51
|
Zhu HX, Yang SH, Gao CY, Bian ZH, Chen XM, Huang RR, Meng QL, Li X, Jin H, Tsuneyama K, Han Y, Li L, Zhao ZB, Gershwin ME, Lian ZX. Targeting pathogenic CD8 + tissue-resident T cells with chimeric antigen receptor therapy in murine autoimmune cholangitis. Nat Commun 2024; 15:2936. [PMID: 38580644 PMCID: PMC10997620 DOI: 10.1038/s41467-024-46654-5] [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: 08/29/2023] [Accepted: 03/01/2024] [Indexed: 04/07/2024] Open
Abstract
Primary biliary cholangitis (PBC) is a cholestatic autoimmune liver disease characterized by autoreactive T cell response against intrahepatic small bile ducts. Here, we use Il12b-/-Il2ra-/- mice (DKO mice) as a model of autoimmune cholangitis and demonstrate that Cd8a knockout or treatment with an anti-CD8α antibody prevents/reduces biliary immunopathology. Using single-cell RNA sequencing analysis, we identified CD8+ tissue-resident memory T (Trm) cells in the livers of DKO mice, which highly express activation- and cytotoxicity-associated markers and induce apoptosis of bile duct epithelial cells. Liver CD8+ Trm cells also upregulate the expression of several immune checkpoint molecules, including PD-1. We describe the development of a chimeric antigen receptor to target PD-1-expressing CD8+ Trm cells. Treatment of DKO mice with PD-1-targeting CAR-T cells selectively depleted liver CD8+ Trm cells and alleviated autoimmune cholangitis. Our work highlights the pathogenic role of CD8+ Trm cells and the potential therapeutic usage of PD-1-targeting CAR-T cells.
Collapse
Affiliation(s)
- Hao-Xian Zhu
- Chronic Disease Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shu-Han Yang
- Chronic Disease Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), School of Medicine, South China University of Technology, Guangzhou, China
| | - Cai-Yue Gao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhen-Hua Bian
- Chronic Disease Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiao-Min Chen
- Chronic Disease Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Rong-Rong Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qian-Li Meng
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xin Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Haosheng Jin
- Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Ying Han
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Liang Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Zhi-Bin Zhao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, Davis, CA, USA.
| | - Zhe-Xiong Lian
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
52
|
Reina-Campos M, Monell A, Ferry A, Luna V, Cheung KP, Galletti G, Scharping NE, Takehara KK, Quon S, Boland B, Lin YH, Wong WH, Indralingam CS, Yeo GW, Chang JT, Heeg M, Goldrath AW. Functional Diversity of Memory CD8 T Cells is Spatiotemporally Imprinted. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.20.585130. [PMID: 38585842 PMCID: PMC10996520 DOI: 10.1101/2024.03.20.585130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Tissue-resident memory CD8 T cells (TRM) kill infected cells and recruit additional immune cells to limit pathogen invasion at barrier sites. Small intestinal (SI) TRM cells consist of distinct subpopulations with higher expression of effector molecules or greater memory potential. We hypothesized that occupancy of diverse anatomical niches imprints these distinct TRM transcriptional programs. We leveraged human samples and a murine model of acute systemic viral infection to profile the location and transcriptome of pathogen-specific TRM cell differentiation at single-transcript resolution. We developed computational approaches to capture cellular locations along three anatomical axes of the small intestine and to visualize the spatiotemporal distribution of cell types and gene expression. TRM populations were spatially segregated: with more effector- and memory-like TRM preferentially localized at the villus tip or crypt, respectively. Modeling ligand-receptor activity revealed patterns of key cellular interactions and cytokine signaling pathways that initiate and maintain TRM differentiation and functional diversity, including different TGFβ sources. Alterations in the cellular networks induced by loss of TGFβRII expression revealed a model consistent with TGFβ promoting progressive TRM maturation towards the villus tip. Ultimately, we have developed a framework for the study of immune cell interactions with the spectrum of tissue cell types, revealing that T cell location and functional state are fundamentally intertwined.
Collapse
Affiliation(s)
- Miguel Reina-Campos
- Division of Biological Sciences, Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Alexander Monell
- Division of Biological Sciences, Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Amir Ferry
- Division of Biological Sciences, Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Vida Luna
- Division of Biological Sciences, Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Kitty P. Cheung
- Division of Biological Sciences, Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Giovanni Galletti
- Division of Biological Sciences, Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Nicole E. Scharping
- Division of Biological Sciences, Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Kennidy K. Takehara
- Division of Biological Sciences, Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Sara Quon
- Division of Biological Sciences, Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Brigid Boland
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Yun Hsuan Lin
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - William H. Wong
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | - Gene W. Yeo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - John T. Chang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Maximilian Heeg
- Division of Biological Sciences, Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Ananda W. Goldrath
- Division of Biological Sciences, Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
- Allen Institute for Immunology, 615 Westlake Avenue N, Seattle, WA 98109, USA
| |
Collapse
|
53
|
Bhattacharya S, Myers JA, Baker C, Guo M, Danopoulos S, Myers JR, Bandyopadhyay G, Romas ST, Huyck HL, Misra RS, Dutra J, Holden-Wiltse J, McDavid AN, Ashton JM, Al Alam D, Potter SS, Whitsett JA, Xu Y, Pryhuber GS, Mariani TJ. Single-Cell Transcriptomic Profiling Identifies Molecular Phenotypes of Newborn Human Lung Cells. Genes (Basel) 2024; 15:298. [PMID: 38540357 PMCID: PMC10970229 DOI: 10.3390/genes15030298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 05/01/2024] Open
Abstract
While animal model studies have extensively defined the mechanisms controlling cell diversity in the developing mammalian lung, there exists a significant knowledge gap with regards to late-stage human lung development. The NHLBI Molecular Atlas of Lung Development Program (LungMAP) seeks to fill this gap by creating a structural, cellular and molecular atlas of the human and mouse lung. Transcriptomic profiling at the single-cell level created a cellular atlas of newborn human lungs. Frozen single-cell isolates obtained from two newborn human lungs from the LungMAP Human Tissue Core Biorepository, were captured, and library preparation was completed on the Chromium 10X system. Data was analyzed in Seurat, and cellular annotation was performed using the ToppGene functional analysis tool. Transcriptional interrogation of 5500 newborn human lung cells identified distinct clusters representing multiple populations of epithelial, endothelial, fibroblasts, pericytes, smooth muscle, immune cells and their gene signatures. Computational integration of data from newborn human cells and with 32,000 cells from postnatal days 1 through 10 mouse lungs generated by the LungMAP Cincinnati Research Center facilitated the identification of distinct cellular lineages among all the major cell types. Integration of the newborn human and mouse cellular transcriptomes also demonstrated cell type-specific differences in maturation states of newborn human lung cells. Specifically, newborn human lung matrix fibroblasts could be separated into those representative of younger cells (n = 393), or older cells (n = 158). Cells with each molecular profile were spatially resolved within newborn human lung tissue. This is the first comprehensive molecular map of the cellular landscape of neonatal human lung, including biomarkers for cells at distinct states of maturity.
Collapse
Affiliation(s)
- Soumyaroop Bhattacharya
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.B.); (S.T.R.); (H.L.H.); (R.S.M.); (G.S.P.); (T.J.M.)
| | - Jacquelyn A. Myers
- Genomic Research Center, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.A.M.); (C.B.); (J.R.M.); (J.M.A.)
| | - Cameron Baker
- Genomic Research Center, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.A.M.); (C.B.); (J.R.M.); (J.M.A.)
| | - Minzhe Guo
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45219, USA; (M.G.); (S.S.P.); (J.A.W.); (Y.X.)
| | - Soula Danopoulos
- Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, University of California Los Angeles, Los Angeles, CA 90024, USA; (S.D.)
| | - Jason R. Myers
- Genomic Research Center, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.A.M.); (C.B.); (J.R.M.); (J.M.A.)
| | - Gautam Bandyopadhyay
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.B.); (S.T.R.); (H.L.H.); (R.S.M.); (G.S.P.); (T.J.M.)
| | - Stephen T. Romas
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.B.); (S.T.R.); (H.L.H.); (R.S.M.); (G.S.P.); (T.J.M.)
| | - Heidie L. Huyck
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.B.); (S.T.R.); (H.L.H.); (R.S.M.); (G.S.P.); (T.J.M.)
| | - Ravi S. Misra
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.B.); (S.T.R.); (H.L.H.); (R.S.M.); (G.S.P.); (T.J.M.)
| | - Jennifer Dutra
- Clinical & Translational Science Institute, University of Rochester, Rochester, NY 14642, USA; (J.D.); (J.H.-W.)
| | - Jeanne Holden-Wiltse
- Clinical & Translational Science Institute, University of Rochester, Rochester, NY 14642, USA; (J.D.); (J.H.-W.)
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Andrew N. McDavid
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - John M. Ashton
- Genomic Research Center, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.A.M.); (C.B.); (J.R.M.); (J.M.A.)
| | - Denise Al Alam
- Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, University of California Los Angeles, Los Angeles, CA 90024, USA; (S.D.)
| | - S. Steven Potter
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45219, USA; (M.G.); (S.S.P.); (J.A.W.); (Y.X.)
| | - Jeffrey A. Whitsett
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45219, USA; (M.G.); (S.S.P.); (J.A.W.); (Y.X.)
| | - Yan Xu
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45219, USA; (M.G.); (S.S.P.); (J.A.W.); (Y.X.)
| | - Gloria S. Pryhuber
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.B.); (S.T.R.); (H.L.H.); (R.S.M.); (G.S.P.); (T.J.M.)
| | - Thomas J. Mariani
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.B.); (S.T.R.); (H.L.H.); (R.S.M.); (G.S.P.); (T.J.M.)
| |
Collapse
|
54
|
Fang J, Lei J, He B, Wu Y, Chen P, Sun Z, Wu N, Huang Y, Wei P, Yin L, Chen Y. Decoding the transcriptional heterogeneity, differentiation lineage, clinical significance in tissue-resident memory CD8 T cell of the small intestine by single-cell analysis. J Transl Med 2024; 22:203. [PMID: 38403590 PMCID: PMC10895748 DOI: 10.1186/s12967-024-04978-2] [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: 11/28/2023] [Accepted: 02/11/2024] [Indexed: 02/27/2024] Open
Abstract
Resident memory T (Trm) cells which are specifically located in non-lymphoid tissues showed distinct phenotypes and functions compared to circulating memory T cells and were vital for the initiation of robust immune response within tissues. However, the heterogeneity in the transcriptional features, development pathways, and cancer response of Trm cells in the small intestine was not demonstrated. Here, we integrated scRNA-seq and scTCR-seq data pan-tissue T cells to explore the heterogeneity of Trm cells and their development pathways. Trm were enriched in tissue-specific immune response and those in the DUO specially interacted with B cells via TNF and MHC-I signatures. T cell lineage analyses demonstrated that Trm might be derived from the T_CD4/CD8 subset within the same organ or migrated from spleen and mesenteric lymph nodes. We compared the immune repertoire of Trm among organs and implied that clonotypes in both DUO and ILE were less expanded and hydrophilic TRB CDR3s were enriched in the DUO. We further demonstrated that Trm in the intestine infiltrated the colorectal cancer and several effector molecules were highly expressed. Finally, the TCGA dataset of colorectal cancer implied that the infiltration of Trm from the DUO and the ILE was beneficial for overall survival and the response to immune checkpoint blockade.
Collapse
Affiliation(s)
- Jialing Fang
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Jun Lei
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Department of Laboratory Medicine, Xixi Hospital of Hangzhou, Hangzhou, China
| | - Boxiao He
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yankang Wu
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Peng Chen
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Zaiqiao Sun
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Ning Wu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafei Huang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengcheng Wei
- School of Medicine, Guangxi University, Nanning, 530004, China
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, 80206, USA
| | - Lei Yin
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.
| | - Yongshun Chen
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.
| |
Collapse
|
55
|
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
|
56
|
Manandhar P, Szymczak-Workman AL, Kane LP. Tim-3 Is Not Required for Establishment of CD8+ T Cell Memory to Lymphocytic Choriomeningitis Virus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:466-474. [PMID: 38108417 PMCID: PMC10906969 DOI: 10.4049/jimmunol.2300401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/21/2023] [Indexed: 12/19/2023]
Abstract
Tim-3 is a transmembrane protein that is best known for being highly expressed on terminally exhausted CD8+ T cells associated with chronic infection and tumors, although its expression is not limited to those settings. Tim-3 is also expressed by CD8+ T cells during acute infection and by multiple other immune cell types, including CD4+ Th1 and regulatory T cells, dendritic cells, and mast cells. In this study, we investigated the role of Tim-3 signaling on CD8+ T cell memory using a Tim-3 conditional knockout mouse model and mice lacking the signaling portion of the Tim-3 cytoplasmic domain. Together, our results indicate that Tim-3 has at most a modest effect on the formation and function of CD8+ memory T cells.
