1
|
Li J, Yang C, Zhang Y, Hong X, Jiang M, Zhu Z, Li J. Deciphering aging-associated prognosis and heterogeneity in gastric cancer through a machine learning-driven approach. iScience 2025; 28:112316. [PMID: 40256325 PMCID: PMC12008712 DOI: 10.1016/j.isci.2025.112316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 04/22/2025] Open
Abstract
Gastric cancer (GC) is a prevalent malignancy with a high mortality rate and limited treatment options. Aging significantly contributes to tumor progression, and GC was confirmed as an aging-related heterogeneous disease. This study established an aging-associated index (AAI) using a machine learning-derived gene panel to stratify GC patients. High AAI scores associated with poor prognosis and indicated potential benefits from adjuvant chemotherapy, while showing resistance to immunotherapy. Single-cell transcriptome analysis revealed that AAI was enriched in monocyte cells within the tumor microenvironment. Two distinct molecular subtypes of GC were identified through unsupervised clustering, leading to the development of a subtype-specific regulatory network highlighting SOX7 and ELK3 as potential therapeutic targets. Drug sensitivity analyses indicated that patients with high ELK3 expression may respond to FDA-approved drugs (axitinib, dacarbazine, crizotinib, and vincristine). Finally, a user-friendly Shiny application was created to facilitate access to the prognostic model and molecular subtype classifier for GC.
Collapse
Affiliation(s)
- Jiang Li
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Chuanlai Yang
- Department of Science and Technology, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Yunxiao Zhang
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Department of Andrology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiaoning Hong
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Mingye Jiang
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhongxu Zhu
- Biomics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
| | - Jiang Li
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Guangdong, Shenzhen, China
| |
Collapse
|
2
|
Pioli KT, Ghosh S, Boulet A, Leary SC, Pioli PD. Lymphopoiesis is attenuated upon hepatocyte-specific deletion of the cytochrome c oxidase assembly factor Sco1. iScience 2025; 28:112151. [PMID: 40177634 PMCID: PMC11964678 DOI: 10.1016/j.isci.2025.112151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/30/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Mutations that negatively impact mitochondrial function are highly prevalent in humans and lead to disorders with a wide spectrum of disease phenotypes, including deficiencies in immune cell development and/or function. Previous analyses of mice with a hepatocyte-specific cytochrome c oxidase (COX) deficiency revealed an unexpected peripheral blood leukopenia associated with splenic and thymic atrophy. Here, we use mice with a hepatocyte-specific deletion of the COX assembly factor Sco1 to show that metabolic defects extrinsic to the hematopoietic compartment lead to a pan-lymphopenia represented by severe losses in both B and T cells. We further demonstrate that immune defects in these mice are associated with the loss of bone marrow lymphoid progenitors common to both lineages and early signs of autoantibody-mediated autoimmunity. Our findings collectively identify hepatocyte dysfunction as a potential instigator of immunodeficiency in patients with congenital mitochondrial defects who suffer from chronic or recurrent infections.
Collapse
Affiliation(s)
- KimAnh T. Pioli
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada
| | - Sampurna Ghosh
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada
| | - Aren Boulet
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada
| | - Scot C. Leary
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada
| | - Peter D. Pioli
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada
| |
Collapse
|
3
|
Komorowska JA, Grammer C, Bălan M, Swann JB. Ndrg3 is a critical regulator of peripheral T cell maturation and homeostasis. SCIENCE ADVANCES 2025; 11:eads5143. [PMID: 40073135 PMCID: PMC11900881 DOI: 10.1126/sciadv.ads5143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025]
Abstract
To provide protection, anticipatory T cell-dependent immunity is reliant on the generation and maintenance of a naïve T cell repertoire, which is sufficiently diverse to ensure recognition of newly encountered antigens. Therefore, under steady-state conditions, a given individual needs to maintain a large pool of naïve T cells, ready to respond to potential threats. Here, we demonstrate that N-myc downstream-regulated gene 3 (Ndrg3) is essential for naïve T cell stability. Mice with T cell-specific Ndrg3 loss are lymphopenic, with reduced numbers of conventional T cells and natural killer T cells. We show that in the absence of Ndrg3, naïve CD8+ T cells exhibit high rates of both proliferation and apoptosis, phenotypes that could be partially rescued by transgenic expression of a high-avidity T cell receptor. Furthermore, Ndrg3-deficient cells were refractory to interleukin-4, resulting in reduced Eomes induction, and a decreased virtual memory population. Our study therefore identifies Ndrg3 as an unexpected, pleiotropic regulator of T cell homeostasis.
Collapse
Affiliation(s)
- Julia A. Komorowska
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Faculty of Biology, Albert Ludwig University, Freiburg, Germany
| | - Christiane Grammer
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Mirela Bălan
- Bioinformatics Core Facility, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Jeremy B. Swann
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| |
Collapse
|
4
|
Alvarez-Sierra D, Martínez-Gallo M, Sánchez-Montalvá A, Fernández-Sanmartín M, Colobran R, Espinosa-Pereiro J, Poyatos-Canton E, Zurera-Egea C, Sánchez-Pla A, Violan C, Parra R, Alzayat H, Vivancos A, Morandeira-Rego F, Urban-Vargas B, Martínez-Cáceres E, Hernández-González M, Bas-Minguet J, Katsikis PD, Teniente-Serra A, Pujol-Borrell R. The immune response to SARS-CoV-2 in COVID-19 as a recall response susceptible to immune imprinting: A prospective cohort study. Clin Immunol 2025; 272:110429. [PMID: 39842683 DOI: 10.1016/j.clim.2025.110429] [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: 11/20/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
The antibody response to SARS-CoV-2 does not follow the immunoglobulin isotype pattern of primary responses, conflicting with the current interpretation of COVID-19. METHODS Prospective cohort study of 191 SARS-CoV-2 infection cases and 44 controls from the second wave of COVID-19. The study stratified patients by severity and analyzed the trajectories of SARS-CoV-2 antibodies and multiple immune variables. RESULTS Isotype-specific antibody time course profiles to SARS-CoV-2 revealed a pattern of recall response in 94.2 % of cases. The time course profiles of plasmablasts, B cells, cTfh high-resolution subsets, and cytokines indicated a secondary response. The transcriptomic data showed that this cohort is strictly comparable to contemporary cohorts. CONCLUSIONS In most cases, the immune response to SARS-CoV-2 is a recall response. This constitutes a favorable scenario for most COVID-19 cases to be subjected to immune imprinting by endemic coronavirus, which, in turn, can influence the immune response to SARS-CoV-2.
Collapse
Affiliation(s)
- Daniel Alvarez-Sierra
- Translational Immunology Research Group, Vall d'Hebron Research Institute (VHIR), Campus Vall d'Hebron, Barcelona, Spain.
| | - Mónica Martínez-Gallo
- Translational Immunology Research Group, Vall d'Hebron Research Institute (VHIR), Campus Vall d'Hebron, Barcelona, Spain; Immunology Department, Hospital Universitari Vall d'Hebron, Campus Vall d'Hebron, Barcelona, Spain; Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma Barcelona, Campus Vall d'Hebron and Campus Bellaterra, Barcelona, Spain
| | - Adrián Sánchez-Montalvá
- Infectious Disease Department, Hospital Universitari Vall d'Hebron, Campus Vall d'Hebró, Barcelona, Spain; International Health Program, Institut Català de la Salut, Vall d'Hebron Research Institute (VHIR), Campus Vall d'Hebron, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Campus Vall d'Hebron, Barcelona, Spain
| | - Marco Fernández-Sanmartín
- Flow Cytometry Laboratory, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Roger Colobran
- Translational Immunology Research Group, Vall d'Hebron Research Institute (VHIR), Campus Vall d'Hebron, Barcelona, Spain; Immunology Department, Hospital Universitari Vall d'Hebron, Campus Vall d'Hebron, Barcelona, Spain; Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma Barcelona, Campus Vall d'Hebron and Campus Bellaterra, Barcelona, Spain
| | - Juan Espinosa-Pereiro
- Infectious Disease Department, Hospital Universitari Vall d'Hebron, Campus Vall d'Hebró, Barcelona, Spain; International Health Program, Institut Català de la Salut, Vall d'Hebron Research Institute (VHIR), Campus Vall d'Hebron, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Campus Vall d'Hebron, Barcelona, Spain
| | - Elísabet Poyatos-Canton
- Immunology Division, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain
| | - Coral Zurera-Egea
- Immunology and Inflammation Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Alex Sánchez-Pla
- Bioinformatics and Statistics Group, University of Barcelona, Barcelona, Spain
| | - Concepción Violan
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Mataró, Barcelona, Spain
| | - Rafael Parra
- Banc de Sang i Teixits, Hospital Universitari Vall Hebron, Barcelona, Spain
| | - Hammad Alzayat
- Flow Cytometry Laboratory, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Ana Vivancos
- Tumor Immunology and Immunotherapy Group, Vall Hebron Institut Oncology (VHIO), Campus Vall d'Hebron, Barcelona, Spain
| | | | - Blanca Urban-Vargas
- Immunology Division, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain
| | - Eva Martínez-Cáceres
- Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma Barcelona, Campus Vall d'Hebron and Campus Bellaterra, Barcelona, Spain; Immunology Department, Hospital Universitari Germans Trias I Pujol, Barcelona, Spain; Immunology and Inflammation Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Manuel Hernández-González
- Translational Immunology Research Group, Vall d'Hebron Research Institute (VHIR), Campus Vall d'Hebron, Barcelona, Spain; Immunology Department, Hospital Universitari Vall d'Hebron, Campus Vall d'Hebron, Barcelona, Spain; Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma Barcelona, Campus Vall d'Hebron and Campus Bellaterra, Barcelona, Spain
| | - Jordi Bas-Minguet
- Immunology Division, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain
| | - Peter D Katsikis
- Dept. of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Aina Teniente-Serra
- Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma Barcelona, Campus Vall d'Hebron and Campus Bellaterra, Barcelona, Spain; Immunology Department, Hospital Universitari Germans Trias I Pujol, Barcelona, Spain; Immunology and Inflammation Research Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Ricardo Pujol-Borrell
- Immunology Department, Hospital Universitari Vall d'Hebron, Campus Vall d'Hebron, Barcelona, Spain; Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma Barcelona, Campus Vall d'Hebron and Campus Bellaterra, Barcelona, Spain; Tumor Immunology and Immunotherapy Group, Vall Hebron Institut Oncology (VHIO), Campus Vall d'Hebron, Barcelona, Spain.
| |
Collapse
|
5
|
Cao JF, Yang GJ, Zhang YA, Chen J. Contribution of interleukins in the regulation of teleost fish immunity: A review from the perspective of regulating macrophages. FISH & SHELLFISH IMMUNOLOGY 2025; 158:110173. [PMID: 39909123 DOI: 10.1016/j.fsi.2025.110173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Interleukins (ILs) are potent secreted regulators of a wide range of cell types and cellular activities, particularly in the immune system. They are able to participate in intercellular communication in homeostasis and disease, thereby exerting immune functions. Macrophages serve as the innate immune cells of vertebrates and play a pivotal role in defending against and eliminating external pathogens. In mammals, the immune response mounted by macrophages is intricately linked to ILs. Given the fact that teleost fish have evolved an innate immune system that closely resembles those of mammals, particularly in terms of the functionality of macrophages, raises the intriguing possibility that the regulatory function of ILs in macrophage-mediated immunity might be evolutionarily conserved across both mammal and teleost fish lineages. Consequently, from the perspective of interleukin regulation of macrophages, this review outlines the relationship between ILs and macrophages in teleost fish, and elucidates the regulatory role of ILs of immune cell function in teleost fish, thereby contributing to our understanding of the key role of these cytokines in the prevention and control of aquaculture diseases.
