1
|
Seo ES, Lee SK, Son YM. Multifaceted functions of tissue-resident memory T cells in tumorigenesis and cancer immunotherapy. Cancer Immunol Immunother 2025; 74:184. [PMID: 40285796 PMCID: PMC12033165 DOI: 10.1007/s00262-025-04035-x] [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: 12/02/2024] [Accepted: 03/24/2025] [Indexed: 04/29/2025]
Abstract
Tissue-resident memory T (TRM) cells are well reported as a strong protective first line of defense against foreign antigens in non-lymphoid tissues. Moreover, TRM cells have demonstrated critical protective roles in antitumor immunity, contributing to enhanced survival and tumor growth inhibition across various cancer types. However, surprisingly, recent studies suggest that TRM cells can exhibit paradoxical effects, potentially promoting tumor progression under certain conditions and leading to adverse outcomes during antitumor immune responses. Understanding the complexities of TRM cell functions will enable us to harness their potential in advancing cancer immunotherapy more effectively. Therefore, this review comprehensively investigates the dual roles of TRM cells in different tumor contexts, highlighting their protective functions in combating cancers and their unfavorable potential to exacerbate tumor development. Additionally, we explore the implications of TRM cell behaviors for future cancer treatment strategies, emphasizing the need for further research to optimize the therapeutic exploitation of TRM cells while mitigating their deleterious effects.
Collapse
Affiliation(s)
- Eun Sang Seo
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Sung-Kyu Lee
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Young Min Son
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea.
| |
Collapse
|
2
|
Xu S, Chen H, Tan A, Yang H, Tong J. Surgical Stress and Non-Surgery Related Stress Synergistically Trigger Meningeal CD8 + T Cells Accumulation and Subsequent Brain Dysfunction in Mice. J Neurochem 2025; 169:e70043. [PMID: 40066934 DOI: 10.1111/jnc.70043] [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: 11/26/2024] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 05/13/2025]
Abstract
Patients are usually highly stressed during the immediate perioperative period. It remains unclear whether increased stress contributes to postoperative brain dysfunction. Here, the clinical perioperative setting was mimicked via exploratory laparotomy and perioperative restraint stress. The stress response was assessed by measuring the levels of stress hormones and IL-6 in peripheral blood. Behaviors were evaluated with the open field, novel object recognition, and Barnes maze tests. We found that surgical stress and non-surgery-related stress synergistically trigger meningeal CD8+T cell accumulation, brain dysfunction, and increased stress hormone and IL-6 levels in the peripheral blood of adult mice, but simple surgical stress or non-surgery-related stress had no significant effect on these parameters. Limiting meningeal CD8+ T cell accumulation with an anti-CD8 antibody alleviated the impact of surgery plus perioperative stress on brain function, neuroinflammation, and neurogenesis. The partial elimination of microglia before surgery alleviated postoperative meningeal CD8+ T cell accumulation, cognitive dysfunction, and decreased hippocampal chemotactic factor levels. Our findings indicate that the synergistic effect of surgical stress and non-surgery-related stress contributes to postoperative brain dysfunction by triggering meningeal CD8+ T cell accumulation, suggesting the potential of limiting non-surgery-related stress as a preventive method for postoperative brain dysfunction.
Collapse
Affiliation(s)
- Shanqing Xu
- Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Brain Research Center, Central South University, Changsha, Hunan, China
| | - Hui Chen
- Department of Clinical Laboratory, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Aihua Tan
- Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Brain Research Center, Central South University, Changsha, Hunan, China
| | - Hui Yang
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Jianbin Tong
- Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Brain Research Center, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Xie D, Lu G, Mai G, Guo Q, Xu G. Tissue-resident memory T cells in diseases and therapeutic strategies. MedComm (Beijing) 2025; 6:e70053. [PMID: 39802636 PMCID: PMC11725047 DOI: 10.1002/mco2.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Tissue-resident memory T (TRM) cells are crucial components of the immune system that provide rapid, localized responses to recurrent pathogens at mucosal and epithelial barriers. Unlike circulating memory T cells, TRM cells are located within peripheral tissues, and they play vital roles in antiviral, antibacterial, and antitumor immunity. Their unique retention and activation mechanisms, including interactions with local epithelial cells and the expression of adhesion molecules, enable their persistence and immediate functionality in diverse tissues. Recent advances have revealed their important roles in chronic inflammation, autoimmunity, and cancer, illuminating both their protective and their pathogenic potential. This review synthesizes current knowledge on TRM cells' molecular signatures, maintenance pathways, and functional dynamics across different tissues. We also explore the interactions of TRM cells with other immune cells, such as B cells, macrophages, and dendritic cells, highlighting the complex network that underpins the efficacy of TRM cells in immune surveillance and response. Understanding the nuanced regulation of TRM cells is essential for developing targeted therapeutic strategies, including vaccines and immunotherapies, to enhance their protective roles while mitigating adverse effects. Insights into TRM cells' biology hold promise for innovative treatments for infectious diseases, cancer, and autoimmune conditions.