Collapse
Affiliation(s)
- Priyanka Manandhar
- Dept. of Immunology, University of Pittsburgh, Pittsburgh, PA 15213
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15213
| | | | - Lawrence P. Kane
- Dept. of Immunology, University of Pittsburgh, Pittsburgh, PA 15213
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA 15213
| |
Collapse
|
57
|
Long B, Zhou S, Gao Y, Fan K, Lai J, Yao C, Li J, Xu X, Yu S. Tissue-Resident Memory T Cells in Allergy. Clin Rev Allergy Immunol 2024; 66:64-75. [PMID: 38381299 DOI: 10.1007/s12016-024-08982-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2024] [Indexed: 02/22/2024]
Abstract
Tissue-resident memory T (TRM) cells constitute a distinct subset within the memory T cell population, serving as the vanguard against invading pathogens and antigens in peripheral non-lymphoid tissues, including the respiratory tract, intestines, and skin. Notably, TRM cells adapt to the specific microenvironment of each tissue, predominantly maintaining a sessile state with distinctive phenotypic and functional attributes. Their role is to ensure continuous immunological surveillance and protection. Recent findings have highlighted the pivotal contribution of TRM cells to the modulation of adaptive immune responses in allergic disorders such as allergic rhinitis, asthma, and dermatitis. A comprehensive understanding of the involvement of TRM cells in allergic diseases bears profound implications for allergy prevention and treatment. This review comprehensively explores the phenotypic characteristics, developmental mechanisms, and functional roles of TRM cells, focusing on their intricate relationship with allergic diseases.
Collapse
Affiliation(s)
- Bojin Long
- Department of Otorhinolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Allergy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Shican Zhou
- Department of Otorhinolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Allergy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Yawen Gao
- Department of Otorhinolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Allergy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Kai Fan
- Department of Otorhinolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Allergy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Ju Lai
- Department of Otorhinolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Allergy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Chunyan Yao
- Department of Otorhinolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Allergy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jingwen Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Allergy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Xiayue Xu
- Department of Otorhinolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Allergy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Shaoqing Yu
- Department of Otorhinolaryngology-Head and Neck Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
- Department of Allergy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| |
Collapse
|
58
|
Smolgovsky S, Theall B, Wagner N, Alcaide P. Fibroblasts and immune cells: at the crossroad of organ inflammation and fibrosis. Am J Physiol Heart Circ Physiol 2024; 326:H303-H316. [PMID: 38038714 PMCID: PMC11219060 DOI: 10.1152/ajpheart.00545.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/13/2023] [Accepted: 11/27/2023] [Indexed: 12/02/2023]
Abstract
The immune and fibrotic responses have evolved to work in tandem to respond to pathogen clearance and promote tissue repair. However, excessive immune and fibrotic responses lead to chronic inflammation and fibrosis, respectively, both of which are key pathological drivers of organ pathophysiology. Fibroblasts and immune cells are central to these responses, and evidence of these two cell types communicating through soluble mediators or adopting functions from each other through direct contact is constantly emerging. Here, we review complex junctions of fibroblast-immune cell cross talk, such as immune cell modulation of fibroblast physiology and fibroblast acquisition of immune cell-like functions, as well as how these systems of communication contribute to organ pathophysiology. We review the concept of antigen presentation by fibroblasts among different organs with different regenerative capacities, and then focus on the inflammation-fibrosis axis in the heart in the complex syndrome of heart failure. We discuss the need to develop anti-inflammatory and antifibrotic therapies, so far unsuccessful to date, that target novel mechanisms that sit at the crossroads of the fibrotic and immune responses.
Collapse
Affiliation(s)
- Sasha Smolgovsky
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, United States
- Immunology Graduate Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States
| | - Brandon Theall
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, United States
- Immunology Graduate Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States
| | - Noah Wagner
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, United States
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, United States
- Immunology Graduate Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States
| |
Collapse
|
59
|
Ackermann-Gäumann R, Lang P, Zens KD. Defining the "Correlate(s) of Protection" to tick-borne encephalitis vaccination and infection - key points and outstanding questions. Front Immunol 2024; 15:1352720. [PMID: 38318179 PMCID: PMC10840404 DOI: 10.3389/fimmu.2024.1352720] [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: 12/08/2023] [Accepted: 01/05/2024] [Indexed: 02/07/2024] Open
Abstract
Tick-borne Encephalitis (TBE) is a severe disease of the Central Nervous System (CNS) caused by the tick-borne encephalitis virus (TBEV). The generation of protective immunity after TBEV infection or TBE vaccination relies on the integrated responses of many distinct cell types at distinct physical locations. While long-lasting memory immune responses, in particular, form the basis for the correlates of protection against many diseases, these correlates of protection have not yet been clearly defined for TBE. This review addresses the immune control of TBEV infection and responses to TBE vaccination. Potential correlates of protection and the durability of protection against disease are discussed, along with outstanding questions in the field and possible areas for future research.
Collapse
Affiliation(s)
- Rahel Ackermann-Gäumann
- Microbiologie, ADMED Analyses et Diagnostics Médicaux, La Chaux-de-Fonds, Switzerland
- Swiss National Reference Center for Tick-transmitted Diseases, La Chaux-de-Fonds, Switzerland
| | - Phung Lang
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Kyra D. Zens
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
- Institute for Experimental Immunology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
60
|
Vazquez J, Mohamed MA, Banerjee S, Keding LT, Koenig MR, Leyva Jaimes F, Fisher RC, Bove EM, Golos TG, Stanic AK. Deciphering decidual leukocyte traffic with serial intravascular staining. Front Immunol 2024; 14:1332943. [PMID: 38268922 PMCID: PMC10806228 DOI: 10.3389/fimmu.2023.1332943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024] Open
Abstract
The decidual immunome is dynamic, dramatically changing its composition across gestation. Early pregnancy is dominated by decidual NK cells, with a shift towards T cells later in pregnancy. However, the degree, timing, and subset-specific nature of leukocyte traffic between the decidua and systemic circulation during gestation remains poorly understood. Herein, we employed intravascular staining in pregnant C57BL/6J mice and cynomolgus macaques (Macaca fascicularis) to examine leukocyte traffic into the decidual basalis during pregnancy. Timed-mated or virgin mice were tail-vein injected with labelled αCD45 antibodies 24 hours and 5 minutes before sacrifice. Pregnant cynomolgus macaques (GD155) were infused with labelled αCD45 at 2 hours or 5 mins before necropsy. Decidual cells were isolated and resulting suspensions analyzed by flow cytometry. We found that the proportion of intravascular (IVAs)-negative leukocytes (cells labeled by the 24h infusion of αCD45 or unlabeled) decreased across murine gestation while recent immigrants (24h label only) increased in mid- to late-gestation. In the cynomolgus model our data confirmed differential labeling of decidual leukocytes by the infused antibody, with the 5 min infused animal having a higher proportion of IVAs+ cells compared to the 2hr infused animal. Decidual tissue sections from both macaques showed the presence of intravascularly labeled cells, either in proximity to blood vessels (5min infused animal) or deeper into decidual stroma (2hr infused animal). These results demonstrate the value of serial intravascular staining as a sensitive tool for defining decidual leukocyte traffic during pregnancy.
Collapse
Affiliation(s)
- Jessica Vazquez
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin National Primate Research Center, Madison, WI, United States
| | - Mona A Mohamed
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Soma Banerjee
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Logan T Keding
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin National Primate Research Center, Madison, WI, United States
| | - Michelle R Koenig
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Fernanda Leyva Jaimes
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin National Primate Research Center, Madison, WI, United States
| | - Rachel C Fisher
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Emily M Bove
- Wisconsin National Primate Research Center, Madison, WI, United States
| | - Thaddeus G Golos
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin National Primate Research Center, Madison, WI, United States
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Aleksandar K Stanic
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
61
|
The SMML, Schreurs RRCE, Drewniak A, Bakx R, de Meij TGJ, Budding AE, Poort L, Cense HA, Heij HA, van Heurn LWE, Gorter RR, Bunders MJ. Enhanced Th17 responses in the appendix of children with complex compared to simple appendicitis are associated with microbial dysbiosis. Front Immunol 2024; 14:1258363. [PMID: 38239362 PMCID: PMC10794624 DOI: 10.3389/fimmu.2023.1258363] [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: 07/13/2023] [Accepted: 11/17/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Appendicitis is one of the most common causes of acute abdominal surgery in children. The clinical course of appendicitis ranges from simple to complex appendicitis. The mechanisms underlying the heterogeneity of appendicitis in children remain largely unclear. Dysregulated T cell responses play an important role in several inflammatory diseases of the intestine, but the extend of T cell dysregulation in appendicitis in children is less well known. Methods To characterize appendiceal T cells in simple and complex appendicitis we performed in-depth immunophenotyping of appendiceal-derived T cells by flow cytometry and correlated this to appendiceal-derived microbiota analyses of the same patient. Results Appendix samples of twenty children with appendicitis (n = 8 simple, n = 12 complex) were collected. T cells in complex appendicitis displayed an increased differentiated phenotype compared to simple appendicitis, including a loss of both CD27 and CD28 by CD4+ T cells and to a lesser extent by CD8+ T cells. Frequencies of phenotypic tissue-resident memory CD69+CD4+ T cells and CD69+CD8+ T cells were decreased in children with complex compared to simple appendicitis, indicating disruption of local tissue-resident immune responses. In line with the increased differentiated phenotype, cytokine production of in particular IL-17A by CD4+ T cells was increased in children with complex compared to simple appendicitis. Furthermore, frequencies of IL-17A+ CD4+ T cells correlated with a dysregulation of the appendiceal microbiota in children with complex appendicitis. Conclusion In conclusion, disruption of local T cell responses, and enhanced pro-inflammatory Th17 responses correlating to changes in the appendiceal microbiota were observed in children with complex compared to simple appendicitis. Further studies are needed to decipher the role of a dysregulated network of microbiota and Th17 cells in the development of complex appendicitis in children.
Collapse
Affiliation(s)
- Sarah-May M. L. The
- Department of Paediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Center (UMC), University of Amsterdam & Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Renée R. C. E. Schreurs
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Paediatrics, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam & Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Agata Drewniak
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Roel Bakx
- Department of Paediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Center (UMC), University of Amsterdam & Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
- Amsterdam Gastroenterology and Metabolism Research Institute, Amsterdam, Netherlands
| | - Tim G. J. de Meij
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
- Amsterdam Gastroenterology and Metabolism Research Institute, Amsterdam, Netherlands
- Department of Paediatric Gastroenterology, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | | | | | - Huib A. Cense
- Department of Surgery, Red Cross Hospital, Beverwijk, Netherlands
| | - Hugo A. Heij
- Department of Paediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Center (UMC), University of Amsterdam & Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - L. W. Ernest van Heurn
- Department of Paediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Center (UMC), University of Amsterdam & Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
- Amsterdam Gastroenterology and Metabolism Research Institute, Amsterdam, Netherlands
| | - Ramon R. Gorter
- Department of Paediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Center (UMC), University of Amsterdam & Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
- Amsterdam Gastroenterology and Metabolism Research Institute, Amsterdam, Netherlands
| | - Madeleine J. Bunders
- Leibniz Institute of Virology, Hamburg, Germany
- Third Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
62
|
Kumar NS, Reddy N, Kumar H, Vemireddy S. Immunomodulatory Plant Natural Products as Therapeutics against Inflammatory Skin Diseases. Curr Top Med Chem 2024; 24:1013-1034. [PMID: 38485678 DOI: 10.2174/0115680266277952240223120435] [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: 10/09/2023] [Revised: 12/30/2023] [Accepted: 01/11/2024] [Indexed: 07/16/2024]
Abstract
Frequently occurring inflammatory skin conditions such as psoriasis, dermatitis, acne, including skin cancer, wounds and other disorders arising out of premature skin aging, deteriorate skin health and adversely impact human life. Even though several synthetic compounds have evolved for treating these skin conditions, natural-product-based therapeutics are gaining popularity with growing evidence of their efficacy and safety for treating skin disorders. Many of these inflammatory skin diseases have underlying disturbances in our immune system and immunomodulatory natural products provide solutions for their effective treatment and aid in understanding the underlying mechanism of such inflammatory skin conditions. Based on this premise, the present review summarizes the possible application of plant-derived immunomodulatory compositions and single molecules for treating inflammatory skin conditions. In vitro, in vivo and mechanistic studies reported the application of selected plant-derived natural products for the treatment of inflammatory skin disorders including, cancer and infections. Several online databases including PubMed, Google Scholar, and Science Direct have been searched for gathering the information covered in this review. Empirical studies demonstrated that most of these natural compounds exhibited therapeutic properties through their immunomodulatory and anti-inflammatory potential supplemented often with anti-microbial, anti-neoplastic, and anti- oxidant activities. Overall, plant-based natural products discussed here are capable of modulating the immune system to minimize or completely suppress the pro-inflammatory markers, scavenge free radicals (ROS), prevent bacteria, fungal, and virus-derived skin infections and often regress skin cancer through the induction of apoptosis. The challenges and opportunities associated with the application of plant-based immunomodulators for skin applications and their safety considerations are also discussed here. The present study indicated that immunomodulatory plant natural products being biologically validated ligands against various biological targets manifested in inflammatory skin diseases, offer an effective, safe and affordable treatment for such disorders affecting skin health. However, further clinical evaluations are needed to substantiate these findings.