Collapse
Affiliation(s)
- Jia-Feng Cao
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315832, China
| | - Guan-Jun Yang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315832, China
| | - Yong-An Zhang
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Jiong Chen
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315832, China.
| |
Collapse
|
6
|
Bachoo S, Gudgeon N, Mann R, Stavrou V, Bishop EL, Kelly A, Uribe AH, Loeliger J, Frick C, Maddocks ODK, Lavender P, Hess C, Dimeloe S. IL-7 promotes integrated glucose and amino acid sensing during homeostatic CD4 + T cell proliferation. Cell Rep 2025; 44:115199. [PMID: 39799568 DOI: 10.1016/j.celrep.2024.115199] [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: 07/29/2024] [Revised: 11/15/2024] [Accepted: 12/20/2024] [Indexed: 01/15/2025] Open
Abstract
Interleukin (IL)-7 promotes T cell expansion during lymphopenia. We studied the metabolic basis in CD4+ T cells, observing increased glucose usage for nucleotide synthesis and oxidation in the tricarboxylic acid (TCA) cycle. Unlike other TCA metabolites, glucose-derived citrate does not accumulate upon IL-7 exposure, indicating diversion into other processes. In agreement, IL-7 promotes glucose-dependent histone acetylation and chromatin accessibility, notable at the loci of the amino acid-sensing Ragulator complex. Consistently, the expression of its subunit late endosomal/lysosomal adaptor, MAPK and mTOR activator 5 (LAMTOR5) is promoted by IL-7 in a glucose-dependent manner, and glucose availability determines amino acid-dependent mechanistic target of rapamycin (mTOR) activation, confirming integrated nutrient sensing. LAMTOR5 deletion impairs IL-7-mediated T cell expansion, establishing that glycolysis in the absence of Ragulator activation is insufficient to support this. Clinically, CD4+ T cells from stem cell transplant recipients demonstrate coordinated upregulation of glycolytic and TCA cycle enzymes, amino acid-sensing machinery, and mTOR targets, highlighting the potential to therapeutically target this pathway to fine-tune lymphopenia-induced T cell proliferation.
Collapse
Affiliation(s)
- Seema Bachoo
- School of Infection, Inflammation and Immunology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Nancy Gudgeon
- School of Infection, Inflammation and Immunology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Rebecca Mann
- School of Infection, Inflammation and Immunology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Victoria Stavrou
- School of Infection, Inflammation and Immunology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Emma L Bishop
- School of Infection, Inflammation and Immunology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Audrey Kelly
- School of Immunology & Microbial Sciences, MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - Alejandro Huerta Uribe
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Jordan Loeliger
- Immunobiology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Corina Frick
- Immunobiology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Oliver D K Maddocks
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Paul Lavender
- School of Immunology & Microbial Sciences, MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - Christoph Hess
- Immunobiology, Department of Biomedicine, University of Basel, Basel, Switzerland; Department of Medicine, CITIID, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Sarah Dimeloe
- School of Infection, Inflammation and Immunology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
7
|
Zhang Z, Zhao L, Huang T, Chen Z, Zhao Y, Liang J, Ao X, Jia X, Kang L, Kong L, Jing Q, Hu J, Gu L, Pan F, Hu Z, He L, Zhou M, Chen J, Guo Z. A self-activated and protective module enhances the preclinical performance of allogeneic anti-CD70 CAR-T cells. Front Immunol 2025; 15:1531294. [PMID: 39906740 PMCID: PMC11792090 DOI: 10.3389/fimmu.2024.1531294] [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: 11/20/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
Introduction Allogeneic chimeric antigen receptor T (CAR-T) therapy, also known as universal CAR-T (UCAR-T) therapy, offers broad applicability, high production efficiency, and reduced costs, enabling quicker access for patients. However, clinical application remains limited by challenges such as immune rejection, and issues with potency and durability. Methods We first screened a safe and effective anti-CD70 scFv to construct anti-CD70 CAR-T cells. Anti-CD70 UCAR-T cells were then generated by knocking out TRAC, B2M, and HLA-DRA. To address the limitations of UCAR-T therapy, we developed an 'all-in-one' self-activated and protective (SAP) module, integrated into the CAR scaffold. The SAP module consists of the CD47 extracellular domain, a mutant interleukin 7 receptor alpha (IL7Rα) transmembrane domain, and the IL7Rα intracellular domain, designed to protect UCAR-T cells from host immune attacks and enhance their survival. Results SAP UCAR-T cells demonstrated significantly reduced immune rejection from the innate immune system, as evidenced by enhanced survival and functionality both in vitro and in vivo. The modified UCAR-T cells exhibited improved persistence, with no observed safety concerns. Furthermore, SAP UCAR-T cells maintained process stability during scale-up production, indicating the potential for large-scale manufacturing. Discussion Our findings highlight the SAP module as a promising strategy for the preclinical development of anti-CD70 UCAR-T, paving the way for an 'off-the-shelf' cell therapy product.
Collapse
Affiliation(s)
- Zhao Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lianfeng Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tinghui Huang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhengliang Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
- Department of Research and Development, Nanjing Calmhome Cell & Gene Engineering Institute Co., Ltd., Nanjing, China
| | - Yaoyao Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
- Department of Research and Development, Nanjing Calmhome Cell & Gene Engineering Institute Co., Ltd., Nanjing, China
| | - Junqing Liang
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Xudong Ao
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Xiaoqiong Jia
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Lei Kang
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Linghui Kong
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Qi Jing
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Jianhua Hu
- Center of Biotherapy, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Lili Gu
- Department of Research and Development, Nanjing Calmhome Cell & Gene Engineering Institute Co., Ltd., Nanjing, China
| | - Feiyan Pan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lingfeng He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Muya Zhou
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jiannan Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
8
|
Ding X, Fan L, Xu L, Ma X, Meng P, Li J, Li J, Yue J. Incomplete Immune Reconstitution and Traditional Chinese Medicine in Patients with HIV/AIDS: Challenges and Perspectives. Infect Drug Resist 2024; 17:5827-5838. [PMID: 39737090 PMCID: PMC11683152 DOI: 10.2147/idr.s497083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/12/2024] [Indexed: 01/01/2025] Open
Abstract
Antiretroviral therapy can reduce human immunodeficiency virus (HIV) load to undetectable levels and restore CD4+ T cells to rebuild immune function in patients with HIV. However, some patients fail to achieve immune reconstitution despite treatment. Traditional Chinese medicine is an important branch of complementary and alternative medicine for the treatment of HIV infection, and a growing number of studies has demonstrated that traditional Chinese medicine can increase CD4+ T cell counts in patients, thereby promoting immune reconstitution, ameliorating symptoms and signs, and improving quality of life. Here, we review pathogenesis in immunological non-responders and research into their treatment with traditional Chinese medicine. Furthermore, we summarize potential future research directions, including elucidation of how traditional Chinese medicine can regulate CD4+ T cells to reduce opportunistic infections and improve quality of life in immunological non-responders.
Collapse
Affiliation(s)
- Xue Ding
- Department of Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Leilei Fan
- Department of Cardiovascular, The First People’s Hospital of Zhengzhou, Zhengzhou, People’s Republic of China
| | - Liran Xu
- Department of the First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Xiuxia Ma
- Department of AIDS Clinical Research Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Pengfei Meng
- Department of AIDS Clinical Research Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Jie Li
- Department of AIDS Clinical Research Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Jiahe Li
- Department of the First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Jingyu Yue
- Department of AIDS Clinical Research Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| |
Collapse
|
9
|
Miyamoto H, Kondo Y, Itobayashi E, Uehara M, Hiraoka A, Kudo M, Kakizaki S, Kagawa T, Miuma S, Suzuki T, Sugi K, Suyama K, Beppu T, Toyoda H, Yoshiji H, Uojima H, Miyase S, Inoue K, Tamori A, Ito T, Shimose S, Suda G, Hayashi T, Onishi M, Narahara S, Watanabe T, Iwatsuki M, Fukushima S, Tanaka Y. Evaluation of the associations of interlukin-7 genetic variants with toxicity and efficacy of immune checkpoint inhibitors: A replication study of a Japanese population, based on the findings of a European genome-wide association study. Hepatol Res 2024; 54:1215-1225. [PMID: 38990762 DOI: 10.1111/hepr.14092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
AIM Recent genome-wide association studies of European populations have identified rs16906115, a single-nucleotide polymorphism in the interleukin-7 gene, as a predictor of immune-related adverse events (irAEs) and the therapeutic efficacy of immune checkpoint inhibitors. We evaluated this single-nucleotide polymorphism in a Japanese population. METHODS From January 2021, we stored host DNA from individuals who received various types of immune checkpoint inhibitors. From this population, we categorized 510 participants into cases (grade ≥2 irAEs) and controls (received ≥3 immune checkpoint inhibitor doses, follow-up ≥12 weeks, no irAEs), and divided 339 hepatocellular carcinoma patients treated with atezolizumab/bevacizumab into responders and non-responders, evaluated using the modified response evaluation criteria in solid tumors. We compared the minor allele frequencies of rs16906115 between cases and controls, and responders and non-responders. RESULTS In the irAE prediction analysis of 234 cases and 276 controls, the minor allele frequency was 0.244 in the case group and 0.265 in the control group. This difference is not significant. In the analysis predicting the therapeutic efficacy for hepatocellular carcinoma patients, the responders had a significantly lower minor allele frequency of 0.220, compared with 0.300 for the non-responders (p = 0.022). Univariate and multivariate analyses identified the minor allele homozygosity as a significant predictor of treatment response, with odds ratios of 0.292 (p = 0.015) in the univariate analysis and 0.315 (p = 0.023) in the multivariate analysis. CONCLUSIONS In our Japanese cohort, no association was found between the rs16906115 minor allele and irAEs or treatment efficacy. The minor allele homozygosity may be associated with a negative therapeutic outcome. CLINICAL TRIAL REGISTRATION UMIN Clinical Trials Registry with the number UMIN000043798.
Collapse
Affiliation(s)
- Hideaki Miyamoto
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
| | - Yasuteru Kondo
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
- Department of Hepatology, Sendai Tokushukai Hospital, Miyagi, Japan
- Department of Hepatology, Sendai Kousei Hospital, Miyagi, Japan
| | - Ei Itobayashi
- Department of Gastroenterology, Asahi General Hospital, Chiba, Japan
| | - Masayoshi Uehara
- Department of Gastroenterology, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Atsushi Hiraoka
- Department of Gastroenterology, Ehime Prefectural Central Hospital, Ehime, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Hospital, Osaka, Japan
| | - Satoru Kakizaki
- Department of Clinical Research, NHO Takasaki General Medical Center, Gunma, Japan
| | - Tatehiro Kagawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Satoshi Miuma
- Department of Gastroenterology and Hepatology, Nagasaki University Hospital, Nagasaki, Japan
| | - Takanori Suzuki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Science, Aichi, Japan
| | - Kazuhiro Sugi
- Department of Gastroenterology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Koichi Suyama
- Department of Medical Oncology, Toranomon Hospital, Tokyo, Japan
| | - Toru Beppu
- Department of Surgery, Yamaga City Medical Center, Kumamoto, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Gifu, Japan
| | - Hitoshi Yoshiji
- Department of Gastroenterology, Nara Medical University, Nara, Japan
| | - Haruki Uojima
- Department of Gastroenterology, Kitasato University Hospital, Kanagawa, Japan
| | - Shiho Miyase
- Department of Gastroenterology and Hepatology, Kumamoto Shinto General Hospital, Kumamoto, Japan
| | - Kaori Inoue
- Department of Liver and Diabetes and Endocrinology, Saga University Hospital, Saga, Japan
| | - Akihiro Tamori
- Department of Hepatology, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Hospital, Aichi, Japan
| | - Shigeo Shimose
- Department of Gastroenterology, Kurume University Hospital, Fukuoka, Japan
| | - Goki Suda
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Hokkaido, Japan
| | - Tsuguru Hayashi
- Department of Hepatology, Sendai Kousei Hospital, Miyagi, Japan
| | - Masaya Onishi
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
- Department of Gastroenterology, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Satoshi Narahara
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
| | - Takehisa Watanabe
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Fukushima
- Faculty of Life Sciences, Department of Dermatology and Plastic Surgery, Kumamoto University, Kumamoto, Japan
| | - Yasuhito Tanaka
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
| |
Collapse
|
10
|
Li Y, Xiao J, Li C, Yang M. Memory inflation: Beyond the acute phase of viral infection. Cell Prolif 2024; 57:e13705. [PMID: 38992867 PMCID: PMC11628752 DOI: 10.1111/cpr.13705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024] Open
Abstract
Memory inflation is confirmed as the most commonly dysregulation of host immunity with antigen-independent manner in mammals after viral infection. By generating large numbers of effector/memory and terminal differentiated effector memory CD8+ T cells with diminished naïve subsets, memory inflation is believed to play critical roles in connecting the viral infection and the onset of multiple diseases. Here, we reviewed the current understanding of memory inflated CD8+ T cells in their distinct phenotypic features that different from exhausted subsets; the intrinsic and extrinsic roles in regulating the formation of memory inflation; and the key proteins in maintaining the expansion and proliferation of inflationary populations. More importantly, based on the evidences from both clinic and animal models, we summarized the potential mechanisms of memory inflation to trigger autoimmune neuropathies, such as Guillain-Barré syndrome and multiple sclerosis; the correlations of memory inflation between tumorigenesis and resistance of tumour immunotherapies; as well as the effects of memory inflation to facilitate vascular disease progression. To sum up, better understanding of memory inflation could provide us an opportunity to beyond the acute phase of viral infection, and shed a light on the long-term influences of CD8+ T cell heterogeneity in dampen host immune homeostasis.