Collapse
Affiliation(s)
- Daoyuan Xie
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Guanting Lu
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Gang Mai
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Qiuyan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, Institute of Chinese Materia MedicaAcademy of Chinese Medical SciencesBeijingChina
| | - Guofeng Xu
- Inflammation & Allergic Diseases Research UnitThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
4
|
Chen Y, Man-Tak Chu J, Liu JX, Duan YJ, Liang ZK, Zou X, Wei M, Xin WJ, Xu T, Tin-Chun Wong G, Feng X. Double negative T cells promote surgery-induced neuroinflammation, microglial engulfment and cognitive dysfunction via the IL-17/CEBPβ/C3 pathway in adult mice. Brain Behav Immun 2025; 123:965-981. [PMID: 39491565 DOI: 10.1016/j.bbi.2024.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/11/2024] [Accepted: 10/26/2024] [Indexed: 11/05/2024] Open
Abstract
CD3(+) CD4(-) CD8(-) double negative T cells (DNTs) manifest themselves in autoimmune diseases and associated inflammation. In the central nervous system, the increased presence of DNTs is associated with the progression of neurological conditions and brain injury. Active DNTs that produce IL-17 have been regarded as a pro-inflammatory phenotype. The IL-17 signaling pathway mediates neuroinflammatory responses by inducing glial activation and producing inflammatory factors. Neuroinflammation is considered integral to the pathogenesis of perioperative neurocognitive disorders (PNDs), commonly developed after surgery in susceptible patients. We and others have demonstrated a significant role for complement C3 in surgery-induced neuroinflammation and cognitive impairment but the regulatory mechanisms for this remain unexplored. We hypothesized that surgery induces DNT infiltration into the CNS that in turn upregulates complement C3 expression and this causes changes that contribute to cognitive impairment. Using an adult murine abdominal surgery model, we investigated perioperative changes in cognitive performance, quantifying the presence of T cell subsets and phenotype, IL-17 signaling pathway activation, glial cell activation and C3 expression in the brain. Postoperative IL-17 specific inhibitor GSK2981278 administration or preoperatively conditional CEBPβ knock-down by AAV9 viral vector were then applied to evaluate the effect of inhibiting IL-17 signaling pathway on postoperative C3 expression and cognitive performance. The results showed an increased hippocampus infiltration of DNTs with augmented IL-17 production, along with C3 upregulation and cognitive impairment. Both inhibition of IL-17 or knock-down of CEBPβ significantly suppressed C3 expression, synaptic engulfment by microglia and attenuated cognitive impairment. These findings indicate that DNTs promote postoperative neuroinflammation and cognitive impairment via the IL-17/CEBPβ/C3 pathway and targeting this IL-17 axis could be a potential therapeutic strategy to ameliorate postoperative neuroinflammation and cognitive impairment.
Collapse
Affiliation(s)
- Ying Chen
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - John Man-Tak Chu
- Department of Anaesthesiology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Room K424, 4Th Floor, Block K, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
| | - Jia-Xin Liu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yu-Juan Duan
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zheng-Kai Liang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xin Zou
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ming Wei
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Wen-Jun Xin
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ting Xu
- Neuroscience Program, Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| | - Gordon Tin-Chun Wong
- Department of Anaesthesiology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Room K424, 4Th Floor, Block K, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China.