Collapse
Affiliation(s)
- Nikhila Sampath Kumar
- Department of Dermatology, Venereology and Leprosy, Kamineni Institute of Medical Sciences (KIMS), Narketpalli, Nalagonda District, Hyderabad, 500 007, Telangana, India
| | - Navaneetha Reddy
- Department of Dermatology, Venereology and Leprosy, Kamineni Institute of Medical Sciences (KIMS), Narketpalli, Nalagonda District, Hyderabad, 500 007, Telangana, India
| | - Halmuthur Kumar
- Vaccine Immunology Laboratory, Indian Institute of Chemical Technology, Hyderabad-500 007, India
| | - Sravanthi Vemireddy
- Vaccine Immunology Laboratory, Indian Institute of Chemical Technology, Hyderabad-500 007, India
| |
Collapse
|
63
|
Lama GIG, Noél G, Márquez FCL, Galarza FFG, Hinojosa AIP, Rivera LEN, Willard-Gallo K, Astorga JRA. Concomitant Expression of CD39, CD69, and CD103 Identifies Antitumor CD8 + T Cells in Breast Cancer Implications for Adoptive Cell Therapy. Curr Pharm Biotechnol 2024; 25:1747-1757. [PMID: 37680154 DOI: 10.2174/1389201025666230901094219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/22/2023] [Accepted: 07/20/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND In cancer, an effective immune response involves the action of several different cell types, among which CD8 T cells play a major role as they can specifically recognize and kill cancer cells via the release of cytotoxic molecules and cytokines, being of major importance for adoptive cell transfer (ACT) of ex vivo expanded tumor-infiltrating lymphocytes (TILs). The inflammation resulting from the tumor growth attracts both activated and bystander T cells. For an effective antitumor response, the T cell must express a specific group of chemokine receptors and integrins which include CD103, CD39, CD69, and CD25. These markers had already been analyzed in various cancers, not including breast cancer and their subsequent subtypes, until now. To analyze, the key receptors on ex vivo expanded tumor-infiltrating lymphocytes in luminal A and luminal B breast cancer (BC) subtypes. MATERIALS AND METHODS We were successful in expanding TILs ex vivo using a standard TIL culture condition from a cohort study of 15 primary luminal A and luminal B breast cancer patients. Furthermore, we examined the expression of CD103, CD39, CD69, and CD25 biomarkers after the expansion by flow cytometry. RESULTS We found that the information about the percentage of TILs obtainable after the ex vivo expansion is not associated to nor it is dependent on the heterogeneity of the TIL population before the expansion and does not differ by the molecular subtype (p>0.05). We also found that there is a major population of memory-resident antitumor CD8+CD103+CD39+ and CD8+CD103+ CD69+ TILs present in the stroma after the expansion when compared to CD4 immunosubtypes (p<0.0001). Only the CD8+CD103+CD39+ subpopulation was related to BC subtype (0.0009). CONCLUSION Evidence from our study suggests that CD8 TILs present in the stroma of luminal A and luminal B breast cancer patients can be quantified and phenotyped by flow cytometry and be further expanded ex vivo. The immuno-phenotyping of these markers may be targeted to improve the success of immunotherapeutic approaches, such as adoptive cellular therapy (ACT) in patients with BC.
Collapse
MESH Headings
- Breast Neoplasms/immunology
- Breast Neoplasms/therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Humans
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Integrin alpha Chains/metabolism
- Integrin alpha Chains/immunology
- Female
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/immunology
- Apyrase/metabolism
- Apyrase/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Immunotherapy, Adoptive/methods
- Lectins, C-Type/metabolism
- Lectins, C-Type/immunology
- Middle Aged
- Aged
Collapse
Affiliation(s)
- Grace Ivonne Gattas Lama
- Universidad Iberoamericana de Torreón, Coahuila, México
- Departamento de Inmunología Molecular, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Coahuila, México
| | | | - Francisco Carlos López Márquez
- Departamento de Inmunología Molecular, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Coahuila, México
| | | | - Adria Imelda Prieto Hinojosa
- Universidad Iberoamericana de Torreón, Coahuila, México
- Departamento de Inmunología Molecular, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Coahuila, México
| | - Lydia Enith Nava Rivera
- Departamento de Inmunología Molecular, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Coahuila, México
| | - Karen Willard-Gallo
- Departamento de Inmunología Molecular, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Coahuila, México
| | - Jesús Rafael Arguëllo Astorga
- Departamento de Inmunología Molecular, Facultad de Medicina, Universidad Autónoma de Coahuila, Torreón, Coahuila, México
- Instituto de Ciencias y Medicina Genómica, Torreón, Coahuila, México
| |
Collapse
|
64
|
Boothby M, Cho SH. Hypoxia and the Hypoxia-Inducible Factors in Lymphocyte Differentiation and Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:115-141. [PMID: 39017842 DOI: 10.1007/978-3-031-62731-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Molecular oxygen doubles as a biomolecular building block and an element required for energy generation and metabolism in aerobic organisms. A variety of systems in mammalian cells sense the concentration of oxygen to which they are exposed and are tuned to the range present in our blood and tissues. The ability to respond to insufficient O2 in tissues is central to regulation of erythroid lineage cells, but challenges also are posed for immune cells by a need to adjust to very different oxygen concentrations. Hypoxia-inducible factors (HIFs) provide a major means of making such adjustments. For adaptive immunity, lymphoid lineages are initially defined in bone marrow niches; T lineage cells arise in the thymus, and B cells complete maturation in the spleen. Lymphocytes move from these first stops into microenvironments (bloodstream, lymphatics, and tissues) with distinct oxygenation in each. Herein, evidence pertaining to functions of the HIF transcription factors (TFs) in lymphocyte differentiation and function is reviewed. For the CD4+ and CD8+ subsets of T cells, the case is very strong that hypoxia and HIFs regulate important differentiation events and functions after the naïve lymphocytes emerge from the thymus. In the B lineage, the data indicate that HIF1 contributes to a balanced regulation of B-cell fates after antigen (Ag) activation during immunity. A model synthesized from the aggregate literature is that HIF in lymphocytes generally serves to modulate function in a manner dependent on the molecular context framed by other TFs and signals.
Collapse
Affiliation(s)
- Mark Boothby
- Departments of Pathology, Microbiology, Immunology (Molecular Pathogenesis Division), Vanderbilt University Medical Center, Nashville, TN, USA.
- Medicine (Rheumatology and Immunology Division), Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Inflammation, Immunity (VI4), Nashville, TN, USA.
| | - Sung Hoon Cho
- Departments of Pathology, Microbiology, Immunology (Molecular Pathogenesis Division), Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Inflammation, Immunity (VI4), Nashville, TN, USA
| |
Collapse
|
65
|
Yu S, Wang K, Cao C, Zhang B, Chen Y, Wu C, Li C, Tang J, Luo W. Tissue-resident memory T cells exhibit phenotypically and functionally heterogeneous in human physiological and pathological nasal mucosa. Clin Immunol 2024; 258:109860. [PMID: 38065369 DOI: 10.1016/j.clim.2023.109860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 01/02/2024]
Abstract
Pathogens commonly enter mucosal barrier tissues and tissue-resident memory T cells (TRM) are essential for preventing mucosal lesions. However, the immunological properties of TRM cells in nasal mucosa are poorly known. In comparison with control tissues, decreasing CD103+ TRM cells were observed in Chronic rhinosinusitis with nasal polyps (CRSwNPs) and sinonasal inverted papilloma (SNIP), which presented high capability to produce effector cytokines. In CRSwNPs, we found that CD103+ TRM cells with higher cytokine and Granzyme B coexpressed high PD-1, CD103- TRM cells expressed higher IL-10. Homogenates isolated from CRSwNPs induced CD103 expression on peripheral T cells which could be inhibited by blocking TGF-β. The frequencies of CD103+ TRM cells in CRSwNPs were extremely negatively correlated with neutrophil infiltration. CD103+ TRM cells from Staphylococcus aureus positive CRSwNPs had a stronger response to SEB. Taken together, two phenotypically and functionally distinct subsets of TRM cells exist in nasal tissues and play critical roles in the progress of CRSwNPs and SNIPs.
Collapse
Affiliation(s)
- Sifei Yu
- Institute of translational medicine, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, PR China
| | - Kai Wang
- Department of Otolaryngology, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, PR China
| | - Chen Cao
- Department of Otolaryngology, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, PR China
| | - Beiying Zhang
- Institute of translational medicine, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, PR China
| | - Youmou Chen
- Department of Otolaryngology, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, PR China; The General Hospital of Western Theater Command, No. 270, Rongdu Avenue, Chengdu 610083, PR China
| | - Changyou Wu
- Clifford Hospital, Jinan University, No.3 Hongfu Road, Guangzhou 511495, PR China
| | - Chunwei Li
- Department of Otolaryngology, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, PR China
| | - Jun Tang
- Department of Otolaryngology, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, PR China.
| | - Wei Luo
- Institute of translational medicine, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, PR China.
| |
Collapse
|
66
|
Delclaux I, Ventre KS, Jones D, Lund AW. The tumor-draining lymph node as a reservoir for systemic immune surveillance. Trends Cancer 2024; 10:28-37. [PMID: 37863720 PMCID: PMC10843049 DOI: 10.1016/j.trecan.2023.09.006] [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: 08/06/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/22/2023]
Abstract
Early in solid tumor development, antigens are presented in tumor-draining lymph nodes (tdLNs), a process that is necessary to set up immune surveillance. Recent evidence indicates that tdLNs fuel systemic tumor-specific T cell responses which may halt cancer progression and facilitate future responses to immunotherapy. These protective responses, however, are subject to progressive dysfunction exacerbated by lymph node (LN) metastasis. We discuss emerging preclinical and clinical literature indicating that the tdLN is a crucial reservoir for systemic immunity that can potentiate immune surveillance. We also discuss the impact of LN metastasis and argue that a better understanding of the relationship between LN metastasis and systemic immunity will be necessary to direct regional disease management in the era of immunotherapy.
Collapse
Affiliation(s)
- Ines Delclaux
- Ronald O. Perelman Department of Dermatology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Katherine S Ventre
- Ronald O. Perelman Department of Dermatology, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Dennis Jones
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, New York University (NYU) Grossman School of Medicine, New York, NY, USA; Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA; Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
67
|
Burn OK, Dasyam N, Hermans IF. Recruiting Natural Killer T Cells to Improve Vaccination: Lessons from Preclinical and Clinical Studies. Crit Rev Oncog 2024; 29:31-43. [PMID: 38421712 DOI: 10.1615/critrevoncog.2023049407] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The capacity of type I natural killer T (NKT) cells to provide stimulatory signals to antigen-presenting cells has prompted preclinical research into the use of agonists as immune adjuvants, with much of this work focussed on stimulating T cell responses to cancer. In attempting to evaluate this approach in the clinic, our recent dendritic-cell based study failed to show an advantage to adding an agonist to the vaccine. Here we present potential limitations of the study, and suggest why other simpler strategies may be more effective. These include strategies to target antigen-presenting cells in the host, either through promoting efficient transfer from injected cell lines, facilitating uptake of antigen and agonist as injected conjugates, or encapsulating the components into injected nanovectors. While the vaccine landscape has changed with the rapid uptake of mRNA vaccines, we suggest that there is still a role for recruiting NKT cells in altering T cell differentiation programmes, notably the induction of resident memory T cells.
Collapse
Affiliation(s)
- Olivia K Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
68
|
Isola JVV, Ocañas SR, Hubbart CR, Ko S, Mondal SA, Hense JD, Carter HNC, Schneider A, Kovats S, Alberola-Ila J, Freeman WM, Stout MB. A single-cell atlas of the aging mouse ovary. NATURE AGING 2024; 4:145-162. [PMID: 38200272 PMCID: PMC10798902 DOI: 10.1038/s43587-023-00552-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 12/01/2023] [Indexed: 01/12/2024]
Abstract
Ovarian aging leads to diminished fertility, dysregulated endocrine signaling and increased chronic disease burden. These effects begin to emerge long before follicular exhaustion. Female humans experience a sharp decline in fertility around 35 years of age, which corresponds to declines in oocyte quality. Despite a growing body of work, the field lacks a comprehensive cellular map of the transcriptomic changes in the aging mouse ovary to identify early drivers of ovarian decline. To fill this gap we performed single-cell RNA sequencing on ovarian tissue from young (3-month-old) and reproductively aged (9-month-old) mice. Our analysis revealed a doubling of immune cells in the aged ovary, with lymphocyte proportions increasing the most, which was confirmed by flow cytometry. We also found an age-related downregulation of collagenase pathways in stromal fibroblasts, which corresponds to rises in ovarian fibrosis. Follicular cells displayed stress-response, immunogenic and fibrotic signaling pathway inductions with aging. This report provides critical insights into mechanisms responsible for ovarian aging phenotypes. The data can be explored interactively via a Shiny-based web application.
