE. Establishment and Maintenance of Conventional and Circulation-Driven Lung-Resident Memory CD8 + T Cells Following Respiratory Virus Infections. Front Immunol 2019; 10:733. [PMID: 31024560 PMCID: PMC6459893 DOI: 10.3389/fimmu.2019.00733] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/19/2019] [Indexed: 12/30/2022] Open
Abstract
Antigen-specific CD8+ tissue-resident memory T cells (TRM cells) persist in the lung following resolution of a respiratory virus infection and provide first-line defense against reinfection. In contrast to other memory T cell populations, such as central memory T cells that circulate between lymph and blood, and effector memory T cells (TEM cells) that circulate between blood and peripheral tissues, TRM cells are best defined by their permanent residency in the tissues and their independence from circulatory T cell populations. Consistent with this, we recently demonstrated that CD8+ TRM cells primarily reside within specific niches in the lung (Repair-Associated Memory Depots; RAMD) that normally exclude CD8+ TEM cells. However, it has also been reported that circulating CD8+ TEM cells continuously convert into CD8+ TRM cells in the lung interstitium, helping to sustain TRM numbers. The relative contributions of these two mechanisms of CD8+ TRM cells maintenance in the lung has been the source of vigorous debate. Here we propose a model in which the majority of CD8+ TRM cells are maintained within RAMD (conventional TRM) whereas a small fraction of TRM are derived from circulating CD8+ TEM cells and maintained in the interstitium. The numbers of both types of TRM cells wane over time due to declines in both RAMD availability and the overall number of TEM in the circulation. This model is consistent with most published reports and has important implications for the development of vaccines designed to elicit protective T cell memory in the lung.
Collapse
Affiliation(s)
- Shiki Takamura
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Jacob E Kohlmeier
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
50
|
Guo R, Zhang T, Meng X, Lin Z, Lin J, Gong Y, Liu X, Yu Y, Zhao G, Ding X, Chen X, Lu L. Lymphocyte mass cytometry identifies a CD3-CD4+ cell subset with a potential role in psoriasis. JCI Insight 2019; 4:125306. [PMID: 30747724 DOI: 10.1172/jci.insight.125306] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 02/05/2019] [Indexed: 02/05/2023] Open
Abstract
Psoriasis (PS) is a systemic, immune-mediated inflammatory disorder. However, the whole lymphocyte compartment and the potential pathologies of PS have not been fully characterized. In the present study, we examined whole lymphocyte subsets and signal transduction proteins using high-dimensional single-cell mass cytometry and a bioinformatics pipeline for an in-depth characterization of the immune cell subsets and protein profiles involved in pathways in the peripheral blood of patients with PS. We identified 15 major immune cell populations in T cell lineages and characterized various CD3+CD4+ Th and CD3+CD8+ T cytotoxic cell populations simultaneously across 24 leukocyte markers and 7 proteins related to the signal transduction pathways. High-dimensional analysis identified 3 new subsets that are abundant in PS peripheral blood, resembling CD3-CD4+ lymphoid tissue inducer cells, Tc17 cells, and CD8+CXCR3+ Tregs. We confirmed the CD3-CD4+ cells, and their features and functions, in an independent PS cohort. The use of single-cell mass cytometry allows systemic-level characterization of lymphocyte subpopulations and dysregulated signaling pathways in the blood of patients with PS, identifying abnormalities of different immune cell subsets. We validated that the CD3-CD4+ cells had elevated OX40 and decreased FRA2 expression, which were positively associated with the PS area and severity index.
Collapse
Affiliation(s)
- Ruru Guo
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Zhang
- Institute for Personalized Medicine, State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyu Meng
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Lin
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinran Lin
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Gong
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | | | - Yuetian Yu
- Department of Critical Care Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guilin Zhao
- Institute for Personalized Medicine, State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xianting Ding
- Institute for Personalized Medicine, State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxiang Chen
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liangjing Lu
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|