| | - Xia Feng
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
5
|
Lagache L, Zirem Y, Le Rhun É, Fournier I, Salzet M. Predicting Protein Pathways Associated to Tumor Heterogeneity by Correlating Spatial Lipidomics and Proteomics: The Dry Proteomic Concept. Mol Cell Proteomics 2025; 24:100891. [PMID: 39644924 PMCID: PMC11773152 DOI: 10.1016/j.mcpro.2024.100891] [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: 05/09/2024] [Revised: 11/20/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024] Open
Abstract
Prediction of proteins and associated biological pathways from lipid analyses via matrix-assisted laser desorption/ionization (MALDI) MSI is a pressing challenge. We introduced "dry proteomics," using MALDI MSI to validate spatial localization of identified optimal clusters in lipid imaging. Consistent cluster appearance across omics images suggests association with specific lipid and protein in distinct biological pathways, forming the basis of dry proteomics. The methodology was refined using rat brain tissue as a model, then applied to human glioblastoma, a highly heterogeneous cancer. Sequential tissue sections underwent omics MALDI MSI and unsupervised clustering. Spatial omics analysis facilitated lipid and protein characterization, leading to a predictive model identifying clusters in any tissue based on unique lipid signatures and predicting associated protein pathways. Application to rat brain slices revealed diverse tissue subpopulations, including successfully predicted cerebellum areas. Similarly, the methodology was applied to a dataset from a cohort of 50 glioblastoma patients, reused from a previous study. However, among the 50 patients, only 13 lipid signatures from MALDI MSI data were available, allowing for the identification of lipid-protein associations that correlated with patient prognosis. For cases lacking lipid imaging data, a classification model based on protein data was developed from dry proteomic results to effectively categorize the remaining cohort.
Collapse
Affiliation(s)
- Laurine Lagache
- Univ.Lille, Inserm, CHU Lille, U1192 - Proteomics Inflammatory Response Mass Spectrometry- PRISM, Lille, France
| | - Yanis Zirem
- Univ.Lille, Inserm, CHU Lille, U1192 - Proteomics Inflammatory Response Mass Spectrometry- PRISM, Lille, France
| | - Émilie Le Rhun
- Univ.Lille, Inserm, CHU Lille, U1192 - Proteomics Inflammatory Response Mass Spectrometry- PRISM, Lille, France; Department of Neurosurgery and Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Isabelle Fournier
- Univ.Lille, Inserm, CHU Lille, U1192 - Proteomics Inflammatory Response Mass Spectrometry- PRISM, Lille, France; Department Institut Universitaire de France, Ministère de l'Enseignement supérieur, de la Recherche et de l'Innovation, Paris, France.
| | - Michel Salzet
- Univ.Lille, Inserm, CHU Lille, U1192 - Proteomics Inflammatory Response Mass Spectrometry- PRISM, Lille, France; Department Institut Universitaire de France, Ministère de l'Enseignement supérieur, de la Recherche et de l'Innovation, Paris, France.
| |
Collapse
|
6
|
Musial SC, Kleist SA, Degefu HN, Ford MA, Chen T, Isaacs JF, Boussiotis VA, Skorput AGJ, Rosato PC. Alarm Functions of PD-1+ Brain-Resident Memory T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1585-1594. [PMID: 39413000 PMCID: PMC11647767 DOI: 10.4049/jimmunol.2400295] [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: 06/03/2024] [Accepted: 09/27/2024] [Indexed: 10/18/2024]
Abstract
Resident memory T cells (TRM cells) have been described in barrier tissues as having a "sensing and alarm" function where, upon sensing cognate Ag, they alarm the surrounding tissue and orchestrate local recruitment and activation of immune cells. In the immunologically unique and tightly restricted CNS, it remains unclear whether and how brain TRM cells, which express the inhibitory receptor programmed cell death protein 1 (PD-1), alarm the surrounding tissue during Ag re-encounter. Using mouse models, we reveal that TRM cells are sufficient to drive the rapid remodeling of the brain immune landscape through activation of microglia, dendritic cells, NK cells, and B cells, expansion of regulatory T cells, and recruitment of macrophages and monocytic dendritic cells. Moreover, we report that although PD-1 restrained granzyme B upregulation in brain TRM cells reactivated via viral peptide, we observed no apparent effect on cytotoxicity in vivo, or downstream alarm responses within 48 h of TRM reactivation. We conclude that TRM cells are sufficient to trigger rapid immune activation and recruitment in the CNS and may have an unappreciated role in driving neuroinflammation.