Collapse
Affiliation(s)
- José V V Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sarah R Ocañas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Neuroscience Department, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Physiology Department, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Chase R Hubbart
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sunghwan Ko
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Neuroscience Department, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Samim Ali Mondal
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Jessica D Hense
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Nutrition College, Federal University of Pelotas, Pelotas, Brazil
| | - Hannah N C Carter
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | | | - Susan Kovats
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - José Alberola-Ila
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
69
|
Shaikh N, Waterhölter A, Gnirck AC, Becker M, Adamiak V, Henneken L, Wunderlich M, Hartmann W, Linnemann L, Huber TB, Krebs CF, Panzer U, Locksley RM, Wilhelm C, Breloer M, Turner JE. Retinoic acid drives intestine-specific adaptation of effector ILC2s originating from distant sites. J Exp Med 2023; 220:e20221015. [PMID: 37773047 PMCID: PMC10541314 DOI: 10.1084/jem.20221015] [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: 06/13/2022] [Revised: 07/26/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023] Open
Abstract
Adaptation of immune cells to tissue-specific microenvironments is a crucial process in homeostasis and inflammation. Here, we show that murine effector type 2 innate lymphoid cells (ILC2s) from various organs are equally effective in repopulating ILC2 niches in other anatomical locations where they adapt tissue-specific phenotypes of target organs. Single-cell transcriptomics of ILC2 populations revealed upregulation of retinoic acid (RA) signaling in ILC2s during adaptation to the small intestinal microenvironment, and RA signaling mediated reprogramming of kidney effector ILC2s toward the small intestinal phenotype in vitro and in vivo. Inhibition of intestinal ILC2 adaptation by blocking RA signaling impaired worm expulsion during Strongyloides ratti infection, indicating functional importance of ILC2 tissue imprinting. In conclusion, this study highlights that effector ILC2s retain the ability to adapt to changing tissue-specific microenvironments, enabling them to exert tissue-specific functions, such as promoting control of intestinal helminth infections.
Collapse
Affiliation(s)
- Nikhat Shaikh
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alex Waterhölter
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ann-Christin Gnirck
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martina Becker
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Virginia Adamiak
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lena Henneken
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malte Wunderlich
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wiebke Hartmann
- Helminth Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Lara Linnemann
- Helminth Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Tobias B. Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian F. Krebs
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Division of Translational Immunology, III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulf Panzer
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Division of Translational Immunology, III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Richard M. Locksley
- Department of Medicine, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Christoph Wilhelm
- Unit for Immunopathology, Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Minka Breloer
- Helminth Immunology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Jan-Eric Turner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
70
|
Schenkel JM, Pauken KE. Localization, tissue biology and T cell state - implications for cancer immunotherapy. Nat Rev Immunol 2023; 23:807-823. [PMID: 37253877 PMCID: PMC11448857 DOI: 10.1038/s41577-023-00884-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2023] [Indexed: 06/01/2023]
Abstract
Tissue localization is a critical determinant of T cell immunity. CD8+ T cells are contact-dependent killers, which requires them to physically be within the tissue of interest to kill peptide-MHC class I-bearing target cells. Following their migration and extravasation into tissues, T cells receive many extrinsic cues from the local microenvironment, and these signals shape T cell differentiation, fate and function. Because major organ systems are variable in their functions and compositions, they apply disparate pressures on T cells to adapt to the local microenvironment. Additional complexity arises in the context of malignant lesions (either primary or metastatic), and this has made understanding the factors that dictate T cell function and longevity in tumours challenging. Moreover, T cell differentiation state influences how cues from the microenvironment are interpreted by tissue-infiltrating T cells, highlighting the importance of T cell state in the context of tissue biology. Here, we review the intertwined nature of T cell differentiation state, location, survival and function, and explain how dysfunctional T cell populations can adopt features of tissue-resident memory T cells to persist in tumours. Finally, we discuss how these factors have shaped responses to cancer immunotherapy.
Collapse
Affiliation(s)
- Jason M Schenkel
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Kristen E Pauken
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
71
|
Liang Y. Pathogenicity and virulence of influenza. Virulence 2023; 14:2223057. [PMID: 37339323 DOI: 10.1080/21505594.2023.2223057] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/03/2023] [Accepted: 06/05/2023] [Indexed: 06/22/2023] Open
Abstract
Influenza viruses, including four major types (A, B, C, and D), can cause mild-to-severe and lethal diseases in humans and animals. Influenza viruses evolve rapidly through antigenic drift (mutation) and shift (reassortment of the segmented viral genome). New variants, strains, and subtypes have emerged frequently, causing epidemic, zoonotic, and pandemic infections, despite currently available vaccines and antiviral drugs. In recent years, avian influenza viruses, such as H5 and H7 subtypes, have caused hundreds to thousands of zoonotic infections in humans with high case fatality rates. The likelihood of these animal influenza viruses acquiring airborne transmission in humans through viral evolution poses great concern for the next pandemic. Severe influenza viral disease is caused by both direct viral cytopathic effects and exacerbated host immune response against high viral loads. Studies have identified various mutations in viral genes that increase viral replication and transmission, alter tissue tropism or species specificity, and evade antivirals or pre-existing immunity. Significant progress has also been made in identifying and characterizing the host components that mediate antiviral responses, pro-viral functions, or immunopathogenesis following influenza viral infections. This review summarizes the current knowledge on viral determinants of influenza virulence and pathogenicity, protective and immunopathogenic aspects of host innate and adaptive immune responses, and antiviral and pro-viral roles of host factors and cellular signalling pathways. Understanding the molecular mechanisms of viral virulence factors and virus-host interactions is critical for the development of preventive and therapeutic measures against influenza diseases.
Collapse
Affiliation(s)
- Yuying Liang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, USA
| |
Collapse
|
72
|
Rosine N, Fogel O, Koturan S, Rogge L, Bianchi E, Miceli-Richard C. T cells in the pathogenesis of axial spondyloarthritis. Joint Bone Spine 2023; 90:105619. [PMID: 37487956 DOI: 10.1016/j.jbspin.2023.105619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/26/2023]
Abstract
Axial spondyloarthritis (axSpA) is the prototype of the spondyloarthritis spectrum. The involvement of T cells in its pathogenesis has long been suspected on the basis of the association with the major histocompatibility complex I molecule HLA-B27 and the pivotal role of interleukin 17 in the inflammatory mechanisms associated with the disease. Moreover, the presence of unconventional or "innate-like" T cells within the axial enthesis suggests an important role for these cells in the pathophysiology of the disease. In this review, we describe the characteristics and the interleukin 17 secretion capacity of the T-cell subsets identified in axSpA. We discuss the genetic and epigenetic mechanisms that support the alteration of T-cell functions and promote their activation in axSpA. We also discuss recent data on T cells that could explain the extra-articular manifestations of the SpA spectrum.
Collapse
Affiliation(s)
- Nicolas Rosine
- Service de rhumatologie, université Angers, CHU d'Angers, Paris, France.
| | - Olivier Fogel
- Department of Rheumatology, EULAR Center of Excellence, hôpital Cochin, Assistance publique-Hôpitaux de Paris, Paris University, Paris, France
| | - Surya Koturan
- Faculty of Medicine, MRC London Institute of Medical Science, Institute of Clinical Sciences, Imperial College, W12 0NN London, United Kingdom
| | - Lars Rogge
- Immunoregulation Unit, Institut Pasteur, université Paris Cité, 75015 Paris, France
| | - Elisabetta Bianchi
- Immunoregulation Unit, Institut Pasteur, université Paris Cité, 75015 Paris, France
| | - Corinne Miceli-Richard
- Department of Rheumatology, EULAR Center of Excellence, hôpital Cochin, Assistance publique-Hôpitaux de Paris, Paris University, Paris, France
| |
Collapse
|
73
|
Park SL, Christo SN, Wells AC, Gandolfo LC, Zaid A, Alexandre YO, Burn TN, Schröder J, Collins N, Han SJ, Guillaume SM, Evrard M, Castellucci C, Davies B, Osman M, Obers A, McDonald KM, Wang H, Mueller SN, Kannourakis G, Berzins SP, Mielke LA, Carbone FR, Kallies A, Speed TP, Belkaid Y, Mackay LK. Divergent molecular networks program functionally distinct CD8 + skin-resident memory T cells. Science 2023; 382:1073-1079. [PMID: 38033053 DOI: 10.1126/science.adi8885] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023]
Abstract
Skin-resident CD8+ T cells include distinct interferon-γ-producing [tissue-resident memory T type 1 (TRM1)] and interleukin-17 (IL-17)-producing (TRM17) subsets that differentially contribute to immune responses. However, whether these populations use common mechanisms to establish tissue residence is unknown. In this work, we show that TRM1 and TRM17 cells navigate divergent trajectories to acquire tissue residency in the skin. TRM1 cells depend on a T-bet-Hobit-IL-15 axis, whereas TRM17 cells develop independently of these factors. Instead, c-Maf commands a tissue-resident program in TRM17 cells parallel to that induced by Hobit in TRM1 cells, with an ICOS-c-Maf-IL-7 axis pivotal to TRM17 cell commitment. Accordingly, by targeting this pathway, skin TRM17 cells can be ablated without compromising their TRM1 counterparts. Thus, skin-resident T cells rely on distinct molecular circuitries, which can be exploited to strategically modulate local immunity.
Collapse
Affiliation(s)
- Simone L Park
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Susan N Christo
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Alexandria C Wells
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Luke C Gandolfo
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, VIC, Australia
- Walter and Eliza Hall Institute for Medical Research, Parkville, VIC, Australia
| | - Ali Zaid
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Yannick O Alexandre
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Thomas N Burn
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Jan Schröder
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Nicholas Collins
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Seong-Ji Han
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Stéphane M Guillaume
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Maximilien Evrard
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Clara Castellucci
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Brooke Davies
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Maleika Osman
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Andreas Obers
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Keely M McDonald
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Huimeng Wang
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - George Kannourakis
- Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC, Australia
- Fiona Elsey Cancer Research Institute, Ballarat, VIC, Australia
| | - Stuart P Berzins
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC, Australia
- Fiona Elsey Cancer Research Institute, Ballarat, VIC, Australia
| | - Lisa A Mielke
- Olivia Newton-John Cancer Research Institute, La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Francis R Carbone
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Axel Kallies
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Terence P Speed
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, VIC, Australia
- Walter and Eliza Hall Institute for Medical Research, Parkville, VIC, Australia
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
- NIAID Microbiome Program, NIAID, National Institutes of Health, Bethesda, MD, USA
| | - Laura K Mackay
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
74
|
Muñoz-Ruiz M, Llorian M, D'Antuono R, Pavlova A, Mavrigiannaki AM, McKenzie D, García-Cassani B, Iannitto ML, Wu Y, Dart R, Davies D, Jamal-Hanjani M, Jandke A, Ushakov DS, Hayday AC. IFN-γ-dependent interactions between tissue-intrinsic γδ T cells and tissue-infiltrating CD8 T cells limit allergic contact dermatitis. J Allergy Clin Immunol 2023; 152:1520-1540. [PMID: 37562754 DOI: 10.1016/j.jaci.2023.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/27/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Elicitation of allergic contact dermatitis (ACD), an inflammatory type 4 hypersensitivity disease, induces skin infiltration by polyclonal effector CD8 αβ T cells and precursors of tissue-resident memory T (TRM) cells. Because TRM have long-term potential to contribute to body-surface immunoprotection and immunopathology, their local regulation needs a fuller understanding. OBJECTIVE We sought to investigate how TRM-cell maturation might be influenced by innate-like T cells pre-existing within many epithelia. METHODS This study examined CD8+ TRM-cell maturation following hapten-induced ACD in wild-type mice and in strains harboring altered compartments of dendritic intraepidermal γδ T cells (DETCs), a prototypic tissue-intrinsic, innate-like T-cell compartment that reportedly regulates ACD, but by no elucidated mechanism. RESULTS In addition to eliciting CD8 TRM, ACD induced DETC activation and an intimate coregulatory association of the 2 cell types. This depended on DETC sensing IFN-γ produced by CD8 cells and involved programmed death-ligand 1 (PD-L1). Thus, in mice lacking DETC or lacking IFN-γ receptor solely on γδ cells, ACD-elicited CD8 T cells showed enhanced proliferative and effector potentials and reduced motility, collectively associated with exaggerated ACD pathology. Comparable dysregulation was elicited by PD-L1 blockade in vitro, and IFN-γ-regulated PD-L1 expression was a trait of human skin-homing and intraepithelial γδ T cells. CONCLUSIONS The size and quality of the tissue-infiltrating CD8 T-cell response during ACD can be profoundly regulated by local innate-like T cells responding to IFN-γ and involving PD-L1. Thus, interindividual and tissue-specific variations in tissue-intrinsic lymphocytes may influence responses to allergens and other challenges and may underpin inflammatory pathologies such as those repeatedly observed in γδ T-cell-deficient settings.