Collapse
Affiliation(s)
- Yanfei Li
- School of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengduChina
| | - Jie Xiao
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Chen Li
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Mu Yang
- School of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengduChina
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
11
|
Azizi G, Van den Broek B, Ishikawa LLW, Naziri H, Yazdani R, Zhang GX, Ciric B, Rostami A. IL-7Rα on CD4 + T cells is required for their survival and the pathogenesis of experimental autoimmune encephalomyelitis. J Neuroinflammation 2024; 21:253. [PMID: 39380064 PMCID: PMC11460225 DOI: 10.1186/s12974-024-03224-2] [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: 05/17/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND The IL-7 receptor alpha (IL-7Rα) binds both IL-7 and thymic stromal lymphopoietin (TSLP). IL-7Rα is essential for the development and survival of naive CD4+ T cells and their differentiation to effector/memory CD4+ T cells. Mice lacking IL-7Rα have severe lymphopenia and are resistant to experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis. However, it has been reported that IL-7Rα on peripheral CD4+ T cells is disposable for their maintenance and EAE pathogenesis, which does not align with the body of knowledge on the role of IL-7Rα in the biology of CD4+ T cells. Given that a definitive study on this important topic is lacking, we revisited it using a novel approach, an inducible knockout of the IL-7Rα gene in CD4+ T cells. METHODS We generated Il7rafl/fl/CD4CreERT2 double transgenic mouse line (henceforth CD4ΔIl7ra), susceptible to tamoxifen-induced knockout of the IL-7Rα gene in CD4+ T cells. CD4ΔIl7ra mice were immunized with MOG35 - 55 for EAE induction and monitored for disease development. The expression of IL-7Rα, CD4+ T cell numbers, and MOG35 - 55-specific CD4+ T cell response was evaluated in the central nervous system (CNS) and lymphoid tissues by flow cytometry. Additionally, splenocytes of CD4ΔIl7ra mice were stimulated with MOG35 - 55 to assess their proliferative response and cytokine production by T helper cells. RESULTS Loss of IL-7Rα from the surface of CD4+ T cells in CD4ΔIl7ra mice was virtually complete several days after tamoxifen treatment. The loss of IL-7Rα in CD4+ T cells led to a gradual and substantial decrease in their numbers in both non-immunized and immunized CD4ΔIl7ra mice, followed by slow repopulation up to the initial numbers. CD4ΔIl7ra mice did not develop EAE. We found a decrease in the total numbers of TNF-, IFN-γ-, IL-17 A-, and GM-CSF-producing CD4+ T cells and regulatory T cells in the spleens and CNS of immunized CD4ΔIl7ra mice. Tracking MOG35 - 55-specific CD4+ T cells revealed a significant reduction in their numbers in CD4ΔIl7ra mice and decreased proliferation and cytokine production in response to MOG35 - 55. CONCLUSION Our study demonstrates that IL-7Rα on peripheral CD4+ T cells is essential for their maintenance, immune response, and EAE pathogenesis.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/genetics
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Mice
- Receptors, Interleukin-7/metabolism
- Receptors, Interleukin-7/genetics
- Mice, Transgenic
- Mice, Inbred C57BL
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Myelin-Oligodendrocyte Glycoprotein/immunology
- Cell Survival/physiology
- Cell Survival/drug effects
- Peptide Fragments/toxicity
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Mice, Knockout
- Cytokines/metabolism
Collapse
Affiliation(s)
- Gholamreza Azizi
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Bram Van den Broek
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | | | - Hamed Naziri
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Reza Yazdani
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, 900 Walnut Street, Suite 300, Philadelphia, PA, 19107, USA.
| |
Collapse
|
12
|
Jabłońska A, Jabłonowska E, Studzińska M, Kamerys J, Paradowska E. Polymorphisms in the genes encoding RLR and TLR3 and CMV DNAemia in subjects coinfected with human immunodeficiency virus and cytomegalovirus. Arch Virol 2024; 169:211. [PMID: 39331212 PMCID: PMC11436440 DOI: 10.1007/s00705-024-06114-3] [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: 03/04/2024] [Accepted: 07/08/2024] [Indexed: 09/28/2024]
Abstract
Cytomegalovirus (CMV) is a pathogen that is common worldwide and is often present in individuals infected with human immunodeficiency virus (HIV). Pattern recognition receptors (PRRs) are host sensors that activate the immune response against infectious agents. However, it is unclear whether PRR single-nucleotide polymorphisms (SNPs) are associated with the occurrence of CMV DNAemia in subjects coinfected with HIV and CMV. HIV/CMV-coinfected patients with and without CMV DNAemia were recruited for this study. The DDX58 rs10813831 and IFIH1 (rs3747517 and rs1990760) polymorphisms were genotyped using the TaqMan Allelic Discrimination Assay, whereas the DDX58 rs12006123 and TLR3 (rs3775291 and rs3775296) SNPs were analyzed using a polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) assay. A mutation present in at least one allele of the DDX58 rs12006123 SNP occurred at least two times more frequently in HIV/CMV-coinfected patients with CMV DNAemia than in coinfected subjects without CMV DNAemia (OR, 2.50; 95% CI, 1.33-4.68; p = 0.004, in the dominant model). A higher level of CMV DNAemia was observed in subjects who had the heterozygous (GA) or homozygous recessive (AA) genotype for the DDX58 rs12006123 SNP compared with those who had the wild-type (GG) genotype (p = 0.0003). Moreover, in subjects with a mutation detected in at least one allele of the DDX58 rs12006123 SNP, a lower serum IFN-β concentration was found compared with those who had a wild-type (GG) genotype for this polymorphism (p = 0.024). The DDX58 rs12006123 SNP is associated with CMV DNAemia in HIV/CMV-coinfected patients.
Collapse
Affiliation(s)
- Agnieszka Jabłońska
- Laboratory of Virology, Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa St, Lodz, 93-232, Poland.
| | - Elżbieta Jabłonowska
- Department of Infectious Diseases and Hepatology, Medical University of Lodz, Lodz, Poland
| | - Mirosława Studzińska
- Laboratory of Virology, Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa St, Lodz, 93-232, Poland
| | - Juliusz Kamerys
- Department of Infectious Diseases and Hepatology, Medical University of Lodz, Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa St, Lodz, 93-232, Poland
| |
Collapse
|
13
|
Russell J, Chen L, Liu A, Wang J, Ghosh S, Zhong X, Shi H, Beutler B, Nair-Gill E. Lrp10 suppresses IL7R limiting CD8 T cell homeostatic expansion and anti-tumor immunity. EMBO Rep 2024; 25:3601-3626. [PMID: 38956225 PMCID: PMC11315911 DOI: 10.1038/s44319-024-00191-w] [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: 12/14/2023] [Revised: 06/07/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024] Open
Abstract
Signals emanating from the T-cell receptor (TCR), co-stimulatory receptors, and cytokine receptors each influence CD8 T-cell fate. Understanding how these signals respond to homeostatic and microenvironmental cues can reveal new ways to therapeutically direct T-cell function. Through forward genetic screening in mice, we discover that loss-of-function mutations in LDL receptor-related protein 10 (Lrp10) cause naive and central memory CD8 T cells to accumulate in peripheral lymphoid organs. Lrp10 encodes a conserved cell surface protein of unknown immunological function. T-cell activation induces Lrp10 expression, which post-translationally suppresses IL7 receptor (IL7R) levels. Accordingly, Lrp10 deletion enhances T-cell homeostatic expansion through IL7R signaling. Lrp10-deficient mice are also intrinsically resistant to syngeneic tumors. This phenotype depends on dense tumor infiltration of CD8 T cells, which display increased memory cell characteristics, reduced terminal exhaustion, and augmented responses to immune checkpoint inhibition. Here, we present Lrp10 as a new negative regulator of CD8 T-cell homeostasis and a host factor that controls tumor resistance with implications for immunotherapy.
Collapse
Affiliation(s)
- Jamie Russell
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8505, USA
| | - Luming Chen
- Medical Scientist Training Program, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8505, USA
| | - Aijie Liu
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8505, USA
| | - Jianhui Wang
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8505, USA
| | - Subarna Ghosh
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8505, USA
| | - Xue Zhong
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8505, USA
| | - Hexin Shi
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8505, USA
| | - Bruce Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8505, USA
| | - Evan Nair-Gill
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8505, USA.
- Department of Internal Medicine, Division of Rheumatic Diseases, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8505, USA.
| |
Collapse
|
14
|
Li W, Yang Y, Zhuo F, Liu S, Zhang K, Zhang W, Huang C, Yu B. Paxbp1 is indispensable for the maintenance of peripheral CD4 T cell homeostasis. Immunology 2024; 172:641-652. [PMID: 38750609 DOI: 10.1111/imm.13802] [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: 01/08/2024] [Accepted: 05/05/2024] [Indexed: 07/05/2024] Open
Abstract
The size and condition of the peripheral CD4 T cell population determine the capacity of the immune response. Under homeostatic conditions, the size of the peripheral CD4 T cell population is maintained through turnover and survival. However, the underlying mechanisms remain inadequately understood. Here, we observed a significant decrease in the percentage of CD4 T cells in the periphery following the targeted deletion of the Paxbp1 gene in mouse T cells. In the absence of Paxbp1, naïve CD4 T cells displayed reduced surface interleukin-7 receptor levels and a decreased capacity to respond to survival signals mediated by interleukin-7. In addition, naïve CD4 T cells deficient in Paxbp1 demonstrated impaired T cell antigen receptor signalling, compromised cell cycle entry, decreased proliferation, and increased apoptosis following stimulation, all of which contributed to the reduction in the number of peripheral CD4 T cells. Therefore, our study highlights the indispensable role of Paxbp1 in maintaining peripheral CD4 T cell homeostasis.