Collapse
Affiliation(s)
- Shawn C. Musial
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Sierra A. Kleist
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Hanna N. Degefu
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Myles A. Ford
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Tiffany Chen
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Jordan F. Isaacs
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Vassiliki A. Boussiotis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | | | - Pamela C. Rosato
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| |
Collapse
|
7
|
Harkins AL, Ambegaokar PP, Keeler AM. Immune responses to central nervous system directed adeno-associated virus gene therapy: Does direct CNS delivery make a difference? Neurotherapeutics 2024; 21:e00435. [PMID: 39180957 PMCID: PMC11386282 DOI: 10.1016/j.neurot.2024.e00435] [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: 04/04/2024] [Revised: 07/11/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
Adeno-associated virus (AAV) mediated gene therapy is a leading gene delivery platform with potential to transform the landscape of treatment for neurological disorders. While AAV is deemed non-immunogenic compared to other viral vectors, adverse immune reactions have been observed in the clinic, raising concerns. As the central nervous system (CNS) has a tightly regulated immune system, characterized by a degree of tolerance, it has been considered a unique target for AAV gene therapy. AAV vectors have shown promising results for the treatment of several CNS disorders including Spinal Muscular Atrophy, Giant Axonal Neuropathy, Amyotrophic Lateral Sclerosis, Tay Sachs Disease, Parkinson's Disease, and others, demonstrating safety and success. The Food and Drug Administration (FDA) approval of Zolgensma and European Medicines Agency (EMA) approval of Upstaza, for Spinal Muscular Atrophy (SMA) and Aromatic l-amino acid decarboxylase deficiency (AADC) respectively, represent this success, all while highlighting significant differences in immune responses to AAV, particularly with regards to therapeutic administration route. AAV therapies like Upstaza that are injected directly into the immune-specialized brain have been characterized by mild immune response profiles and minor adverse events, whereas therapies like Zolgensma that are injected systemically demonstrate more robust immune stimulation and off-target toxicities. Despite these contrasting parallels, these therapeutics and others in the clinic have demonstrated clinical benefit for patients, warranting further exploration of immune responses to CNS-directed AAV clinical trials. Thus, in this review, we discuss effects of different routes of AAV administration on eliciting local and peripheral immune responses specifically observed in CNS-targeted trials.
Collapse
Affiliation(s)
- Ashley L Harkins
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, United States; Department of Neurology, University of Massachusetts Chan Medical School, United States; Horae Gene Therapy Center, University of Massachusetts Chan Medical School, United States
| | - Prajakta P Ambegaokar
- Graduate Program in Translational Science, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, United States; Horae Gene Therapy Center, University of Massachusetts Chan Medical School, United States
| | - Allison M Keeler
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, United States; Graduate Program in Translational Science, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, United States; NeuroNexus Institute, University of Massachusetts Chan Medical School, United States; Department of Pediatrics, University of Massachusetts Chan Medical School, United States; Horae Gene Therapy Center, University of Massachusetts Chan Medical School, United States.
| |
Collapse
|
8
|
Ghosh Roy S, Karim AF, Brumeanu TD, Casares SA. Reconstitution of human microglia and resident T cells in the brain of humanized DRAGA mice. Front Cell Infect Microbiol 2024; 14:1367566. [PMID: 38983114 PMCID: PMC11231403 DOI: 10.3389/fcimb.2024.1367566] [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: 01/08/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024] Open
Abstract
Humanized mouse models are valuable tools for investigating the human immune system in response to infection and injury. We have previously described the human immune system (HIS)-DRAGA mice (HLA-A2.HLA-DR4.Rag1KO.IL-2RgKO.NOD) generated by infusion of Human Leukocyte Antigen (HLA)-matched, human hematopoietic stem cells from umbilical cord blood. By reconstituting human cells, the HIS-DRAGA mouse model has been utilized as a "surrogate in vivo human model" for infectious diseases such as Human Immunodeficiency Virus (HIV), Influenza, Coronavirus Disease 2019 (COVID-19), scrub typhus, and malaria. This humanized mouse model bypasses ethical concerns about the use of fetal tissues for the humanization of laboratory animals. Here in, we demonstrate the presence of human microglia and T cells in the brain of HIS-DRAGA mice. Microglia are brain-resident macrophages that play pivotal roles against pathogens and cerebral damage, whereas the brain-resident T cells provide surveillance and defense against infections. Our findings suggest that the HIS-DRAGA mouse model offers unique advantages for studying the functions of human microglia and T cells in the brain during infections, degenerative disorders, tumors, and trauma, as well as for testing therapeutics in these pathological conditions.