Collapse
Affiliation(s)
- Miguel Muñoz-Ruiz
- Immunosurveillance Laboratory, The Francis Crick Institute, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom; Department of Immunology, Ophthalmology and Ear, Nose and Throat, Complutense University School of Medicine and 12 de Octubre Health Research Institute, Madrid, Spain
| | - Miriam Llorian
- Bioinformatics and Biostatistics science technology platform (STP), The Francis Crick Institute, London, United Kingdom
| | - Rocco D'Antuono
- Light Microscopy STP, The Francis Crick Institute, London, United Kingdom
| | - Anna Pavlova
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Erlangen, Germany
| | | | - Duncan McKenzie
- Immunosurveillance Laboratory, The Francis Crick Institute, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom
| | - Bethania García-Cassani
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Maria Luisa Iannitto
- Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom
| | - Yin Wu
- Immunosurveillance Laboratory, The Francis Crick Institute, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom; Centre for Inflammation Biology and Cancer Immunology, King's College London, London, United Kingdom
| | - Robin Dart
- Immunosurveillance Laboratory, The Francis Crick Institute, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom
| | - Daniel Davies
- Immunosurveillance Laboratory, The Francis Crick Institute, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Anett Jandke
- Immunosurveillance Laboratory, The Francis Crick Institute, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom
| | - Dmitry S Ushakov
- Immunosurveillance Laboratory, The Francis Crick Institute, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom; Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Adrian C Hayday
- Immunosurveillance Laboratory, The Francis Crick Institute, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom; Centre for Inflammation Biology and Cancer Immunology, King's College London, London, United Kingdom.
| |
Collapse
|
75
|
Ulibarri MR, Lin Y, Ramprashad JR, Han G, Hasan MH, Mithila FJ, Ma C, Gopinath S, Zhang N, Milner JJ, Beura LK. Epithelial organoid supports resident memory CD8 T cell differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569395. [PMID: 38076957 PMCID: PMC10705482 DOI: 10.1101/2023.12.01.569395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Resident Memory T cells (TRM) play a vital role in regional immune defense in barrier organs. Although laboratory rodents have been extensively used to study fundamental TRM biology, poor isolation efficiency, sampling bias and low cell survival rates have limited our ability to conduct TRM-focused high-throughput assays. Here, we engineered a murine vaginal epithelial organoid (VEO)-CD8 T cell co-culture system that supports CD8 TRM differentiation in vitro. The three-dimensional VEOs established from murine adult stem cells resembled stratified squamous vaginal epithelium and induced gradual differentiation of activated CD8 T cells into epithelial TRM. These in vitro generated TRM were phenotypically and transcriptionally similar to in vivo TRM, and key tissue residency features were reinforced with a second cognate-antigen exposure during co-culture. TRM differentiation was not affected even when VEOs and CD8 T cells were separated by a semipermeable barrier, indicating soluble factors' involvement. Pharmacological and genetic approaches showed that TGF-β signaling played a crucial role in their differentiation. We found that the VEOs in our model remained susceptible to viral infections and the CD8 T cells were amenable to genetic manipulation; both of which will allow detailed interrogation of antiviral CD8 T cell biology in a reductionist setting. In summary, we established a robust model which captures bonafide TRM differentiation that is scalable, open to iterative sampling, and can be subjected to high throughput assays that will rapidly add to our understanding of TRM.
Collapse
Affiliation(s)
- Max R. Ulibarri
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
| | - Ying Lin
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
- Pathobiology Graduate Program, Brown University, Providence, RI, 02912
| | - Julian R. Ramprashad
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
| | - Geongoo Han
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
| | - Mohammad H. Hasan
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
| | - Farha J. Mithila
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
- Molecular Biology, Cell Biology and Biochemistry Graduate Program, Brown University, Providence, RI, 02912
| | - Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, TX, 78229
| | - Smita Gopinath
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Cambridge, MA, 02115
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, TX, 78229
- South Texas Veterans Health Care System, San Antonio, TX, 78229
| | - J. Justin Milner
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, 27599
| | - Lalit K. Beura
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912
| |
Collapse
|
76
|
Chen K, Gu X, Yang S, Tao R, Fan M, Bao W, Wang X. Research progress on intestinal tissue-resident memory T cells in inflammatory bowel disease. Scand J Immunol 2023; 98:e13332. [PMID: 38441381 DOI: 10.1111/sji.13332] [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: 02/08/2023] [Revised: 09/18/2023] [Accepted: 09/21/2023] [Indexed: 03/07/2024]
Abstract
Tissue-resident memory T (TRM) cells are a recently discovered subpopulation of memory T cells that reside in non-lymphoid tissues such as the intestine and skin and do not enter the bloodstream. The intestine encounters numerous pathogens daily. Intestinal mucosal immunity requires a balance between immune responses to pathogens and tolerance to food antigens and symbiotic microbiota. Therefore, intestinal TRM cells exhibit unique characteristics. In healthy intestines, TRM cells induce necessary inflammation to strengthen the intestinal barrier and inhibit bacterial translocation. During intestinal infections, TRM cells rapidly eliminate pathogens by proliferating, releasing cytokines, and recruiting other immune cells. Moreover, certain TRM cell subsets may have regulatory functions. The involvement of TRM cells in inflammatory bowel disease (IBD) is increasingly recognized as a critical factor. In IBD, the number of pro-inflammatory TRM cells increases, whereas the number of regulatory subgroups decreases. Additionally, the classic markers, CD69 and CD103, are not ideal for intestinal TRM cells. Here, we review the phenotype, development, maintenance, and function of intestinal TRM cells, as well as the latest findings in the context of IBD. Further understanding of the function of intestinal TRM cells and distinguishing their subgroups is crucial for developing therapeutic strategies to target these cells.
Collapse
Affiliation(s)
- Ke Chen
- Nanjing Medical University, Nanjing, China
| | - Xin Gu
- Nanjing Medical University, Nanjing, China
| | | | - Rui Tao
- Nanjing Medical University, Nanjing, China
| | | | | | - Xiaoyun Wang
- Wuxi Second Hospital Affiliated to Nanjing Medical University, Wuxi, China
| |
Collapse
|
77
|
Maseda D, Manfredo-Vieira S, Payne AS. T cell and bacterial microbiota interaction at intestinal and skin epithelial interfaces. DISCOVERY IMMUNOLOGY 2023; 2:kyad024. [PMID: 38567051 PMCID: PMC10917213 DOI: 10.1093/discim/kyad024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/28/2023] [Accepted: 11/24/2023] [Indexed: 04/04/2024]
Abstract
Graphical Abstract.
Collapse
Affiliation(s)
- Damian Maseda
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Silvio Manfredo-Vieira
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aimee S Payne
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
78
|
Kaminski A, Hager FT, Kopplin L, Ticconi F, Leufgen A, Vendelova E, Rüttger L, Gasteiger G, Cerovic V, Kastenmüller W, Pabst O, Ugur M. Resident regulatory T cells reflect the immune history of individual lymph nodes. Sci Immunol 2023; 8:eadj5789. [PMID: 37874251 DOI: 10.1126/sciimmunol.adj5789] [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: 07/04/2023] [Accepted: 10/10/2023] [Indexed: 10/25/2023]
Abstract
Regulatory T cells (Tregs) are present in lymphoid and nonlymphoid tissues where they restrict immune activation, prevent autoimmunity, and regulate inflammation. Tregs in nonlymphoid tissues are typically resident, whereas those in lymph nodes (LNs) are considered to recirculate. However, Tregs in LNs are not a homogenous population, and circulation kinetics of different Treg subsets are poorly characterized. Furthermore, whether Tregs can acquire memory T cell properties and persist for extended periods after their activation in LNs is unclear. Here, we used in situ labeling with a stabilized photoconvertible protein to uncover turnover rates of Tregs in LNs in vivo. We found that, whereas most Tregs in LNs recirculate, 10 to 20% are memory-like resident cells that remain in their respective LNs for weeks to months. Single-cell RNA sequencing revealed that LN-resident cells are a functionally and ontogenetically heterogeneous population and share the same core residency gene signature with conventional CD4+ and CD8+ T cells. Resident cells in LNs did not actively proliferate and did not require continuous T cell receptor (TCR) signaling for their residency. However, resident and circulating Tregs had distinct TCR repertoires, and each LN contained exclusive clonal subpopulations of resident Tregs. Our results demonstrate that, similar to conventional T cells, Tregs can form resident memory-like populations in LNs after adaptive immune responses. Specific and local suppression of immune responses by resident Tregs in draining LNs might provide previously unidentified therapeutic opportunities for the treatment of local chronic inflammatory conditions.
Collapse
Affiliation(s)
- Anne Kaminski
- Institute of Molecular Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Fabian Tobias Hager
- Institute of Molecular Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Lydia Kopplin
- Institute of Molecular Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Fabio Ticconi
- Institute of Molecular Medicine, RWTH Aachen University, Aachen 52074, Germany
- Institute for Computational Genomics, RWTH Aachen University, Aachen 52074, Germany
| | - Andrea Leufgen
- Institute of Molecular Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Emilia Vendelova
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Würzburg 97078, Germany
| | - Lennart Rüttger
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Würzburg 97078, Germany
| | - Georg Gasteiger
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Würzburg 97078, Germany
| | - Vuk Cerovic
- Institute of Molecular Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Wolfgang Kastenmüller
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Würzburg 97078, Germany
| | - Oliver Pabst
- Institute of Molecular Medicine, RWTH Aachen University, Aachen 52074, Germany
| | - Milas Ugur
- Institute of Molecular Medicine, RWTH Aachen University, Aachen 52074, Germany
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Würzburg 97078, Germany
| |
Collapse
|
79
|
Kingstad-Bakke B, Cleven T, Bussan H, Yount BL, Uraki R, Iwatsuki-Horimoto K, Koga M, Yamamoto S, Yotsuyanagi H, Park H, Mishra JS, Kumar S, Baric RS, Halfmann PJ, Kawaoka Y, Suresh M. Airway surveillance and lung viral control by memory T cells induced by COVID-19 mRNA vaccine. JCI Insight 2023; 8:e172510. [PMID: 37796612 PMCID: PMC10721330 DOI: 10.1172/jci.insight.172510] [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: 05/23/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023] Open
Abstract
Although SARS-CoV-2 evolution seeds a continuous stream of antibody-evasive viral variants, COVID-19 mRNA vaccines provide robust protection against severe disease and hospitalization. Here, we asked whether mRNA vaccine-induced memory T cells limit lung SARS-CoV-2 replication and severe disease. We show that mice and humans receiving booster BioNTech mRNA vaccine developed potent CD8 T cell responses and showed similar kinetics of expansion and contraction of granzyme B/perforin-expressing effector CD8 T cells. Both monovalent and bivalent mRNA vaccines elicited strong expansion of a heterogeneous pool of terminal effectors and memory precursor effector CD8 T cells in spleen, inguinal and mediastinal lymph nodes, pulmonary vasculature, and most surprisingly in the airways, suggestive of systemic and regional surveillance. Furthermore, we document that: (a) CD8 T cell memory persists in multiple tissues for > 200 days; (b) following challenge with pathogenic SARS-CoV-2, circulating memory CD8 T cells rapidly extravasate to the lungs and promote expeditious viral clearance, by mechanisms that require CD4 T cell help; and (c) adoptively transferred splenic memory CD8 T cells traffic to the airways and promote lung SARS-CoV-2 clearance. These findings provide insights into the critical role of memory T cells in preventing severe lung disease following breakthrough infections with antibody-evasive SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Brock Kingstad-Bakke
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Thomas Cleven
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Hailey Bussan
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Boyd L. Yount
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ryuta Uraki
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | | | - Michiko Koga
- Department of Infectious Diseases and Applied Immunology, IMSUT Hospital of The Institute of Medical Science, and
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Shinya Yamamoto
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiroshi Yotsuyanagi
- Department of Infectious Diseases and Applied Immunology, IMSUT Hospital of The Institute of Medical Science, and
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hongtae Park
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jay S. Mishra
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sathish Kumar
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ralph S. Baric
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Peter J. Halfmann
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
- The University of Tokyo, Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), Tokyo, Japan
| | - M. Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
80
|
DeDreu J, Basta MD, Walker JL, Menko AS. Immune Responses Induced at One Hour Post Cataract Surgery Wounding of the Chick Lens. Biomolecules 2023; 13:1615. [PMID: 38002297 PMCID: PMC10668984 DOI: 10.3390/biom13111615] [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: 07/31/2023] [Revised: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
While the lens is an avascular tissue with an immune-privileged status, studies have now revealed that there are immune responses specifically linked to the lens. The response to lens injury, such as following cataract surgery, has been shown to involve the activation of the resident immune cell population of the lens and the induction of immunomodulatory factors by the wounded epithelium. However, there has been limited investigation into the immediate response of the lens to wounding, particularly those induced factors that are intrinsic to the lens and its associated resident immune cells. Using an established chick embryo ex vivo cataract surgery model has made it possible to determine the early immune responses of this tissue to injury, including its resident immune cells, through a transcriptome analysis. RNA-seq studies were performed to determine the gene expression profile at 1 h post wounding compared to time 0. The results provided evidence that, as occurs in other tissues, the resident immune cells of the lens rapidly acquired a molecular signature consistent with their activation. These studies also identified the expression of many inflammatory factors by the injured lens that are associated with both the induction and regulation of the immune response.
Collapse
Affiliation(s)
- JodiRae DeDreu
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (M.D.B.); (J.L.W.)
| | - Morgan D. Basta
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (M.D.B.); (J.L.W.)
| | - Janice L. Walker
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (M.D.B.); (J.L.W.)
- Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - A. Sue Menko
- Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA (M.D.B.); (J.L.W.)
- Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
81
|
Suchanek O, Ferdinand JR, Tuong ZK, Wijeyesinghe S, Chandra A, Clauder AK, Almeida LN, Clare S, Harcourt K, Ward CJ, Bashford-Rogers R, Lawley T, Manz RA, Okkenhaug K, Masopust D, Clatworthy MR. Tissue-resident B cells orchestrate macrophage polarisation and function. Nat Commun 2023; 14:7081. [PMID: 37925420 PMCID: PMC10625551 DOI: 10.1038/s41467-023-42625-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 10/17/2023] [Indexed: 11/06/2023] Open
Abstract
B cells play a central role in humoral immunity but also have antibody-independent functions. Studies to date have focused on B cells in blood and secondary lymphoid organs but whether B cells reside in non-lymphoid organs (NLO) in homeostasis is unknown. Here we identify, using intravenous labeling and parabiosis, a bona-fide tissue-resident B cell population in lung, liver, kidney and urinary bladder, a substantial proportion of which are B-1a cells. Tissue-resident B cells are present in neonatal tissues and also in germ-free mice NLOs, albeit in lower numbers than in specific pathogen-free mice and following co-housing with 'pet-store' mice. They spatially co-localise with macrophages and regulate their polarization and function, promoting an anti-inflammatory phenotype, in-part via interleukin-10 production, with effects on bacterial clearance during urinary tract infection. Thus, our data reveal a critical role for tissue-resident B cells in determining the homeostatic 'inflammatory set-point' of myeloid cells, with important consequences for tissue immunity.
Collapse
Affiliation(s)
- Ondrej Suchanek
- Molecular Immunity Unit, University of Cambridge Department of Medicine, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - John R Ferdinand
- Molecular Immunity Unit, University of Cambridge Department of Medicine, Cambridge, UK
| | - Zewen K Tuong
- Molecular Immunity Unit, University of Cambridge Department of Medicine, Cambridge, UK
| | - Sathi Wijeyesinghe
- Department of Microbiology and Immunology, Centre for Immunology, University of Minnesota, Minneapolis, MI, USA
| | - Anita Chandra
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ann-Katrin Clauder
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Germany
| | - Larissa N Almeida
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Germany
| | - Simon Clare
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Christopher J Ward
- Molecular Immunity Unit, University of Cambridge Department of Medicine, Cambridge, UK
| | | | - Trevor Lawley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Rudolf A Manz
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Germany
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - David Masopust
- Department of Microbiology and Immunology, Centre for Immunology, University of Minnesota, Minneapolis, MI, USA
| | - Menna R Clatworthy
- Molecular Immunity Unit, University of Cambridge Department of Medicine, Cambridge, UK.
- Cambridge University Hospitals NHS Foundation Trust, and NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
| |
Collapse
|
82
|
Westerhof LM, Noonan J, Hargrave KE, Chimbayo ET, Cheng Z, Purnell T, Jackson MR, Borcherding N, MacLeod MKL. Multifunctional cytokine production marks influenza A virus-specific CD4 T cells with high expression of survival molecules. Eur J Immunol 2023; 53:e2350559. [PMID: 37490492 PMCID: PMC10947402 DOI: 10.1002/eji.202350559] [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: 05/08/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/27/2023]
Abstract
Cytokine production by memory T cells is a key mechanism of T cell mediated protection. However, we have limited understanding of the persistence of cytokine producing T cells during memory cell maintenance and secondary responses. We interrogated antigen-specific CD4 T cells using a mouse influenza A virus infection model. Although CD4 T cells detected using MHCII tetramers declined in lymphoid and non-lymphoid organs, we found similar numbers of cytokine+ CD4 T cells at days 9 and 30 in the lymphoid organs. CD4 T cells with the capacity to produce cytokines expressed higher levels of pro-survival molecules, CD127 and Bcl2, than non-cytokine+ cells. Transcriptomic analysis revealed a heterogeneous population of memory CD4 T cells with three clusters of cytokine+ cells. These clusters match flow cytometry data and reveal an enhanced survival signature in cells capable of producing multiple cytokines. Following re-infection, multifunctional T cells expressed low levels of the proliferation marker, Ki67, whereas cells that only produce the anti-viral cytokine, interferon-γ, were more likely to be Ki67+ . Despite this, multifunctional memory T cells formed a substantial fraction of the secondary memory pool. Together these data indicate that survival rather than proliferation may dictate which populations persist within the memory pool.
Collapse
Affiliation(s)
| | - Jonathan Noonan
- Baker Heart and Diabetes Institute & Baker Department of Cardiometabolic HealthUniversity of MelbourneMelbourneAustralia
| | | | - Elizabeth T. Chimbayo
- School of Infection and ImmunityUniversity of GlasgowGlasgowUK
- Malawi Liverpool Wellcome CentreBlantyreMalawi
| | - Zhiling Cheng
- School of Infection and ImmunityUniversity of GlasgowGlasgowUK
| | - Thomas Purnell
- School of Infection and ImmunityUniversity of GlasgowGlasgowUK
| | | | | | | |
Collapse
|
83
|
Zheng C, Cao T, Ye C, Zou Y. Neutrophil recruitment by CD4 tissue-resident memory T cells induces chronic recurrent inflammation in atopic dermatitis. Clin Immunol 2023; 256:109805. [PMID: 37832861 DOI: 10.1016/j.clim.2023.109805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/18/2023] [Accepted: 10/07/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic inflammatory skin disease that continues to impose significant physical, mental, and economic burdens on patients. Recent research has suggested the significant role of tissue-resident memory (TRM) cells in AD. However, the precise role and mechanisms of action of TRM cells in AD remain unclear. A deeper understanding of the involvement of TRM cells in AD will unveil promising pathways for future innovative therapeutic strategies. METHODS To investigate the involvement of TRM cells in AD, we used diverse mouse models and employed experimental techniques to manipulate cell formation and depletion. We assessed the inflammatory response by analyzing mouse ear phenotype, measuring ear thickness, and performing hematoxylin and eosin staining. Flow cytometry and immunofluorescence staining were used to identify different cell types and evaluate changes in cell quantity. Additionally, we used qPCR to analyze gene expression of relevant chemokines and cytokines. RESULTS Our study revealed the presence of TRM cells in the skin after exposure to calcipotriol. After a 24-h re-challenge, we observed substantial neutrophil infiltration into the previously irritated skin. Neutrophil depletion prior to re-challenge effectively prevented early flare-up responses during AD recurrence. Furthermore, we demonstrate that CD4+TRM cells upregulate expression of cytokines INF-γ and TNF-α, which may induce the expression of CXCL1, thereby recruiting neutrophils and contributing to the chronic recurrent inflammation observed in AD. CONCLUSIONS We have established a novel, chronic recurrent mouse model for investigating TRM cells in AD. Our findings demonstrate that CD4+TRM cells in the skin mediate early flare-up response during AD recurrence and influence the chronic recurrent inflammation of AD by recruiting neutrophils. Targeting CD4+TRM cells may represent a promising approach for the treatment of chronic recurrent inflammation in AD.
Collapse
Affiliation(s)
- Chunjiao Zheng
- Skin and Cosmetic Research Department, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ting Cao
- Skin and Cosmetic Research Department, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chengbin Ye
- Skin and Cosmetic Research Department, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Zou
- Skin and Cosmetic Research Department, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
84
|
Zhang P, Wang Y, Miao Q, Chen Y. The therapeutic potential of PD-1/PD-L1 pathway on immune-related diseases: Based on the innate and adaptive immune components. Biomed Pharmacother 2023; 167:115569. [PMID: 37769390 DOI: 10.1016/j.biopha.2023.115569] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/30/2023] Open
Abstract
Currently, immunotherapy targeting programmed cell death 1 (PD-1) or programmed death ligand 1 (PD-L1) has revolutionized the treatment strategy of human cancer patients. Meanwhile, PD-1/PD-L1 pathway has also been implicated in the pathogenesis of many immune-related diseases, such as autoimmune diseases, chronic infection diseases and adverse pregnancy outcomes, by regulating components of the innate and adaptive immune systems. Given the power of the new therapy, a better understanding of the regulatory effects of PD-1/PD-L1 pathway on innate and adaptive immune responses in immune-related diseases will facilitate the discovery of novel biomarkers and therapeutic drug targets. Targeting this pathway may successfully halt or potentially even reverse these pathological processes. In this review, we discuss recent major advances in PD-1/PD-L1 axis regulating innate and adaptive immune components in immune-related diseases. We reveal that the impact of PD-1/PD-L1 axis on the immune system is complex and manifold and multi-strategies on the targeted PD-1/PD-L1 axis are taken in the treatment of immune-related diseases. Consequently, targeting PD-1/PD-L1 pathway, alone or in combination with other treatments, may represent a novel strategy for future therapeutic intervention on immune-related diseases.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Yuting Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Qianru Miao
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Ying Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention (China Medical University), Ministry of Education, Shenyang 110122, Liaoning, China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
85
|
Santosa EK, Sun JC. Cardinal features of immune memory in innate lymphocytes. Nat Immunol 2023; 24:1803-1812. [PMID: 37828377 PMCID: PMC10998651 DOI: 10.1038/s41590-023-01607-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/28/2023] [Indexed: 10/14/2023]
Abstract
The ability of vertebrates to 'remember' previous infections had once been attributed exclusively to adaptive immunity. We now appreciate that innate lymphocytes also possess memory properties akin to those of adaptive immune cells. In this Review, we draw parallels from T cell biology to explore the key features of immune memory in innate lymphocytes, including quantity, quality, and location. We discuss the signals that trigger clonal or clonal-like expansion in innate lymphocytes, and highlight recent studies that shed light on the complex cellular and molecular crosstalk between metabolism, epigenetics, and transcription responsible for differentiating innate lymphocyte responses towards a memory fate. Additionally, we explore emerging evidence that activated innate lymphocytes relocate and establish themselves in specific peripheral tissues during infection, which may facilitate an accelerated response program akin to those of tissue-resident memory T cells.
Collapse
Affiliation(s)
- Endi K Santosa
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA
| | - Joseph C Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA.
| |
Collapse
|
86
|
Zemanek T, Nova Z, Nicodemou A. Tumor-Infiltrating Lymphocytes and Adoptive Cell Therapy: State of the Art in Colorectal, Breast and Lung Cancer. Physiol Res 2023; 72:S209-S224. [PMID: 37888965 PMCID: PMC10669950 DOI: 10.33549/physiolres.935155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/07/2023] [Indexed: 12/01/2023] Open
Abstract
Our knowledge of tumor-infiltrating lymphocytes (TILs) is dramatically expanding. These cells have proven prognostic and therapeutic value for many cancer outcomes and potential to treat also disseminated breast, colorectal, or lung cancer. However, the therapeutical outcome of TILs is negatively affected by tumor mutational burden and neoantigens. On the other hand, it can be improved in combination with checkpoint blockade therapy. This knowledge and rapid detection techniques alongside gene editing allow us to classify and modify T cells in many ways. Hence, to tailor them precisely to the patient´s needs as to program T cell receptors to recognize specific tumor-associated neoantigens and to insert them into lymphocytes or to select tumor neoantigen-specific T cells, for the development of vaccines that recognize tumor-specific antigens in tumors or metastases. Further studies and clinical trials in the field are needed for an even better-detailed understanding of TILs interactions and aiming in the fight against multiple cancers.
Collapse
Affiliation(s)
- T Zemanek
- Lambda Life, Bratislava, Slovak Republic.
| | | | | |
Collapse
|
87
|
Depew CE, McSorley SJ. The role of tissue resident memory CD4 T cells in Salmonella infection: Implications for future vaccines. Vaccine 2023; 41:6426-6433. [PMID: 37739887 DOI: 10.1016/j.vaccine.2023.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 07/20/2023] [Accepted: 09/05/2023] [Indexed: 09/24/2023]
Abstract
Salmonella infections cause a wide range of intestinal and systemic disease that affects global human health. While some vaccines are available, they do not mitigate the impact of Salmonella on endemic areas. Research using Salmonella mouse models has revealed the important role of CD4 T cells and antibody in the development of protective immunity against Salmonella infection. Recent work points to a critical role for hepatic tissue-resident memory lymphocytes in naturally acquired immunity to systemic infection. Thus, understanding the genesis and function of this Salmonella-specific population is an important objective and is the primary focus of this review. Greater understanding of how these memory lymphocytes contribute to bacterial elimination could suggest new approaches to vaccination against an important human pathogen.