Collapse
Affiliation(s)
- Wenting Li
- Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Yang Yang
- Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Fan Zhuo
- Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Shenglin Liu
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, College of Biological and Food Engineering, Huaihua University, Huaihua, Hunan Province, China
| | - Kaoyuan Zhang
- Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Wei Zhang
- Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China
| | - Cong Huang
- Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Bo Yu
- Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| |
Collapse
|
15
|
Zhao J, Zhang Y, Wang JY, Wei B, Liu YG. Expression pattern of serum interleukin-7 in elderly septic patients and its prognostic value for predicting short-term mortality. Cytokine 2024; 180:156664. [PMID: 38795605 DOI: 10.1016/j.cyto.2024.156664] [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: 02/19/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND The identification of novel prognostic biomarkers in elderly septic patients are essential for the improvement of mortality in sepsis in the context of precision medicine. The purpose of this study was to explore the expression pattern and prognostic value of serum interleukin-7 (IL-7) in predicting 28-day mortality in elderly patients with sepsis. METHODS Patients were retrospectively enrolled according to the sepsis-3.0 diagnostic criteria and divided into the survival group and non-survival group based on the clinical outcome at the 28-day interval. The baseline characteristic data, samples for the laboratory tests, and the SOFA, Acute Physiology and Chronic Health Evaluation (APACHE II), as well as Glasgow coma scale (GCS) scores, were recorded within 24 h after admission to the emergency department. Serum levels of IL-7 and TNF-α of the patients were quantified by the Luminex assay. Spearman correlation analysis, logistic regressive analysis and receiver operating characteristic curve (ROC) analysis were performed, respectively. RESULTS Totally, 220 elderly patients with sepsis were enrolled, 151 of whom died in a 28-day period. Albumin (ALB), high-density lipoprotein (HDL), systolic pressure (SBP), and platelet (PLT) were found to be significantly higher in the survival group (p < 0.05). IL-7 was shown to be correlated with TNF-α in the non-survival group (p = 0.030) but not in the survival group (p = 0.194). No correlation was shown between IL-7 and other factors (p > 0.05). IL-7 and TNF-α were found to be independent risk factors associated with the 28-day mortality (OR = 1.215, 1.420). Combination of IL-7, SOFA and ALB can make an AUROC of 0.874 with the specificity of 90.77 %. Combination of IL-7 and TNF-α can make an AUROC of 0.901 with the sensitivity of 90.41 % while the combination of IL-7, TNF-α, and ALB can make an AUROC of 0.898 with the sensitivity of 94.52 %. CONCLUSIONS This study highlights the importance of monitoring the serum level of IL-7 and TNF-α in elderly septic patients as well as evaluating the combinations with other routine risk factors which can be potentially used for the identification of elderly septic patients with higher risk of mortality.
Collapse
Affiliation(s)
- Jingjing Zhao
- Department of Infectious Disease and Clinical Microbiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China
| | - Ye Zhang
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Clinical Center for Medicine in Acute Infection, Capital Medical University, Beijing 100043, China
| | - Jun-Yu Wang
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Clinical Center for Medicine in Acute Infection, Capital Medical University, Beijing 100043, China
| | - Bing Wei
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Clinical Center for Medicine in Acute Infection, Capital Medical University, Beijing 100043, China.
| | - Yu-Geng Liu
- Department of Infectious Disease and Clinical Microbiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China; Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Clinical Center for Medicine in Acute Infection, Capital Medical University, Beijing 100043, China.
| |
Collapse
|
16
|
Benito JM, Jiménez-Carretero D, Restrepo C, Ligos JM, Valentín-Quiroga J, Mahillo I, Cabello A, López-Collazo E, Sánchez-Cabo F, Górgolas M, Estrada V, Rallón N. T Cell Homeostasis Disturbances in a Cohort of Long-Term Elite Controllers of HIV Infection. Int J Mol Sci 2024; 25:5937. [PMID: 38892124 PMCID: PMC11172696 DOI: 10.3390/ijms25115937] [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: 04/16/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Elite controllers (ECs) are people living with HIV (PLWH) able to control HIV replication without antiretroviral therapy and have been proposed as a model of a functional HIV cure. Much evidence suggests that this spontaneous control of HIV has a cost in terms of T cell homeostasis alterations. We performed a deep phenotypic study to obtain insight into T cell homeostasis disturbances in ECs maintaining long-term virologic and immunologic control of HIV (long-term elite controllers; LTECs). Forty-seven PLWH were included: 22 LTECs, 15 non-controllers under successful antiretroviral therapy (onART), and 10 non-controllers not receiving ART (offART). Twenty uninfected participants (UCs) were included as a reference. T cell homeostasis was analyzed by spectral flow cytometry and data were analyzed using dimensionality reduction and clustering using R software v3.3.2. Dimensionality reduction and clustering yielded 57 and 54 different CD4 and CD8 T cell clusters, respectively. The offART group showed the highest perturbation of T cell homeostasis, with 18 CD4 clusters and 15 CD8 clusters significantly different from those of UCs. Most of these alterations were reverted in the onART group. Interestingly, LTECs presented several disturbances of T cell homeostasis with 15 CD4 clusters and 13 CD8 clusters different from UC. Moreover, there was a specific profile of T cell homeostasis alterations associated with LTECs, characterized by increases in clusters of naïve T cells, increases in clusters of non-senescent effector CD8 cells, and increases in clusters of central memory CD4 cells. These results demonstrate that, compared to ART-mediated control of HIV, the spontaneous control of HIV is associated with several disturbances in CD4 and CD8 T cell homeostasis. These alterations could be related to the existence of a potent and efficient virus-specific T cell response, and to the ability to halt disease progression by maintaining an adequate pool of CD4 T cells.
Collapse
Affiliation(s)
- José M. Benito
- HIV and Viral Hepatitis Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (N.R.)
- Hospital Universitario Rey Juan Carlos, 28933 Móstoles, Spain
| | - Daniel Jiménez-Carretero
- Unidad de Bioinformática, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (D.J.-C.); (F.S.-C.)
| | - Clara Restrepo
- HIV and Viral Hepatitis Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (N.R.)
- Hospital Universitario Rey Juan Carlos, 28933 Móstoles, Spain
| | | | - Jaime Valentín-Quiroga
- Grupo de Respuesta Inmune Innata, IdiPAZ, Hospital Universitario La Paz, 28046 Madrid, Spain; (J.V.-Q.); (E.L.-C.)
| | - Ignacio Mahillo
- Department of Statistics, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain;
| | - Alfonso Cabello
- Hospital Universitario Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.C.); (M.G.)
| | - Eduardo López-Collazo
- Grupo de Respuesta Inmune Innata, IdiPAZ, Hospital Universitario La Paz, 28046 Madrid, Spain; (J.V.-Q.); (E.L.-C.)
| | - Fátima Sánchez-Cabo
- Unidad de Bioinformática, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (D.J.-C.); (F.S.-C.)
| | - Miguel Górgolas
- Hospital Universitario Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.C.); (M.G.)
| | - Vicente Estrada
- Hospital Universitario Clínico San Carlos, 28040 Madrid, Spain;
| | - Norma Rallón
- HIV and Viral Hepatitis Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (C.R.); (N.R.)
- Hospital Universitario Rey Juan Carlos, 28933 Móstoles, Spain
| |
Collapse
|
17
|
Beumer-Chuwonpad A, Behr FM, van Alphen FPJ, Kragten NAM, Hoogendijk AJ, van den Biggelaar M, van Gisbergen KPJM. Intestinal tissue-resident memory T cells maintain distinct identity from circulating memory T cells after in vitro restimulation. Eur J Immunol 2024; 54:e2350873. [PMID: 38501878 DOI: 10.1002/eji.202350873] [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: 11/01/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/20/2024]
Abstract
Resident memory T (TRM) cells have been recently established as an important subset of memory T cells that provide early and essential protection against reinfection in the absence of circulating memory T cells. Recent findings showing that TRM expand in vivo after repeated antigenic stimulation indicate that these memory T cells are not terminally differentiated. This suggests an opportunity for in vitro TRM expansion to apply in an immunotherapy setting. However, it has also been shown that TRM may not maintain their identity and form circulating memory T cells after in vivo restimulation. Therefore, we set out to determine how TRM respond to antigenic activation in culture. Using Listeria monocytogenes and LCMV infection models, we found that TRM from the intraepithelial compartment of the small intestine expand in vitro after antigenic stimulation and subsequent resting in homeostatic cytokines. A large fraction of the expanded TRM retained their phenotype, including the expression of key TRM markers CD69 and CD103 (ITGAE). The optimal culture of TRM required low O2 pressure to maintain the expression of these and other TRM-associated molecules. Expanded TRM retained their effector capacity to produce cytokines after restimulation, but did not acquire a highly glycolytic profile indicative of effector T cells. The proteomic analysis confirmed TRM profile retention, including expression of TRM-related transcription factors, tissue retention factors, adhesion molecules, and enzymes involved in fatty acid metabolism. Collectively, our data indicate that limiting oxygen conditions supports in vitro expansion of TRM cells that maintain their TRM phenotype, at least in part, suggesting an opportunity for therapeutic strategies that require in vitro expansion of TRM.
Collapse
MESH Headings
- Animals
- Memory T Cells/immunology
- Immunologic Memory/immunology
- Mice
- Listeria monocytogenes/immunology
- Antigens, CD/metabolism
- Antigens, CD/immunology
- Integrin alpha Chains/metabolism
- Mice, Inbred C57BL
- Listeriosis/immunology
- Lectins, C-Type/metabolism
- Lectins, C-Type/immunology
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Cytokines/metabolism
- Cytokines/immunology
- Lymphocyte Activation/immunology
- Lymphocytic choriomeningitis virus/immunology
- Intestinal Mucosa/immunology
- CD8-Positive T-Lymphocytes/immunology
- Intestine, Small/immunology
- Cells, Cultured
Collapse
Affiliation(s)
- Ammarina Beumer-Chuwonpad
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Felix M Behr
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Floris P J van Alphen
- Department of Research Facilities, Sanquin Research and Laboratory Services, Amsterdam, the Netherlands
| | - Natasja A M Kragten
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Arie J Hoogendijk
- Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | | | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, the Netherlands
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, the Netherlands
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| |
Collapse
|
18
|
Fiuza-Luces C, Valenzuela PL, Gálvez BG, Ramírez M, López-Soto A, Simpson RJ, Lucia A. The effect of physical exercise on anticancer immunity. Nat Rev Immunol 2024; 24:282-293. [PMID: 37794239 DOI: 10.1038/s41577-023-00943-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/06/2023]
Abstract
Regular physical activity is associated with lower cancer incidence and mortality, as well as with a lower rate of tumour recurrence. The epidemiological evidence is supported by preclinical studies in animal models showing that regular exercise delays the progression of cancer, including highly aggressive malignancies. Although the mechanisms underlying the antitumorigenic effects of exercise remain to be defined, an improvement in cancer immunosurveillance is likely important, with different immune cell subtypes stimulated by exercise to infiltrate tumours. There is also evidence that immune cells from blood collected after an exercise bout could be used as adoptive cell therapy for cancer. In this Perspective, we address the importance of muscular activity for maintaining a healthy immune system and discuss the effects of a single bout of exercise (that is, 'acute' exercise) and those of 'regular' exercise (that is, repeated bouts) on anticancer immunity, including tumour infiltrates. We also address the postulated mechanisms and the clinical implications of this emerging area of research.
Collapse
Affiliation(s)
- Carmen Fiuza-Luces
- Physical Activity and Health Research Group ('PaHerg'), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain.
| | - Pedro L Valenzuela
- Physical Activity and Health Research Group ('PaHerg'), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain
- Systems Biology Department, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Beatriz G Gálvez
- Physical Activity and Health Research Group ('PaHerg'), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Ramírez
- Oncohematology Unit, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Biomedical Research Foundation, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- La Princesa Institute of Heah, Madrid, Spain
| | - Alejandro López-Soto
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Asturias, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Asturias, Spain.
| | - Richard J Simpson
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ, USA
- Department of Paediatrics, The University of Arizona, Tucson, AZ, USA
- Department of Immunobiology, The University of Arizona, Tucson, AZ, USA
| | - Alejandro Lucia
- CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, Spain.