Collapse
Affiliation(s)
- Sounak Ghosh Roy
- Agile Vaccines & Therapeutics, Defense Infectious Diseases Directorate, Naval Medical Research Command, Silver Spring, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Ahmad F. Karim
- Agile Vaccines & Therapeutics, Defense Infectious Diseases Directorate, Naval Medical Research Command, Silver Spring, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Teodor-D. Brumeanu
- Department of Medicine, Division of Immunology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Sofia A. Casares
- Agile Vaccines & Therapeutics, Defense Infectious Diseases Directorate, Naval Medical Research Command, Silver Spring, MD, United States
| |
Collapse
|
9
|
Musial SC, Kleist SA, Degefu HN, Ford MA, Chen T, Isaacs JF, Boussiotis VA, Skorput AGJ, Rosato PC. Alarm functions of PD-1+ brain resident memory T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597370. [PMID: 38895249 PMCID: PMC11185697 DOI: 10.1101/2024.06.06.597370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Resident memory T cells (T RM ) have been described in barrier tissues as having a 'sensing and alarm' function where, upon sensing cognate antigen, they alarm the surrounding tissue and orchestrate local recruitment and activation of immune cells. In the immunologically unique and tightly restricted CNS, it remains unclear if and how brain T RM , which express the inhibitory receptor PD-1, alarm the surrounding tissue during antigen re-encounter. Here, we reveal that T RM are sufficient to drive the rapid remodeling of the brain immune landscape through activation of microglia, DCs, NK cells, and B cells, expansion of Tregs, and recruitment of macrophages and monocytic dendritic cells. Moreover, we report that while PD-1 restrains granzyme B expression by reactivated brain T RM , it has no effect on cytotoxicity or downstream alarm responses. We conclude that T RM are sufficient to trigger rapid immune activation and recruitment in the CNS and may have an unappreciated role in driving neuroinflammation.
Collapse
|
10
|
Yin X, Zhang S, Lee JH, Dong H, Mourgkos G, Terwilliger G, Kraus A, Geraldo LH, Poulet M, Fischer S, Zhou T, Mohammed FS, Zhou J, Wang Y, Malloy S, Rohner N, Sharma L, Salinas I, Eichmann A, Thomas JL, Saltzman WM, Huttner A, Zeiss C, Ring A, Iwasaki A, Song E. Compartmentalized ocular lymphatic system mediates eye-brain immunity. Nature 2024; 628:204-211. [PMID: 38418880 PMCID: PMC10990932 DOI: 10.1038/s41586-024-07130-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/29/2024] [Indexed: 03/02/2024]
Abstract
The eye, an anatomical extension of the central nervous system (CNS), exhibits many molecular and cellular parallels to the brain. Emerging research demonstrates that changes in the brain are often reflected in the eye, particularly in the retina1. Still, the possibility of an immunological nexus between the posterior eye and the rest of the CNS tissues remains unexplored. Here, studying immune responses to herpes simplex virus in the brain, we observed that intravitreal immunization protects mice against intracranial viral challenge. This protection extended to bacteria and even tumours, allowing therapeutic immune responses against glioblastoma through intravitreal immunization. We further show that the anterior and posterior compartments of the eye have distinct lymphatic drainage systems, with the latter draining to the deep cervical lymph nodes through lymphatic vasculature in the optic nerve sheath. This posterior lymphatic drainage, like that of meningeal lymphatics, could be modulated by the lymphatic stimulator VEGFC. Conversely, we show that inhibition of lymphatic signalling on the optic nerve could overcome a major limitation in gene therapy by diminishing the immune response to adeno-associated virus and ensuring continued efficacy after multiple doses. These results reveal a shared lymphatic circuit able to mount a unified immune response between the posterior eye and the brain, highlighting an understudied immunological feature of the eye and opening up the potential for new therapeutic strategies in ocular and CNS diseases.