Collapse
Affiliation(s)
- Claire E Depew
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA.
| | - Stephen J McSorley
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
88
|
Douguet L, Fert I, Lopez J, Vesin B, Le Chevalier F, Moncoq F, Authié P, Nguyen T, Noirat A, Névo F, Blanc C, Bourgine M, Hardy D, Anna F, Majlessi L, Charneau P. Full eradication of pre-clinical human papilloma virus-induced tumors by a lentiviral vaccine. EMBO Mol Med 2023; 15:e17723. [PMID: 37675835 PMCID: PMC10565635 DOI: 10.15252/emmm.202317723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023] Open
Abstract
Human papillomavirus (HPV) infections are the cause of all cervical and numerous oropharyngeal and anogenital cancers. The currently available HPV vaccines, which induce neutralizing antibodies, have no therapeutic effect on established tumors. Here, we developed an immuno-oncotherapy against HPV-induced tumors based on a non-integrative lentiviral vector encoding detoxified forms of the Early E6 and E7 oncoproteins of HPV16 and 18 genotypes, namely, "Lenti-HPV-07". A single intramuscular injection of Lenti-HPV-07 into mice bearing established HPV-induced tumors resulted in complete tumor eradication in 100% of the animals and was also effective against lung metastases. This effect correlated with CD8+ T-cell induction and profound remodeling of the tumor microenvironment. In the intra-tumoral infiltrates of vaccinated mice, the presence of large amounts of activated effector, resident memory, and transcription factor T cell factor-1 (TCF-1)+ "stem-like" CD8+ T cells was associated with full tumor eradication. The Lenti-HPV-07-induced immunity was long-lasting and prevented tumor growth after a late re-challenge, mimicking tumor relapse. Lenti-HPV-07 therapy synergizes with an anti-checkpoint inhibitory treatment and therefore shows promise as an immuno-oncotherapy against established HPV-mediated malignancies.
Collapse
Affiliation(s)
- Laëtitia Douguet
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Ingrid Fert
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Jodie Lopez
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Benjamin Vesin
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Fabien Le Chevalier
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Fanny Moncoq
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Pierre Authié
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Trang‐My Nguyen
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Amandine Noirat
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Fabien Névo
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Catherine Blanc
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Maryline Bourgine
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - David Hardy
- Histopathology Platform, Institut PasteurUniversité de ParisParisFrance
| | - François Anna
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Laleh Majlessi
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| | - Pierre Charneau
- Virology Department, Pasteur‐TheraVectys Joint Lab, Institut PasteurUniversité de ParisParisFrance
| |
Collapse
|
89
|
Rotrosen E, Kupper TS. Assessing the generation of tissue resident memory T cells by vaccines. Nat Rev Immunol 2023; 23:655-665. [PMID: 37002288 PMCID: PMC10064963 DOI: 10.1038/s41577-023-00853-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2023] [Indexed: 04/03/2023]
Abstract
Vaccines have been a hugely successful public health intervention, virtually eliminating many once common diseases of childhood. However, they have had less success in controlling endemic pathogens including Mycobacterium tuberculosis, herpesviruses and HIV. A focus on vaccine-mediated generation of neutralizing antibodies, which has been a successful approach for some pathogens, has been complicated by the emergence of escape variants, which has been seen for pathogens such as influenza viruses and SARS-CoV-2, as well as for HIV-1. We discuss how vaccination strategies aimed at generating a broad and robust T cell response may offer superior protection against pathogens, particularly those that have been observed to mutate rapidly. In particular, we consider here how a focus on generating resident memory T cells may be uniquely effective for providing immunity to pathogens that typically infect (or become reactivated in) the skin, respiratory mucosa or other barrier tissues.
Collapse
Affiliation(s)
- Elizabeth Rotrosen
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, USA
- Boston University School of Medicine, Boston, MA, USA
| | - Thomas S Kupper
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
90
|
Lyon De Ana C, Shenoy AT, Barker KA, Arafa EI, Etesami NS, Korkmaz FT, Soucy AM, Breen MP, Martin IMC, Tilton BR, Devarajan P, Crossland NA, Pihl RMF, Goltry WN, Belkina AC, Jones MR, Quinton LJ, Mizgerd JP. GL7 ligand expression defines a novel subset of CD4 + T RM cells in lungs recovered from pneumococcus. Mucosal Immunol 2023; 16:699-710. [PMID: 37604254 PMCID: PMC10591822 DOI: 10.1016/j.mucimm.2023.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/26/2023] [Indexed: 08/23/2023]
Abstract
Streptococcus pneumoniae is the most common etiology of bacterial pneumonia, one of the leading causes of death in children and the elderly worldwide. During non-lethal infections with S. pneumoniae, lymphocytes accumulate in the lungs and protect against reinfection with serotype-mismatched strains. Cluster of differentiation CD4+ resident memory T (TRM) cells are known to be crucial for this protection, but the diversity of lung CD4+ TRM cells has yet to be fully delineated. We aimed to identify unique subsets and their contributions to lung immunity. After recovery from pneumococcal infections, we identified a distinct subset of CD4+ T cells defined by the phenotype CD11ahiCD69+GL7+ in mouse lungs. Phenotypic analyses for markers of lymphocyte memory and residence demonstrated that GL7+ T cells are a subset of CD4+ TRM cells. Functional studies revealed that unlike GL7- TRM subsets that were mostly (RAR-related Orphan Receptor gamma T) RORγT+, GL7+ TRM cells exhibited higher levels of (T-box expressed in T cells) T-bet and Gata-3, corresponding with increased synthesis of interferon-γ, interleukin-13, and interleukin-5, inherent to both T helper 1 (TH1) and TH2 functions. Thus, we propose that these cells provide novel contributions during pneumococcal pneumonia, serving as important determinants of lung immunity.
Collapse
Affiliation(s)
- Carolina Lyon De Ana
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Virology, Immunology, & Microbiology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Anukul T Shenoy
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department. of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kimberly A Barker
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Virology, Immunology, & Microbiology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Emad I Arafa
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Neelou S Etesami
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Virology, Immunology, & Microbiology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Filiz T Korkmaz
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Alicia M Soucy
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Michael P Breen
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Virology, Immunology, & Microbiology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Ian M C Martin
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Brian R Tilton
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Priyadharshini Devarajan
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Nicholas A Crossland
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Riley M F Pihl
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Flow Cytometry Core Facility, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Wesley N Goltry
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Anna C Belkina
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Flow Cytometry Core Facility, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Matthew R Jones
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Lee J Quinton
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Virology, Immunology, & Microbiology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Virology, Immunology, & Microbiology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Medicine, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA; Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedesian School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
91
|
Berton RR, McGonagil PW, Jensen IJ, Ybarra TK, Bishop GA, Harty JT, Griffith TS, Badovinac VP. Sepsis leads to lasting changes in phenotype and function of naïve CD8 T cells. PLoS Pathog 2023; 19:e1011720. [PMID: 37824591 PMCID: PMC10597476 DOI: 10.1371/journal.ppat.1011720] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 10/24/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
Sepsis, an amplified immune response to systemic infection, is characterized by a transient cytokine storm followed by chronic immune dysfunction. Consequently, sepsis survivors are highly susceptible to newly introduced infections, suggesting sepsis can influence the function and composition of the naïve CD8 T cell pool and resulting pathogen-induced primary CD8 T cell responses. Here, we explored the extent to which sepsis induces phenotypic and functional changes within the naïve CD8 T cell pool. To interrogate this, the cecal ligation and puncture (CLP) mouse model of polymicrobial sepsis was used. In normal, non-septic mice, we show type-I interferon (IFN I)-mediated signaling plays an important role in driving the phenotypic and functional heterogeneity in the naïve CD8 T cell compartment leading to increased representation of Ly6C+ naïve CD8 T cells. In response to viral infection after sepsis resolution, naïve Ly6C+ CD8 T cells generated more primary effector and memory CD8 T cells with slower conversion to a central memory CD8 T cell phenotype (Tcm) than Ly6C- naïve CD8 T cells. Importantly, as a potent inducer of cytokine storm and IFN I production, sepsis leads to increased representation of Ly6C+ naïve CD8 T cells that maintained their heightened ability to respond (i.e., effector and memory CD8 T cell accumulation and cytokine production) to primary LCMV infection. Lastly, longitudinal analyses of peripheral blood samples obtained from septic patients revealed profound changes in CD8 T cell subset composition and frequency compared to healthy controls. Thus, sepsis has the capacity to alter the composition of naïve CD8 T cells, directly influencing primary CD8 T cell responses to newly introduced infections.
Collapse
Affiliation(s)
- Roger R. Berton
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Patrick W. McGonagil
- Department of Surgery, University of Iowa, Iowa City, Iowa, United States of America
| | - Isaac J. Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York City, New York, United States of America
| | - Tiffany K. Ybarra
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Gail A. Bishop
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - John T. Harty
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Thomas S. Griffith
- Department of Urology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota, United States of America
| | - Vladimir P. Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
92
|
Armitage E, Quan D, Flórido M, Palendira U, Triccas JA, Britton WJ. CXCR3 Provides a Competitive Advantage for Retention of Mycobacterium tuberculosis-Specific Tissue-Resident Memory T Cells Following a Mucosal Tuberculosis Vaccine. Vaccines (Basel) 2023; 11:1549. [PMID: 37896952 PMCID: PMC10611282 DOI: 10.3390/vaccines11101549] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Mycobacterium tuberculosis is a major human pathogen, and new vaccines are needed to prevent transmission. Mucosal vaccination may confer protection against M. tuberculosis by stimulating tissue-resident memory (TRM) CD4+ T cells in the lungs. The chemokine receptor CXCR3 promotes lung recruitment of T cells, but its role in TRM development is unknown. This study demonstrates the recombinant influenza A virus vaccine PR8.p25, expressing the immunodominant M. tuberculosis T cell epitope p25, induces CXCR3 expression on p25-specific CD4+ T cells in the lungs so that the majority of vaccine-induced CD4+ TRM expresses CXCR3 at 6 weeks. However, CXCR3-/- mice developed equivalent antigen-specific CD4+ T cell responses to wild-type (WT) mice following PR8.p25, and surprisingly retained more p25-specific CD4+ TRM in the lungs than WT mice at 6 weeks. The adoptive transfer of CXCR3-/- and WT P25 T cells into WT mice revealed that the initial recruitment of vaccine-induced CD4+ T cells into the lungs was independent of CXCR3, but by 6 weeks, CXCR3-deficient P25 T cells, and especially CXCR3-/- TRM, were significantly reduced compared to CXCR3-sufficient P25 T cells. Therefore, although CXCR3 was not essential for CD4+ TRM recruitment or retention, it provided a competitive advantage for the induction of M. tuberculosis-specific CD4+ TRM in the lungs following pulmonary immunization.
Collapse
Affiliation(s)
- Ellis Armitage
- Centenary Institute, The University of Sydney, Sydney, NSW 2006, Australia; (E.A.); (D.Q.); (M.F.); (U.P.)
| | - Diana Quan
- Centenary Institute, The University of Sydney, Sydney, NSW 2006, Australia; (E.A.); (D.Q.); (M.F.); (U.P.)
| | - Manuela Flórido
- Centenary Institute, The University of Sydney, Sydney, NSW 2006, Australia; (E.A.); (D.Q.); (M.F.); (U.P.)
| | - Umaimainthan Palendira
- Centenary Institute, The University of Sydney, Sydney, NSW 2006, Australia; (E.A.); (D.Q.); (M.F.); (U.P.)
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
| | - James A. Triccas
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
- The University of Sydney Infectious Diseases Institute (Sydney ID), Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Warwick J. Britton
- Centenary Institute, The University of Sydney, Sydney, NSW 2006, Australia; (E.A.); (D.Q.); (M.F.); (U.P.)
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| |
Collapse
|
93
|
Clatch A, Christo SN, Mackay LK. T RM cells: not born this way. Trends Immunol 2023; 44:663-664. [PMID: 37591711 DOI: 10.1016/j.it.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/19/2023]
Abstract
Human tissue-resident memory T (TRM) cells seeded early in life undergo an age-associated functional maturation and residency acquisition throughout childhood.
Collapse
Affiliation(s)
- Allison Clatch
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Susan N Christo
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Laura K Mackay
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| |
Collapse
|
94
|
Collier ARY, Modest AM, Aguayo RA, Bondzie EA, Patel S, Hacker MR, Barouch DH. Altered Cytokine Production in Human Intervillous Blood T Cells in Preeclampsia. Reprod Sci 2023; 30:2655-2664. [PMID: 36749459 PMCID: PMC10404629 DOI: 10.1007/s43032-023-01165-4] [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: 09/19/2022] [Accepted: 12/28/2022] [Indexed: 02/08/2023]
Abstract
Conventional and regulatory T cells (Treg) are dynamic mediators of maternal immune tolerance to the developing feto-placental unit. Functional evaluation of T cells at the maternal-fetal interface is crucial to elucidate the immunologic basis of obstetric complications. Our objective was to define the T cell phenotype and function of uterine intervillous blood (IVB) in pregnancy with and without preeclampsia. We hypothesize that preeclampsia is associated with impaired immune tolerance and a pro-inflammatory uterine T cell microenvironment. In this cross-sectional study, maternal peripheral blood (PB) and uterine IVB (obtained from the surgical sponge used to clean the placental bed during cesarean delivery) were collected from participants with and without preeclampsia. Proportion, activation, and cytokine production of T cell subsets were quantified by flow cytometry. T cell parameters were compared by tissue source and by preeclampsia status. Sixty participants, 26 with preeclampsia, were included. Induced Treg made up a greater proportion of IVB T cells compared to PB and had greater cytokine-producing capacity. Preeclampsia was associated with increased ratio of pro-inflammatory IL-17α to suppressive IL-10 cytokine production by CD4 T cell subsets in IVB, but not in PB. Human uterine IVB is composed of activated, cytokine-producing T cell subsets distinct from maternal PB. Preeclampsia is associated with a pro-inflammatory IVB profile, with increased IL-17α /IL-10 ratio in all CD4 T cell subsets. IVB sampling is a useful tool for investigating human T cell biology at the maternal-fetal interface that may inform immunotherapeutic strategies for preeclampsia.