- Faculty of Sport Sciences, Universidad Europea, Madrid, Spain.
| |
Collapse
|
19
|
Yamakawa M, Rexach JE. Cell States and Interactions of CD8 T Cells and Disease-Enriched Microglia in Human Brains with Alzheimer's Disease. Biomedicines 2024; 12:308. [PMID: 38397909 PMCID: PMC10886701 DOI: 10.3390/biomedicines12020308] [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: 12/01/2023] [Revised: 01/05/2024] [Accepted: 01/13/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a multi-stage neurodegenerative disorder characterized by beta-amyloid accumulation, hyperphosphorylated Tau deposits, neurodegeneration, neuroinflammation, and cognitive impairment. Recent studies implicate CD8 T cells as neuroimmune responders to the accumulation of AD pathology in the brain and potential contributors to toxic neuroinflammation. However, more evidence is needed to understand lymphocytes in disease, including their functional states, molecular mediators, and interacting cell types in diseased brain tissue. The scarcity of lymphocytes in brain tissue samples has limited the unbiased profiling of disease-associated cell types, cell states, drug targets, and relationships to common AD genetic risk variants based on transcriptomic analyses. However, using recent large-scale, high-quality single-nuclear sequencing datasets from over 84 Alzheimer's disease and control cases, we leverage single-nuclear RNAseq data from 800 lymphocytes collected from 70 individuals to complete unbiased molecular profiling. We demonstrate that effector memory CD8 T cells are the major lymphocyte subclass enriched in the brain tissues of individuals with AD dementia. We define disease-enriched interactions involving CD8 T cells and multiple brain cell subclasses including two distinct microglial disease states that correlate, respectively, to beta-amyloid and tau pathology. We find that beta-amyloid-associated microglia are a major hub of multicellular cross-talk gained in disease, including interactions involving both vulnerable neuronal subtypes and CD8 T cells. We reproduce prior reports that amyloid-response microglia are depleted in APOE4 carriers. Overall, these human-based studies provide additional support for the potential relevance of effector memory CD8 T cells as a lymphocyte population of interest in AD dementia and provide new candidate interacting partners and drug targets for further functional study.
Collapse
Affiliation(s)
| | - Jessica E. Rexach
- Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA;
| |
Collapse
|
20
|
Russell J, Chen L, Liu A, Wang J, Ghosh S, Zhong X, Shi H, Beutler B, Nair-Gill E. Lrp10 suppresses IL7R limiting CD8 T cell homeostatic expansion and anti-tumor immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.08.570738. [PMID: 38106103 PMCID: PMC10723380 DOI: 10.1101/2023.12.08.570738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Signals emanating from the T cell receptor (TCR), co-stimulatory receptors, and cytokine receptors each influence CD8 T cell fate. Understanding how these signals respond to homeostatic and microenvironmental cues can reveal new ways to therapeutically direct T cell function. Through forward genetic screening in mice, we discovered that loss-of-function mutations in LDL receptor related protein 10 ( Lrp10 ) caused naïve and central memory CD8 T cells to accumulate in peripheral lymphoid organs. Lrp10 encodes a conserved cell surface protein of unknown immunological function. Lrp10 was induced with T cell activation and its expression post-translationally suppressed IL7 receptor (IL7R) levels. Accordingly, Lrp10 deletion enhanced T cell homeostatic expansion through IL7R signaling. Lrp10 -deficient mice were also intrinsically resistant to syngeneic tumors. This phenotype depended on dense tumor infiltration of CD8 T cells that displayed increased memory cell characteristics, reduced terminal exhaustion, and augmented responses to immune checkpoint inhibition. Here, we present Lrp10 as a new negative regulator of CD8 T cell homeostasis and a host factor that controls tumor resistance with implications for immunotherapy.
Collapse
|
21
|
Luo M, Gong W, Zhang Y, Li H, Ma D, Wu K, Gao Q, Fang Y. New insights into the stemness of adoptively transferred T cells by γc family cytokines. Cell Commun Signal 2023; 21:347. [PMID: 38049832 PMCID: PMC10694921 DOI: 10.1186/s12964-023-01354-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/11/2023] [Indexed: 12/06/2023] Open
Abstract
T cell-based adoptive cell therapy (ACT) has exhibited excellent antitumoral efficacy exemplified by the clinical breakthrough of chimeric antigen receptor therapy (CAR-T) in hematologic malignancies. It relies on the pool of functional T cells to retain the developmental potential to serially kill targeted cells. However, failure in the continuous supply and persistence of functional T cells has been recognized as a critical barrier to sustainable responses. Conferring stemness on infused T cells, yielding stem cell-like memory T cells (TSCM) characterized by constant self-renewal and multilineage differentiation similar to pluripotent stem cells, is indeed necessary and promising for enhancing T cell function and sustaining antitumor immunity. Therefore, it is crucial to identify TSCM cell induction regulators and acquire more TSCM cells as resource cells during production and after infusion to improve antitumoral efficacy. Recently, four common cytokine receptor γ chain (γc) family cytokines, encompassing interleukin-2 (IL-2), IL-7, IL-15, and IL-21, have been widely used in the development of long-lived adoptively transferred TSCM in vitro. However, challenges, including their non-specific toxicities and off-target effects, have led to substantial efforts for the development of engineered versions to unleash their full potential in the induction and maintenance of T cell stemness in ACT. In this review, we summarize the roles of the four γc family cytokines in the orchestration of adoptively transferred T cell stemness, introduce their engineered versions that modulate TSCM cell formation and demonstrate the potential of their various combinations. Video Abstract.
Collapse
Affiliation(s)
- Mengshi Luo
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjian Gong
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuewen Zhang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huayi Li
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Ma
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinglei Gao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yong Fang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
22
|
Jackman RP, Darst O, Gaillard B, Tran JQ, Tomayko MM, Muench MO. Enhanced alloresponse to platelet transfusion due to immune dysregulation following ablative chemotherapy in mice. Front Immunol 2023; 14:1281123. [PMID: 38090570 PMCID: PMC10711281 DOI: 10.3389/fimmu.2023.1281123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction Alloimmunization is common following platelet transfusion and can result in negative outcomes for recipients such as refractoriness to subsequent transfusions and rejection of transplants. Healthy people do not receive blood transfusions, and the diseases and therapies that result in a need to transfuse have significant impacts on the immunological environment to which these alloantigens are introduced. Ablative chemotherapies are common among platelet recipients and have potent immunological effects. In this study, we modeled the impact of chemotherapy on the alloresponse to platelet transfusion. As chemotherapies are generally regarded as immunosuppressive, we hypothesized that that they would result in a diminished alloresponse. Methods Mice were given a combination chemotherapeutic treatment of cytarabine and doxorubicin followed by transfusion of allogeneic platelets, and compared to controls given no treatment, chemotherapy alone, or transfusion alone. Alloantibody responses were measured 2 weeks after transfusion, and cellular responses and growth factors were monitored over time. Results Contrary to our hypothesis, we found that chemotherapy led to increased alloantibody responses to allogeneic platelet transfusion. This enhanced response was antigen-specific and was associated with increased CD4+ and CD8+ T cell responses. Chemotherapy led to rapid lymphocyte depletion followed by reconstitution, non-specific activation of transitional B cells with the highest levels of activation in the least mature subsets, and increased serum levels of B cell activating factor (BAFF). Conclusion These data suggest that ablative chemotherapy can increase the risk of alloimmunization and, if confirmed clinically, that additional measures to protect these patient populations may be warranted.
Collapse
Affiliation(s)
- Rachael P. Jackman
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Orsolya Darst
- Vitalant Research Institute, San Francisco, CA, United States
| | - Betty Gaillard
- Vitalant Research Institute, San Francisco, CA, United States
| | - Johnson Q. Tran
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Mary M. Tomayko
- Department of Dermatology, Yale School of Medicine, New Haven, CT, United States
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States
| | - Marcus O. Muench
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
23
|
Ju T, Jiang D, Zhong C, Zhang H, Huang Y, Zhu C, Yang S, Yan D. Characteristics of circulating immune cells in HBV-related acute-on-chronic liver failure following artificial liver treatment. BMC Immunol 2023; 24:47. [PMID: 38007423 PMCID: PMC10676598 DOI: 10.1186/s12865-023-00579-8] [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: 07/18/2023] [Accepted: 10/19/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND AND AIM Liver failure, which is predominantly caused by hepatitis B (HBV) can be improved by an artificial liver support system (ALSS). This study investigated the phenotypic heterogeneity of immunocytes in patients with HBV-related acute-on-chronic liver failure (HBV-ACLF) before and after ALSS therapy. METHODS A total of 22 patients with HBV-ACLF who received ALSS therapy were included in the study. Patients with Grade I according to the ACLF Research Consortium score were considered to have improved. Demographic and laboratory data were collected and analyzed during hospitalization. Immunological features of peripheral blood in the patients before and after ALSS were detected by mass cytometry analyses. RESULTS In total, 12 patients improved and 10 patients did not. According to the immunological features data after ALSS, the proportion of circulating monocytes was significantly higher in non-improved patients, but there were fewer γδT cells compared with those in improved patients. Characterization of 37 cell clusters revealed that the frequency of effector CD8+ T (P = 0.003), CD4+ TCM (P = 0.033), CD4+ TEM (P = 0.039), and inhibitory natural killer (NK) cells (P = 0.029) decreased in HBV-ACLF patients after ALSS therapy. Sub group analyses after treatment showed that the improved patients had higher proportions of CD4+ TCM (P = 0.010), CD4+ TEM (P = 0.021), and γδT cells (P = 0.003) and a lower proportion of monocytes (P = 0.012) compared with the non-improved patients. CONCLUSIONS Changes in effector CD8+ T cells, effector and memory CD4+ T cells, and inhibitory NK cells are associated with ALSS treatment of HBV-ACLF. Moreover, monocytes and γδT cells exhibited the main differences when patients obtained different prognoses. The phenotypic heterogeneity of lymphocytes and monocytes may contribute to the prognosis of ALSS and future immunotherapy strategies.
Collapse
Affiliation(s)
- Tao Ju
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Daixi Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Chengli Zhong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Huafen Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Yandi Huang
- Department of Laboratory Medicine, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310003, China
| | - Chunxia Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Shigui Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China.
| | - Dong Yan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China.
| |
Collapse
|
24
|
Li F, Chen D, Zeng Q, Du Y. Possible Mechanisms of Lymphopenia in Severe Tuberculosis. Microorganisms 2023; 11:2640. [PMID: 38004652 PMCID: PMC10672989 DOI: 10.3390/microorganisms11112640] [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: 09/14/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Tuberculosis (TB) is a chronic infectious disease caused by Mycobacterium tuberculosis (M. tuberculosis). In lymphopenia, T cells are typically characterized by progressive loss and a decrease in their count results. Lymphopenia can hinder immune responses and lead to systemic immunosuppression, which is strongly associated with mortality. Lymphopenia is a significant immunological abnormality in the majority of patients with severe and advanced TB, and its severity is linked to disease outcomes. However, the underlying mechanism remains unclear. Currently, the research on the pathogenesis of lymphopenia during M. tuberculosis infection mainly focuses on how it affects lymphocyte production, survival, or tissue redistribution. This includes impairing hematopoiesis, inhibiting T-cell proliferation, and inducing lymphocyte apoptosis. In this study, we have compiled the latest research on the possible mechanisms that may cause lymphopenia during M. tuberculosis infection. Lymphopenia may have serious consequences in severe TB patients. Additionally, we discuss in detail potential intervention strategies to prevent lymphopenia, which could help understand TB immunopathogenesis and achieve the goal of preventing and treating severe TB.
Collapse
Affiliation(s)
- Fei Li
- Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (D.C.); (Q.Z.); (Y.D.)
| | | | | | | |
Collapse
|
25
|
Dower WJ, Park AI, Bakker AV, Cwirla SE, Pongtornpipat P, Williams BM, Joshi P, Baxter BA, Needels MC, Barrett RW. A mechanistically novel peptide agonist of the IL-7 receptor that addresses limitations of IL-7 cytokine therapy. PLoS One 2023; 18:e0286834. [PMID: 37874823 PMCID: PMC10597491 DOI: 10.1371/journal.pone.0286834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023] Open
Abstract
Interleukin (IL)-7 is broadly active on T-cell populations, and modified versions have been clinically evaluated for a variety of therapeutic applications, including cancer, lymphopenia, and infectious diseases; and found to be relatively well-tolerated and biologically active. Here we describe novel IL-7R agonists that are unrelated in structure to IL-7, bind to the receptor subunits differently from IL-7, but closely emulate IL-7 biology. The small size, low structural complexity, and the natural amino acid composition of the pharmacologically active peptide MDK1472 allows facile incorporation into protein structures, such as the IgG2-Fc fusion MDK-703. This molecule possesses properties potentially better suited to therapeutic applications than native IL-7 or its derivatives. We compared these compounds with IL-7 for immune cell selectivity, induction of IL-7R signaling, receptor-mediated internalization, proliferation, and generation of immune cell phenotypes in human and non-human primate (NHP) peripheral blood cells in vitro; and found them to be similar in biological activity to IL-7. In cynomolgus macaques, MDK-703 exhibits a circulating half-life of 46 hr and produces sustained T-cell expansion characteristic of IL-7 treatment. In the huCD34+-engrafted NSG mouse model of the human immune system, MDK-703 induces an immune cell profile very similar to that generated by IL-7-derived compounds; including the pronounced expansion of memory T-cells, particularly the population of stem-like memory T-cells (Tscm) which may be important for anti-tumor activities reported with IL-7 treatment. Clinical administration of IL-7 and modified variants has been reported to induce anti-drug antibodies (ADAs), including IL-7 neutralizing antibodies. The novel peptide agonist reported here scores very low in predicted immunogenicity, and because the peptide lacks sequence similarity with IL-7, the problematic immunogenic neutralization of endogenous cytokine should not occur. The properties we report here implicate MDK-703 as a candidate for clinical evaluation in oncology, anti-viral and other infectious disease, vaccine enhancement, and treatment of lymphopenia.