Collapse
Affiliation(s)
- Xiangyun Yin
- Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Sophia Zhang
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Ju Hyun Lee
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, CT, USA
| | - Huiping Dong
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - George Mourgkos
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Gordon Terwilliger
- Section of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Aurora Kraus
- Center of Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM, USA
| | - Luiz Henrique Geraldo
- Department of Internal Medicine, Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Mathilde Poulet
- Department of Internal Medicine, Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Suzanne Fischer
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Ting Zhou
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
| | - Farrah Shalima Mohammed
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Jiangbing Zhou
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Yongfu Wang
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Seth Malloy
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Lokesh Sharma
- Section of Pulmonary and Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Irene Salinas
- Center of Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM, USA
| | - Anne Eichmann
- Department of Internal Medicine, Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
- Université de Paris, INSERM, PARCC, Paris, France
| | - Jean-Leon Thomas
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
- Department of Chemical & Environmental Engineering, Yale School of Engineering and Applied Science, New Haven, CT, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Anita Huttner
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Caroline Zeiss
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Aaron Ring
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Eric Song
- Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT, USA.
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
11
|
Foo IJH, Chua BY, Clemens EB, Chang SY, Jia X, McQuilten HA, Yap AHY, Cabug AF, Ashayeripanah M, McWilliam HEG, Villadangos JA, Evrard M, Mackay LK, Wakim LM, Fazakerley JK, Kedzierska K, Kedzierski L. Prior infection with unrelated neurotropic virus exacerbates influenza disease and impairs lung T cell responses. Nat Commun 2024; 15:2619. [PMID: 38521764 PMCID: PMC10960853 DOI: 10.1038/s41467-024-46822-7] [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: 07/28/2023] [Accepted: 03/07/2024] [Indexed: 03/25/2024] Open
Abstract
Immunity to infectious diseases is predominantly studied by measuring immune responses towards a single pathogen, although co-infections are common. In-depth mechanisms on how co-infections impact anti-viral immunity are lacking, but are highly relevant to treatment and prevention. We established a mouse model of co-infection with unrelated viruses, influenza A (IAV) and Semliki Forest virus (SFV), causing disease in different organ systems. SFV infection eight days before IAV infection results in prolonged IAV replication, elevated cytokine/chemokine levels and exacerbated lung pathology. This is associated with impaired lung IAV-specific CD8+ T cell responses, stemming from suboptimal CD8+ T cell activation and proliferation in draining lymph nodes, and dendritic cell paralysis. Prior SFV infection leads to increased blood brain barrier permeability and presence of IAV RNA in brain, associated with increased trafficking of IAV-specific CD8+ T cells and establishment of long-term tissue-resident memory. Relative to lung IAV-specific CD8+ T cells, brain memory IAV-specific CD8+ T cells have increased TCR repertoire diversity within immunodominant DbNP366+CD8+ and DbPA224+CD8+ responses, featuring suboptimal TCR clonotypes. Overall, our study demonstrates that infection with an unrelated neurotropic virus perturbs IAV-specific immune responses and exacerbates IAV disease. Our work provides key insights into therapy and vaccine regimens directed against unrelated pathogens.
Collapse
Affiliation(s)
- Isabelle Jia-Hui Foo
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Veterinary Biosciences, Faculty of Science, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Brendon Y Chua
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - E Bridie Clemens
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - So Young Chang
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hayley A McQuilten
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Ashley Huey Yiing Yap
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Aira F Cabug
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Mitra Ashayeripanah
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hamish E G McWilliam
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jose A Villadangos
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Biochemistry and Pharmacology; Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Maximilien Evrard
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Laura K Mackay
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - John K Fazakerley
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Veterinary Biosciences, Faculty of Science, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| |
Collapse
|
12
|
Vecchiarelli HA, Tremblay MÈ. Disrupting T cell memory to promote stress resilience: A role for CD74? Brain Behav Immun 2023; 114:240-241. [PMID: 37574175 DOI: 10.1016/j.bbi.2023.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023] Open
Affiliation(s)
- Haley A Vecchiarelli
- Division of Medical Sciences, Centre for Advanced Materials and Related Technology (CAMTEC), and Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
| | - Marie-Ève Tremblay
- Division of Medical Sciences, Centre for Advanced Materials and Related Technology (CAMTEC), and Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada; Département de médecine moléculaire, Axe neurosciences, Centre de recherche du CHU de Québec, Université Laval, Québec City, QC, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|