Collapse
Affiliation(s)
- Ai-Ris Y Collier
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Obstetrics and Gynecology, Kirstein 3rd floor, Boston, MA, 02215, USA.
- Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA.
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Anna M Modest
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Obstetrics and Gynecology, Kirstein 3rd floor, Boston, MA, 02215, USA
- Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | - Ricardo A Aguayo
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Obstetrics and Gynecology, Kirstein 3rd floor, Boston, MA, 02215, USA
| | - Esther A Bondzie
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Shivani Patel
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michele R Hacker
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Obstetrics and Gynecology, Kirstein 3rd floor, Boston, MA, 02215, USA
- Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
95
|
Reina-Campos M, Heeg M, Kennewick K, Mathews IT, Galletti G, Luna V, Nguyen Q, Huang H, Milner JJ, Hu KH, Vichaidit A, Santillano N, Boland BS, Chang JT, Jain M, Sharma S, Krummel MF, Chi H, Bensinger SJ, Goldrath AW. Metabolic programs of T cell tissue residency empower tumour immunity. Nature 2023; 621:179-187. [PMID: 37648857 PMCID: PMC11238873 DOI: 10.1038/s41586-023-06483-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 07/26/2023] [Indexed: 09/01/2023]
Abstract
Tissue resident memory CD8+ T (TRM) cells offer rapid and long-term protection at sites of reinfection1. Tumour-infiltrating lymphocytes with characteristics of TRM cells maintain enhanced effector functions, predict responses to immunotherapy and accompany better prognoses2,3. Thus, an improved understanding of the metabolic strategies that enable tissue residency by T cells could inform new approaches to empower immune responses in tissues and solid tumours. Here, to systematically define the basis for the metabolic reprogramming supporting TRM cell differentiation, survival and function, we leveraged in vivo functional genomics, untargeted metabolomics and transcriptomics of virus-specific memory CD8+ T cell populations. We found that memory CD8+ T cells deployed a range of adaptations to tissue residency, including reliance on non-steroidal products of the mevalonate-cholesterol pathway, such as coenzyme Q, driven by increased activity of the transcription factor SREBP2. This metabolic adaptation was most pronounced in the small intestine, where TRM cells interface with dietary cholesterol and maintain a heightened state of activation4, and was shared by functional tumour-infiltrating lymphocytes in diverse tumour types in mice and humans. Enforcing synthesis of coenzyme Q through deletion of Fdft1 or overexpression of PDSS2 promoted mitochondrial respiration, memory T cell formation following viral infection and enhanced antitumour immunity. In sum, through a systematic exploration of TRM cell metabolism, we reveal how these programs can be leveraged to fuel memory CD8+ T cell formation in the context of acute infections and enhance antitumour immunity.
Collapse
Affiliation(s)
- Miguel Reina-Campos
- School of Biological Sciences, Department of Molecular Biology, University of California, San Diego, San Diego, CA, USA
| | - Maximilian Heeg
- School of Biological Sciences, Department of Molecular Biology, University of California, San Diego, San Diego, CA, USA
| | - Kelly Kennewick
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ian T Mathews
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Giovanni Galletti
- School of Biological Sciences, Department of Molecular Biology, University of California, San Diego, San Diego, CA, USA
| | - Vida Luna
- School of Biological Sciences, Department of Molecular Biology, University of California, San Diego, San Diego, CA, USA
| | - Quynhanh Nguyen
- School of Biological Sciences, Department of Molecular Biology, University of California, San Diego, San Diego, CA, USA
| | - Hongling Huang
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - J Justin Milner
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Kenneth H Hu
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Amy Vichaidit
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Natalie Santillano
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Brigid S Boland
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - John T Chang
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Mohit Jain
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Sonia Sharma
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Steven J Bensinger
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ananda W Goldrath
- School of Biological Sciences, Department of Molecular Biology, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
96
|
Heim TA, Schultz AC, Delclaux I, Cristaldi V, Churchill MJ, Lund AW. Lymphatic vessel transit seeds precursors to cytotoxic resident memory T cells in skin draining lymph nodes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.29.555369. [PMID: 37693469 PMCID: PMC10491166 DOI: 10.1101/2023.08.29.555369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Resident memory T cells (TRM) provide rapid, localized protection in peripheral tissues to pathogens and cancer. While TRM are also found in lymph nodes (LN), how they develop during primary infection and their functional significance remains largely unknown. Here, we track the anatomical distribution of anti-viral CD8+ T cells as they simultaneously seed skin and LN TRM using a model of skin infection with restricted antigen distribution. We find exquisite localization of LN TRM to the draining LN of infected skin. LN TRM formation depends on lymphatic transport and specifically egress of effector CD8+ T cells that appear poised for residence as early as 12 days post infection. Effector CD8+ T cell transit through skin is necessary and sufficient to populate LN TRM in draining LNs, a process reinforced by antigen encounter in skin. Importantly, we demonstrate that LN TRM are sufficient to provide protection against pathogenic rechallenge. These data support a model whereby a subset of tissue infiltrating CD8+ T cells egress during viral clearance, and establish regional protection in the draining lymphatic basin as a mechanism to prevent pathogen spread.
Collapse
Affiliation(s)
- Taylor A. Heim
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Austin C. Schultz
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Ines Delclaux
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Vanessa Cristaldi
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Madeline J. Churchill
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR
| | - Amanda W. Lund
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
97
|
Gordon H, Rodger B, Lindsay JO, Stagg AJ. Recruitment and Residence of Intestinal T Cells - Lessons for Therapy in Inflammatory Bowel Disease. J Crohns Colitis 2023; 17:1326-1341. [PMID: 36806613 DOI: 10.1093/ecco-jcc/jjad027] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Indexed: 02/23/2023]
Abstract
Targeting leukocyte trafficking in the management of inflammatory bowel disease [IBD] has been a significant therapeutic advance over the past 15 years. However, as with other advanced therapies, phase III clinical trials report response to trafficking inhibitors in only a proportion of patients, with fewer achieving clinical remission or mucosal healing. Additionally, there have been significant side effects, most notably progressive multifocal leukoencephalopathy in association with the α4 inhibitor natalizumab. This article reviews the mechanisms underpinning T cell recruitment and residence, to provide a background from which the strength and limitations of agents that disrupt leukocyte trafficking can be further explored. The therapeutic impact of trafficking inhibitors is underpinned by the complexity and plasticity of the intestinal immune response. Pathways essential for gut homing in health may be bypassed in the inflamed gut, thus providing alternative routes of entry when conventional homing molecules are targeted. Furthermore, there is conservation of trafficking architecture between proinflammatory and regulatory T cells. The persistence of resident memory cells within the gut gives rise to local established pro-inflammatory populations, uninfluenced by inhibition of trafficking. Finally, trafficking inhibitors may give rise to effects beyond the intended response, such as the impact of vedolizumab on innate immunity, as well as on target side effects. With significant research efforts into predictive biomarkers already underway, it is ultimately hoped that a better understanding of trafficking and residence will help us predict which patients are most likely to respond to inhibition of leukocyte trafficking, and how best to combine therapies.
Collapse
Affiliation(s)
- Hannah Gordon
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine, Barts & The London Medical School, Queen Mary University of London, London, UK
- Department of Gastroenterology, Barts Health NHS Trust, London, UK
| | - Beverley Rodger
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine, Barts & The London Medical School, Queen Mary University of London, London, UK
| | - James O Lindsay
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine, Barts & The London Medical School, Queen Mary University of London, London, UK
- Department of Gastroenterology, Barts Health NHS Trust, London, UK
| | - Andrew J Stagg
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine, Barts & The London Medical School, Queen Mary University of London, London, UK
| |
Collapse
|
98
|
Connors TJ, Matsumoto R, Verma S, Szabo PA, Guyer R, Gray J, Wang Z, Thapa P, Dogra P, Poon MML, Rybkina K, Bradley MC, Idzikowski E, McNichols J, Kubota M, Pethe K, Shen Y, Atkinson MA, Brusko M, Brusko TM, Yates AJ, Sims PA, Farber DL. Site-specific development and progressive maturation of human tissue-resident memory T cells over infancy and childhood. Immunity 2023; 56:1894-1909.e5. [PMID: 37421943 PMCID: PMC10527943 DOI: 10.1016/j.immuni.2023.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/23/2023] [Accepted: 06/13/2023] [Indexed: 07/10/2023]
Abstract
Infancy and childhood are critical life stages for generating immune memory to protect against pathogens; however, the timing, location, and pathways for memory development in humans remain elusive. Here, we investigated T cells in mucosal sites, lymphoid tissues, and blood from 96 pediatric donors aged 0-10 years using phenotypic, functional, and transcriptomic profiling. Our results revealed that memory T cells preferentially localized in the intestines and lungs during infancy and accumulated more rapidly in mucosal sites compared with blood and lymphoid organs, consistent with site-specific antigen exposure. Early life mucosal memory T cells exhibit distinct functional capacities and stem-like transcriptional profiles. In later childhood, they progressively adopt proinflammatory functions and tissue-resident signatures, coincident with increased T cell receptor (TCR) clonal expansion in mucosal and lymphoid sites. Together, our findings identify staged development of memory T cells targeted to tissues during the formative years, informing how we might promote and monitor immunity in children.
Collapse
Affiliation(s)
- Thomas J Connors
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Rei Matsumoto
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Shivali Verma
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peter A Szabo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rebecca Guyer
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Joshua Gray
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Zicheng Wang
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Puspa Thapa
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Pranay Dogra
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Maya M L Poon
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ksenia Rybkina
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Marissa C Bradley
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Emma Idzikowski
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - James McNichols
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Masaru Kubota
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kalpana Pethe
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Maigan Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Todd M Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
99
|
Zhu W, Park J, Pho T, Wei L, Dong C, Kim J, Ma Y, Champion JA, Wang BZ. ISCOMs/MPLA-Adjuvanted SDAD Protein Nanoparticles Induce Improved Mucosal Immune Responses and Cross-Protection in Mice. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301801. [PMID: 37162451 PMCID: PMC10524461 DOI: 10.1002/smll.202301801] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/20/2023] [Indexed: 05/11/2023]
Abstract
The epidemics caused by the influenza virus are a serious threat to public health and the economy. Adding appropriate adjuvants to improve immunogenicity and finding effective mucosal vaccines to combat respiratory infection at the portal of virus entry are important strategies to boost protection. In this study, a novel type of core/shell protein nanoparticle consisting of influenza nucleoprotein (NP) as the core and NA1-M2e or NA2-M2e fusion proteins as the coating antigens by SDAD hetero-bifunctional crosslinking is exploited. Immune-stimulating complexes (ISCOMs)/monophosphoryl lipid A (MPLA) adjuvants further boost the NP/NA-M2e SDAD protein nanoparticle-induced immune responses when administered intramuscularly. The ISCOMs/MPLA-adjuvanted protein nanoparticles are delivered through the intranasal route to validate the application as mucosal vaccines. ISCOMs/MPLA-adjuvanted nanoparticles induce significantly strengthened antigen-specific antibody responses, cytokine-secreting splenocytes in the systemic compartment, and higher levels of antigen-specific IgA and IgG in the local mucosa. Meanwhile, significantly expanded lung resident memory (RM) T and B cells (TRM /BRM ) and alveolar macrophages population are observed in ISCOMs/MPLA-adjuvanted nanoparticle-immunized mice with a 100% survival rate after homogeneous and heterogeneous H3N2 viral challenges. Taken together, ISCOMs/MPLA-adjuvanted protein nanoparticles could improve strong systemic and mucosal immune responses conferring protection in different immunization routes.
Collapse
Affiliation(s)
- Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Jaeyoung Park
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Thomas Pho
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Bioengineering Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Joo Kim
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Julie A. Champion
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Bioengineering Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
100
|
Li H, Zhou Y, Guo X, Zhang Q, Ding X. The effects of irreversible electroporation triggering anti-tumor immunity and the value of its combination with immunotherapy. J Interv Med 2023; 6:107-110. [PMID: 37846332 PMCID: PMC10577062 DOI: 10.1016/j.jimed.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/08/2023] [Accepted: 07/08/2023] [Indexed: 10/18/2023] Open
Abstract
Recently, interventional ablation techniques have gained prominence in tumor treatment guidelines and complement traditional approaches, such as surgery, chemotherapy, and radiotherapy. Conventional ablation techniques, such as microwave, radiofrequency, and cryoablation, have been used; however, they have certain limitations, including the risk of damaging surrounding normal tissues and the heat sink effect caused by tumor blood flow.1 Irreversible electroporation (IRE), an ablation technology independent of thermal energy, is a promising alternative.2 Clinical studies have demonstrated IRE's efficacy in treating tumors, such as pancreatic and liver tumors.3 Recent research has shown that IRE can elicit specific anti-tumor immune responses in the body.5 IRE also plays a crucial role in eliminating residual tumor cells postoperatively and preventing tumor recurrence.
Collapse
Affiliation(s)
- Hengyu Li
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Yu Zhou
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Xiaoxia Guo
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Qiwei Zhang
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Xiaoyi Ding
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| |
Collapse
|