Collapse
Affiliation(s)
- William J. Dower
- Medikine, Inc., Menlo Park, California, United States of America
| | | | - Alice V. Bakker
- Medikine, Inc., Menlo Park, California, United States of America
| | - Steven E. Cwirla
- Medikine, Inc., Menlo Park, California, United States of America
| | | | - Blake M. Williams
- Medikine, Inc., Menlo Park, California, United States of America
- Department of Biomedical Engineering, and Center for Quantitative Bioinformatics and Quantitative Biology, Colleges of Engineering and Medicine, University of Illinois Chicago, IL, United States of America
| | - Prarthana Joshi
- Medikine, Inc., Menlo Park, California, United States of America
| | - Bryan A. Baxter
- Medikine, Inc., Menlo Park, California, United States of America
| | | | | |
Collapse
|
26
|
Ji Z, Lu W, Wu S, Zhang Y, Meng D, Zhang X, Dai X, Chen H, Ma L, Sun Y, Jiang L, Kong X. Single-Cell RNA-Sequencing Reveals Peripheral T Helper Cells Promoting the Development of IgG4-Related Disease by Enhancing B Cell Activation and Differentiation. Int J Mol Sci 2023; 24:13735. [PMID: 37762039 PMCID: PMC10530310 DOI: 10.3390/ijms241813735] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/17/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Abnormal B cell differentiation plays a critical role in IgG4-related disease (IgG4-RD), but the underlying mechanism remains largely unknown. We investigated the cell landscape from three IgG4-RD retroperitoneal tissues and three control tissues using single-cell RNA-sequencing. Critical cell type or markers were further validated in the peripheral blood from the patients with IgG4-RD and healthy controls via flow cytometry as well as in the IgG4-RD and control tissue via immunofluorescence staining. The increases in B cells, plasma cells, and CD4+ T cells were found in IgG4-RD retroperitoneal tissue. Importantly, among CD4+ T cells, an increase in CD4+CXCR5-PD1hi peripheral T helper (Tph) cells with a high expression of IL-21 and TIGIT was discovered in IgG4-RD tissue, which was further validated in peripheral blood of the patients with IgG4-RD. The Tph cell and TIGIT+ Tph cell proportion were remarkably higher in active IgG4-RD patients and correlated with disease activity. Moreover, TIGIT+CD4+ cells were able to promote B cell differentiation via IL-21. Our study revealed that Tph cells are increased in IgG4-RD and probably play critical roles in B cell differentiation through TIGIT-IL-21 axis. Peripheral Tph cell and TIGIT+Tph cell are potential markers for IgG4-RD disease activity.
Collapse
Affiliation(s)
- Zongfei Ji
- Department of Rheumatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China; (Z.J.)
| | - Weiqi Lu
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Sifan Wu
- Department of Rheumatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China; (Z.J.)
| | - Yong Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Dan Meng
- Department of Rheumatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China; (Z.J.)
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiao Zhang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China; (Z.J.)
| | - Xiaojuan Dai
- Department of Rheumatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China; (Z.J.)
| | - Huiyong Chen
- Department of Rheumatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China; (Z.J.)
| | - Lili Ma
- Department of Rheumatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China; (Z.J.)
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ying Sun
- Department of Rheumatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China; (Z.J.)
| | - Lindi Jiang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China; (Z.J.)
- Evidence-Based Medicine Center, Fudan University, Shanghai 200032, China
| | - Xiufang Kong
- Department of Rheumatology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China; (Z.J.)
- Evidence-Based Medicine Center, Fudan University, Shanghai 200032, China
| |
Collapse
|
27
|
Collier-Bain HD, Brown FF, Causer AJ, Emery A, Oliver R, Moore S, Murray J, Turner JE, Campbell JP. Harnessing the immunomodulatory effects of exercise to enhance the efficacy of monoclonal antibody therapies against B-cell haematological cancers: a narrative review. Front Oncol 2023; 13:1244090. [PMID: 37681023 PMCID: PMC10482436 DOI: 10.3389/fonc.2023.1244090] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) are standard care for many B-cell haematological cancers. The modes of action for these mAbs include: induction of cancer cell lysis by activating Fcγ-receptors on innate immune cells; opsonising target cells for antibody-dependent cellular cytotoxicity or phagocytosis, and/or triggering the classical complement pathway; the simultaneous binding of cancer cells with T-cells to create an immune synapse and activate perforin-mediated T-cell cytotoxicity against cancer cells; blockade of immune checkpoints to facilitate T-cell cytotoxicity against immunogenic cancer cell clones; and direct delivery of cytotoxic agents via internalisation of mAbs by target cells. While treatment regimens comprising mAb therapy can lead to durable anti-cancer responses, disease relapse is common due to failure of mAb therapy to eradicate minimal residual disease. Factors that limit mAb efficacy include: suboptimal effector cell frequencies, overt immune exhaustion and/or immune anergy, and survival of diffusely spread tumour cells in different stromal niches. In this review, we discuss how immunomodulatory changes arising from exposure to structured bouts of acute exercise might improve mAb treatment efficacy by augmenting (i) antibody-dependent cellular cytotoxicity, (ii) antibody-dependent cellular phagocytosis, (iii) complement-dependent cytotoxicity, (iv) T-cell cytotoxicity, and (v) direct delivery of cytotoxic agents.
Collapse
Affiliation(s)
| | - Frankie F. Brown
- Department for Health, University of Bath, Bath, United Kingdom
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom
| | - Adam J. Causer
- Department for Health, University of Bath, Bath, United Kingdom
| | - Annabelle Emery
- Department for Health, University of Bath, Bath, United Kingdom
| | - Rebecca Oliver
- Department for Health, University of Bath, Bath, United Kingdom
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, Bath, United Kingdom
| | - Sally Moore
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, Bath, United Kingdom
| | - James Murray
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, Bath, United Kingdom
| | - James E. Turner
- Department for Health, University of Bath, Bath, United Kingdom
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | | |
Collapse
|
28
|
Roberts J, Chevalier A, Hawerkamp HC, Yeow A, Matarazzo L, Schwartz C, Hams E, Fallon PG. Retinoic Acid-Related Orphan Receptor α Is Required for Generation of Th2 Cells in Type 2 Pulmonary Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:626-632. [PMID: 37387671 PMCID: PMC10404816 DOI: 10.4049/jimmunol.2200896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 06/08/2023] [Indexed: 07/01/2023]
Abstract
The transcription factor retinoic acid-related orphan receptor α (RORα) is important in regulating several physiological functions, such as cellular development, circadian rhythm, metabolism, and immunity. In two in vivo animal models of type 2 lung inflammation, Nippostrongylus brasiliensis infection and house dust mite (HDM) sensitization, we show a role for Rora in Th2 cellular development during pulmonary inflammation. N. brasiliensis infection and HDM challenge induced an increase in frequency of Rora-expressing GATA3+CD4 T cells in the lung. Using staggerer mice, which have a ubiquitous deletion of functional RORα, we generated bone marrow chimera mice, and we observed a delayed worm expulsion and reduced frequency in the expansion of Th2 cells and innate lymphoid type 2 cells (ILC2s) in the lungs after N. brasiliensis infection. ILC2-deficient mouse (Rorafl/flIl7raCre) also had delayed worm expulsion with associated reduced frequency of Th2 cells and ILC2s in the lungs after N. brasiliensis infection. To further define the role for Rora-expressing Th2 cells, we used a CD4-specific Rora-deficient mouse (Rorafl/flCD4Cre), with significantly reduced frequency of lung Th2 cells, but not ILC2, after N. brasiliensis infection and HDM challenge. Interestingly, despite the reduction in pulmonary Th2 cells in Rorafl/flCD4Cre mice, this did not impact the expulsion of N. brasiliensis after primary and secondary infection, or the generation of lung inflammation after HDM challenge. This study demonstrates a role for RORα in Th2 cellular development during pulmonary inflammation that could be relevant to the range of inflammatory diseases in which RORα is implicated.
Collapse
Affiliation(s)
- Joseph Roberts
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Anne Chevalier
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Heike C. Hawerkamp
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Aoife Yeow
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Laura Matarazzo
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Christian Schwartz
- Mikrobiologisches Institut–Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Emily Hams
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Padraic G. Fallon
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
29
|
Marton C, Minaud A, Coupet CA, Chauvin M, Dhiab J, Vallet H, Boddaert J, Kehrer N, Bastien B, Inchauspe G, Barraud L, Sauce D. IL-7 producing immunotherapy improves ex vivo T cell functions of immunosenescent patients, especially post hip fracture. Hum Vaccin Immunother 2023; 19:2232247. [PMID: 37417353 PMCID: PMC10332238 DOI: 10.1080/21645515.2023.2232247] [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: 12/21/2022] [Accepted: 06/29/2023] [Indexed: 07/08/2023] Open
Abstract
Following acute stress such as trauma or sepsis, most of critically ill elderly patients become immunosuppressed and susceptible to secondary infections and enhanced mortality. We have developed a virus-based immunotherapy encoding human interleukin-7 (hIL-7) aiming at restoring both innate an adaptative immune homeostasis in these patients. We assessed the impact of this encoded hIL-7 on the ex vivo immune functions of T cells from PBMC of immunosenescent patients with or without hip fracture. T-cell ex vivo phenotyping was characterized in terms of senescence (CD57), IL-7 receptor (CD127) expression, and T cell differentiation profile. Then, post stimulation, activation status, and functionality (STAT5/STAT1 phosphorylation and T cell proliferation assays) were evaluated by flow cytometry. Our data show that T cells from both groups display immunosenescence features, express CD127 and are activated after stimulation by virotherapy-produced hIL-7-Fc. Interestingly, hip fracture patients exhibit a unique functional ability: An important T cell proliferation occurred compared to controls following stimulation with hIL-7-Fc. In addition, stimulation led to an increased naïve T cell as well as a decreased effector memory T cell proportions compared to controls. This preliminary study indicates that the produced hIL-7-Fc is well recognized by T cells and initiates IL-7 signaling through STAT5 and STAT1 phosphorylation. This signaling efficiently leads to T cell proliferation and activation and enables a T cell "rejuvenation." These results are in favor of the clinical development of the hIL-7-Fc expressing virotherapy to restore or induce immune T cell responses in immunosenescent hip fracture patients.
Collapse
Affiliation(s)
- Chrystel Marton
- Centre d’Immunologie et des Maladies Infectieuses, Sorbonne Université, Inserm, CIMI-Paris, Paris, France
- ImmmunResQ Department, Transgene, Lyon, France
| | - Alix Minaud
- Centre d’Immunologie et des Maladies Infectieuses, Sorbonne Université, Inserm, CIMI-Paris, Paris, France
| | | | - Manon Chauvin
- Centre d’Immunologie et des Maladies Infectieuses, Sorbonne Université, Inserm, CIMI-Paris, Paris, France
| | - Jamila Dhiab
- Centre d’Immunologie et des Maladies Infectieuses, Sorbonne Université, Inserm, CIMI-Paris, Paris, France
| | - Hélène Vallet
- Centre d’Immunologie et des Maladies Infectieuses, Sorbonne Université, Inserm, CIMI-Paris, Paris, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Unité de Gériatrie Aigue, Paris, France
| | - Jacques Boddaert
- Centre d’Immunologie et des Maladies Infectieuses, Sorbonne Université, Inserm, CIMI-Paris, Paris, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpétrière, Unité périopératoire gériatrique, Paris, France
| | | | | | | | - Luc Barraud
- ImmmunResQ Department, Transgene, Lyon, France
| | - Delphine Sauce
- Centre d’Immunologie et des Maladies Infectieuses, Sorbonne Université, Inserm, CIMI-Paris, Paris, France
| |
Collapse
|
30
|
Hesari M, Attar Z, Soltani-Shirazi S, Keshavarzian O, Taheri R, Tabrizi R, Fouladseresht H. The Therapeutic Values of IL-7/IL-7R and the Recombinant Derivatives in Glioma: A Narrative Review. J Interferon Cytokine Res 2023; 43:319-334. [PMID: 37566474 DOI: 10.1089/jir.2023.0050] [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] [Indexed: 08/13/2023] Open
Abstract
Interleukin-7 (IL-7) is essential for maintaining the immune system's defense functions by regulating the development and homeostasis of lymphocytes. Findings have shown the high efficacy of IL-7/IL-7 receptor (IL-7R)-based immunotherapy on various malignancies, with confirmation in both animal models and humans. In recent years, the progression-free survival and overall survival of patients suffering from gliomas significantly increased by introducing C7R-expressing chimeric antigen receptor (CAR)-T cells and long-acting IL-7 agonists such as NT-I7 (rhIL-7-hyFc, Efineptakin alfa). However, the effect of IL-7-based immunotherapies on the resistance of tumor cells to chemotherapy (when used simultaneously with chemotherapy agents) is still ambiguous and requires further studies. This article first reviews the pathophysiological roles of IL-7/IL-7R in tumors, focusing on gliomas. Subsequently, it discusses the therapeutic values of IL-7/IL-7R and the recombinant derivatives in gliomas.
Collapse
Affiliation(s)
| | - Zeinab Attar
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Department of Pharmacology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shakiba Soltani-Shirazi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Reza Taheri
- Department of Neurosurgery, Fasa University of Medical Sciences, Fasa, Iran
| | - Reza Tabrizi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Hamed Fouladseresht
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
31
|
Micevic G, Daniels A, Flem-Karlsen K, Park K, Talty R, McGeary M, Mirza H, Blackburn HN, Sefik E, Cheung JF, Hornick NI, Aizenbud L, Joshi NS, Kluger H, Iwasaki A, Bosenberg MW, Flavell RA. IL-7R licenses a population of epigenetically poised memory CD8 + T cells with superior antitumor efficacy that are critical for melanoma memory. Proc Natl Acad Sci U S A 2023; 120:e2304319120. [PMID: 37459511 PMCID: PMC10372654 DOI: 10.1073/pnas.2304319120] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023] Open
Abstract
Recurrence of advanced melanoma after therapy is a major risk factor for reduced survival, and treatment options are limited. Antitumor immune memory plays a critical role in preventing melanoma recurrence and memory T cells could be a potent cell-based therapy, but the identity, and functional properties of the required immune cells are incompletely understood. Here, we show that an IL-7Rhi tumor-specific CD8+ population is critical for antitumor memory and can be epigenetically augmented to drive powerful antitumor immune responses. Using a model of functional antimelanoma memory, we found that high IL-7R expression selectively marks a CD8+ population in lymphoid organs that plays critical roles in maintaining tumor remission after immunotherapy or surgical resection. This population has intrinsic cytotoxic activity, lacks markers of exhaustion and has superior antitumor efficacy. IL-7Rhi cells have a functionally poised epigenetic landscape regulated by DNA methylation, which can be augmented by hypomethylating agents to confer improved survival and complete melanoma clearance in naive mice. Importantly, greater than 95% of tumor-specific T cells in draining lymph nodes after therapy express high levels of IL-7R. This overlap between IL-7Rhi and antigen-specific T cells allows for enrichment of a potent functional CD8+ population without determining antigen-specificity, which we demonstrate in a melanoma model without a known antigen. We identify that IL-7R expression in human melanoma is an independent prognostic factor of improved survival. These findings advance our basic understanding of antitumor memory and suggest a cell-based therapy using high IL-7R expression to enrich for a lymph node population with superior antitumor activity that can be augmented by hypomethylating agents.
Collapse
Affiliation(s)
- Goran Micevic
- Department of Immunobiology, Yale School of Medicine, New Haven, CT06520
- Department of Dermatology, Yale School of Medicine, New Haven, CT06520
| | - Andrew Daniels
- Department of Immunobiology, Yale School of Medicine, New Haven, CT06520
- Department of Pathology, Yale School of Medicine, New Haven, CT06520
| | | | - Koonam Park
- Department of Dermatology, Yale School of Medicine, New Haven, CT06520
| | - Ronan Talty
- Department of Pathology, Yale School of Medicine, New Haven, CT06520
| | - Meaghan McGeary
- Department of Pathology, Yale School of Medicine, New Haven, CT06520
| | - Haris Mirza
- Department of Immunobiology, Yale School of Medicine, New Haven, CT06520
- Department of Pathology, Yale School of Medicine, New Haven, CT06520
| | - Holly N. Blackburn
- Department of Immunobiology, Yale School of Medicine, New Haven, CT06520
- Department of Surgery, Yale School of Medicine, New Haven, CT06520
| | - Esen Sefik
- Department of Immunobiology, Yale School of Medicine, New Haven, CT06520
| | - Julie F. Cheung
- Department of Immunobiology, Yale School of Medicine, New Haven, CT06520
| | - Noah I. Hornick
- Department of Immunobiology, Yale School of Medicine, New Haven, CT06520
| | - Lilach Aizenbud
- Yale Cancer Center, Yale School of Medicine, New Haven, CT06520
- Department of Medicine (Medical Oncology), Yale School of Medicine, New Haven, CT06520
| | - Nikhil S. Joshi
- Department of Immunobiology, Yale School of Medicine, New Haven, CT06520
| | - Harriet Kluger
- Yale Cancer Center, Yale School of Medicine, New Haven, CT06520
- Department of Medicine (Medical Oncology), Yale School of Medicine, New Haven, CT06520
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT06520
| | - Akiko Iwasaki
- Department of Immunobiology, Yale School of Medicine, New Haven, CT06520
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT06520
- HHMI, Chevy Chase, MD20815
| | - Marcus W. Bosenberg
- Department of Immunobiology, Yale School of Medicine, New Haven, CT06520
- Department of Dermatology, Yale School of Medicine, New Haven, CT06520
- Department of Pathology, Yale School of Medicine, New Haven, CT06520
- Yale Cancer Center, Yale School of Medicine, New Haven, CT06520
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT06520
- Yale Center for Immuno-Oncology, Yale School of Medicine, New Haven, CT06520
| | - Richard A. Flavell
- Department of Immunobiology, Yale School of Medicine, New Haven, CT06520
- Yale Cancer Center, Yale School of Medicine, New Haven, CT06520
- HHMI, Chevy Chase, MD20815
| |
Collapse
|
32
|
Shireman JM, Gonugunta N, Zhao L, Pattnaik A, Distler E, Her S, Wang X, Das R, Galipeau J, Dey M. GM-CSF and IL-7 fusion cytokine engineered tumor vaccine generates long-term Th-17 memory cells and increases overall survival in aged syngeneic mouse models of glioblastoma. Aging Cell 2023; 22:e13864. [PMID: 37165998 PMCID: PMC10352573 DOI: 10.1111/acel.13864] [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: 02/02/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/12/2023] Open
Abstract
Age-related immune dysfunctions, such as decreased T-cell output, are closely related to pathologies like cancers and lack of vaccine efficacy among the elderly. Engineered fusokine, GIFT-7, a fusion of interleukin 7 (IL-7) and GM-CSF, can reverse aging-related lymphoid organ atrophy. We generated a GIFT-7 fusokine tumor vaccine and employed it in aged syngeneic mouse models of glioblastoma and found that peripheral vaccination with GIFT-7TVax resulted in thymic regeneration and generated durable long-term antitumor immunity specifically in aged mice. Global cytokine analysis showed increased pro-inflammatory cytokines including IL-1β in the vaccinated group that resulted in hyperactivation of dendritic cells. In addition, GIFT-7 vaccination resulted in increased T-cell trafficking to the brain and robust Th-17 long-term effector memory T-cell formation. TCR-seq analysis showed increased productive frequency among detected rearrangements within the vaccinated group. Overall, our data demonstrate that aging immune system can be therapeutically augmented to generate lasting antitumor immunity.
Collapse
Affiliation(s)
- Jack M. Shireman
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Nikita Gonugunta
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Lei Zhao
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Akshita Pattnaik
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Emily Distler
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Skyler Her
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Xiaohu Wang
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Rahul Das
- Department of Medicine, Division of Hematology and OncologyUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Jaques Galipeau
- Department of Medicine, Division of Hematology and OncologyUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| | - Mahua Dey
- Department of NeurosurgeryUniversity of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center,MadisonWisconsinUSA
| |
Collapse
|
33
|
Li F, Ma Y, Li X, Zhang D, Han J, Tan D, Mi Y, Yang X, Wang J, Zhu B. Severe persistent mycobacteria antigen stimulation causes lymphopenia through impairing hematopoiesis. Front Cell Infect Microbiol 2023; 13:1079774. [PMID: 36743311 PMCID: PMC9889370 DOI: 10.3389/fcimb.2023.1079774] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/02/2023] [Indexed: 01/20/2023] Open
Abstract
Miliary tubersculosis (TB), an acute systemic blood disseminated tuberculosis mainly caused by Mycobacterium tuberculosis (M. tuberculosis), can cause signs of lymphopenia in clinical patients. To investigate whether/how persistent mycobacteria antigen stimulation impairs hematopoiesis and the therapeutic effect of interleukin-7 (IL-7), a mouse model of Mycobacterium Bovis Bacillus Calmette-Guérin (BCG) intravenous infection with/without an additional stimulation with M. tuberculosis multi-antigen cocktail containing ESAT6-CFP10 (EC) and Mtb10.4-HspX (MH) was established. Consistent with what happened in miliary TB, high dose of BCG intravenous infection with/without additional antigen stimulation caused lymphopenia in peripheral blood. In which, the levels of cytokines IFN-γ and TNF-α in serum increased, and consequently the expression levels of transcription factors Batf2 and IRF8 involved in myeloid differentiation were up-regulated, while the expression levels of transcription factors GATA2 and NOTCH1 involved in lymphoid commitment were down-regulated, and the proliferating activity of bone marrow (BM) lineage- c-Kit+ (LK) cells decreased. Furthermore, recombinant Adeno-Associated Virus 2-mediated IL-7 (rAAV2-IL-7) treatment could significantly promote the elevation of BM lymphoid progenitors. It suggests that persistent mycobacteria antigen stimulation impaired lymphopoiesis of BM hematopoiesis, which could be restored by complement of IL-7.
Collapse
Affiliation(s)
- Fei Li
- Gansu Provincial Key Laboratory of Evidence-Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yanlin Ma
- Gansu Provincial Key Laboratory of Evidence-Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiaoping Li
- Inpatient Ward 1, Lanzhou Pulmonary Hospital, Lanzhou, China
| | - Dan Zhang
- Gansu Provincial Key Laboratory of Evidence-Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jiangyuan Han
- Gansu Provincial Key Laboratory of Evidence-Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Daquan Tan
- Gansu Provincial Key Laboratory of Evidence-Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Youjun Mi
- Gansu Provincial Key Laboratory of Evidence-Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China,Institute of Pathophysiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiaojuan Yang
- Gansu Provincial Key Laboratory of Evidence-Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China,Department of Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Juan Wang
- Gansu Provincial Key Laboratory of Evidence-Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Bingdong Zhu
- Gansu Provincial Key Laboratory of Evidence-Based Medicine and Clinical Translation and Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China,State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou, China,*Correspondence: Bingdong Zhu,
| |
Collapse
|
34
|
Shin MS, Park HJ, Young J, Kang I. Implication of IL-7 receptor alpha chain expression by CD8 + T cells and its signature in defining biomarkers in aging. Immun Ageing 2022; 19:66. [PMID: 36544153 PMCID: PMC9768896 DOI: 10.1186/s12979-022-00324-6] [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: 06/30/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022]
Abstract
CD8+ T cells play an important role in host defense against infections and malignancies as well as contribute to the development of inflammatory disorders. Alterations in the frequency of naïve and memory CD8+ T cells are one of the most significant changes in the immune system with age. As the world population rapidly ages, a better understanding of aging immune function or immunosenescence could become a basis for discovering treatments of illnesses that commonly occur in older adults. In particular, biomarkers for immune aging could be utilized to identify individuals at high risk of developing age-associated conditions and help monitor the efficacy of therapeutic interventions targeting such conditions. This review details the possible role of CD8+ T cell subsets expressing different levels of the cytokine receptor IL-7 receptor alpha chain (IL-7Rα) and the gene signature associated with IL-7Rα as potential biomarkers for immune aging given the association of CD8+ T cells in host defense, inflammation, and immunosenescence.
Collapse
Affiliation(s)
- Min Sun Shin
- Departments of Internal Medicine, Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, S525C TAC, 300 Cedar Street, New Haven, CT, 06520, USA
| | - Hong-Jai Park
- Departments of Internal Medicine, Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, S525C TAC, 300 Cedar Street, New Haven, CT, 06520, USA
| | - Juan Young
- Departments of Psychiatry, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Insoo Kang
- Departments of Internal Medicine, Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, S525C TAC, 300 Cedar Street, New Haven, CT, 06520, USA.
| |
Collapse
|
35
|
Oh D, Choi H, Kim M, Cai L, Lee J, Jawad A, Kim S, Zheng H, Lee G, Jeon Y, Hyun SH. Interleukin-7 enhances in vitro development and blastocyst quality in porcine parthenogenetic embryos. Front Vet Sci 2022; 9:1052856. [PMID: 36570506 PMCID: PMC9772438 DOI: 10.3389/fvets.2022.1052856] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Interleukin-7 (IL-7), a vital factor that affects cell development, proliferation, and survival, plays an important role in oocyte maturation. However, its role in embryonic development remains unknown. Therefore, in this study, we aimed to investigate the effects of IL-7 supplementation on in vitro culture (IVC) of porcine embryos after parthenogenetic activation (PA) based on characteristics such as cleavage, blastocyst formation rate, intracellular glutathione (GSH) and reactive oxygen species (ROS) levels in cleaved embryos, total cell number, apoptosis rate, and cell lineage specification in blastocysts. Immunofluorescence revealed that IL-7 and its receptor, IL-7Rα (IL-7R) localized in the cytoplasm of porcine parthenote embryos. By supplementing the IVC medium (PZM5) with various concentrations of IL-7, an optimal concentration that enhanced embryonic development, promoted intracellular GSH, and decreased ROS levels in the cleavage stage during porcine embryo IVC was determined. Investigation of mRNA expression patterns via qRT-PCR suggested that IL-7 possibly regulated maternal mRNA clearance and zygotic genome activation. Furthermore, IL-7 supplementation reduced blastocyst apoptosis, enhanced the expression of the inner cell mass marker SOX2, and phosphorylated STAT5 levels in the blastocysts. Moreover, it altered the transcription patterns of genes that regulate apoptosis, IL-7 signaling, and development. Thus, we demonstrated the localization of IL-7 and IL-7R in porcine preimplantation embryos in vitro for the first time. Furthermore, we suggest that IL-7 supplementation can be employed to enhance embryonic development and blastocyst quality based on the activation of the transcripts of genes that are involved in developmental competence and IL-7 signaling during in vitro porcine embryo development following PA.
Collapse
Affiliation(s)
- Dongjin Oh
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, South Korea,Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea
| | - Hyerin Choi
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, South Korea,Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea
| | - Mirae Kim
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, South Korea,Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea
| | - Lian Cai
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, South Korea,Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea,Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, South Korea
| | - Joohyeong Lee
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, South Korea,Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea
| | - Ali Jawad
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, South Korea,Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea
| | - Sohee Kim
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, South Korea,Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea
| | - Haomiao Zheng
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, South Korea,Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea
| | - Gabsang Lee
- Department of Neurology, Institute for Cell Engineering, School of Medicine, Johns Hopkins Medicine, Baltimore, ML, United States
| | - Yubyeol Jeon
- Laboratory of Theriogenology and Reproductive Biotechnology, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea,*Correspondence: Yubyeol Jeon
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, South Korea,Institute of Stem Cell and Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, South Korea,Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, South Korea,Sang-Hwan Hyun
| |
Collapse
|
36
|
Taylor CA, Watson RA, Tong O, Ye W, Nassiri I, Gilchrist JJ, de Los Aires AV, Sharma PK, Koturan S, Cooper RA, Woodcock VK, Jungkurth E, Shine B, Coupe N, Payne MJ, Church DN, Naranbhai V, Groha S, Emery P, Mankia K, Freedman ML, Choueiri TK, Middleton MR, Gusev A, Fairfax BP. IL7 genetic variation and toxicity to immune checkpoint blockade in patients with melanoma. Nat Med 2022; 28:2592-2600. [PMID: 36526722 PMCID: PMC9800275 DOI: 10.1038/s41591-022-02095-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/18/2022] [Indexed: 12/23/2022]
Abstract
Treatment with immune checkpoint blockade (ICB) frequently triggers immune-related adverse events (irAEs), causing considerable morbidity. In 214 patients receiving ICB for melanoma, we observed increased severe irAE risk in minor allele carriers of rs16906115, intronic to IL7. We found that rs16906115 forms a B cell-specific expression quantitative trait locus (eQTL) to IL7 in patients. Patients carrying the risk allele demonstrate increased pre-treatment B cell IL7 expression, which independently associates with irAE risk, divergent immunoglobulin expression and more B cell receptor mutations. Consistent with the role of IL-7 in T cell development, risk allele carriers have distinct ICB-induced CD8+ T cell subset responses, skewing of T cell clonality and greater proportional repertoire occupancy by large clones. Finally, analysis of TCGA data suggests that risk allele carriers independently have improved melanoma survival. These observations highlight key roles for B cells and IL-7 in both ICB response and toxicity and clinical outcomes in melanoma.
Collapse
Affiliation(s)
- Chelsea A Taylor
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Robert A Watson
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| | - Orion Tong
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Weiyu Ye
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Isar Nassiri
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - James J Gilchrist
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Alba Verge de Los Aires
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Piyush Kumar Sharma
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Surya Koturan
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Rosalin A Cooper
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Victoria K Woodcock
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| | - Elsita Jungkurth
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Brian Shine
- Department of Clinical Biochemistry, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Nicholas Coupe
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| | - Miranda J Payne
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| | - David N Church
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Vivek Naranbhai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Center for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Stefan Groha
- Department of Medical Oncology, Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard & MIT, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- National Institute for Health Research Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Kulveer Mankia
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- National Institute for Health Research Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Matthew L Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard & MIT, Cambridge, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard & MIT, Cambridge, MA, USA
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Mark R Middleton
- Department of Oncology, University of Oxford, Oxford, UK
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Alexander Gusev
- Department of Medical Oncology, Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard & MIT, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
| | - Benjamin P Fairfax
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
- Department of Oncology, University of Oxford, Oxford, UK.
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
37
|
Choi JO, Ham JH, Hwang SS. RNA Metabolism in T Lymphocytes. Immune Netw 2022; 22:e39. [PMID: 36381959 PMCID: PMC9634142 DOI: 10.4110/in.2022.22.e39] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/18/2022] [Accepted: 08/30/2022] [Indexed: 01/25/2023] Open
Abstract
RNA metabolism plays a central role in regulating of T cell-mediated immunity. RNA processing, modifications, and regulations of RNA decay influence the tight and rapid regulation of gene expression during T cell phase transition. Thymic selection, quiescence maintenance, activation, differentiation, and effector functions of T cells are dependent on selective RNA modulations. Recent technical improvements have unveiled the complex crosstalk between RNAs and T cells. Moreover, resting T cells contain large amounts of untranslated mRNAs, implying that the regulation of RNA metabolism might be a key step in controlling gene expression. Considering the immunological significance of T cells for disease treatment, an understanding of RNA metabolism in T cells could provide new directions in harnessing T cells for therapeutic implications.
Collapse
Affiliation(s)
- Jin Ouk Choi
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Severance Biomedical Science Institute and Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jeong Hyeon Ham
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Severance Biomedical Science Institute and Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Soo Seok Hwang
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Severance Biomedical Science Institute and Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea.,Chronic Intractable Disease Systems Medicine Research Center, Institute of Genetic Science, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
38
|
Wang C, Kong L, Kim S, Lee S, Oh S, Jo S, Jang I, Kim TD. The Role of IL-7 and IL-7R in Cancer Pathophysiology and Immunotherapy. Int J Mol Sci 2022; 23:ijms231810412. [PMID: 36142322 PMCID: PMC9499417 DOI: 10.3390/ijms231810412] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 12/03/2022] Open
Abstract
Interleukin-7 (IL-7) is a multipotent cytokine that maintains the homeostasis of the immune system. IL-7 plays a vital role in T-cell development, proliferation, and differentiation, as well as in B cell maturation through the activation of the IL-7 receptor (IL-7R). IL-7 is closely associated with tumor development and has been used in cancer clinical research and therapy. In this review, we first summarize the roles of IL-7 and IL-7Rα and their downstream signaling pathways in immunity and cancer. Furthermore, we summarize and discuss the recent advances in the use of IL-7 and IL-7Rα as cancer immunotherapy tools and highlight their potential for therapeutic applications. This review will help in the development of cancer immunotherapy regimens based on IL-7 and IL-7Rα, and will also advance their exploitation as more effective and safe immunotherapy tools.
Collapse
Affiliation(s)
- Chunli Wang
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
| | - Lingzu Kong
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Seokmin Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Sunyoung Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| | - Sechan Oh
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Seona Jo
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Inhwan Jang
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
| | - Tae-Don Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea
- Correspondence:
| |
Collapse
|
39
|
Takada I, Hidano S, Takahashi S, Yanaka K, Ogawa H, Tsuchiya M, Yokoyama A, Sato S, Ochi H, Nakagawa T, Kobayashi T, Nakagawa S, Makishima M. Transcriptional coregulator Ess2 controls survival of post-thymic CD4 + T cells through the Myc and IL-7 signaling pathways. J Biol Chem 2022; 298:102342. [PMID: 35933014 PMCID: PMC9436822 DOI: 10.1016/j.jbc.2022.102342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 07/18/2022] [Accepted: 07/26/2022] [Indexed: 11/21/2022] Open
Abstract
Ess2, also known as Dgcr14, is a transcriptional co-regulator of CD4+ T cells. Ess2 is located in a chromosomal region, the loss of which has been associated with 22q11.2 deletion syndrome (22q11DS), which causes heart defects, skeletal abnormalities, and immunodeficiency. However, the specific association of Ess2 with 22q11DS remains unclear. To elucidate the role of Ess2 in T-cell development, we generated Ess2 floxed (Ess2fl/fl) and CD4+ T cell-specific Ess2 KO (Ess2ΔCD4/ΔCD4) mice using the Cre/loxP system. Interestingly, Ess2ΔCD4/ΔCD4 mice exhibited reduced naïve T-cell numbers in the spleen, while the number of thymocytes (CD4-CD8-, CD4+CD8+, CD4+CD8-, and CD4-CD8+) in the thymus remained unchanged. Furthermore, Ess2ΔCD4/ΔCD4 mice had decreased NKT cells and increased γδT cells in the thymus and spleen. A genome-wide expression analysis using RNA-seq revealed that Ess2 deletion alters the expression of many genes in CD4 single-positive thymocytes, including genes related to the immune system and Myc target genes. In addition, Ess2 enhanced the transcriptional activity of c-Myc. Some genes identified as Ess2 targets in mice show expressional correlation with ESS2 in human immune cells. Moreover, Ess2ΔCD4/ΔCD4 naïve CD4+ T cells did not maintain survival in response to IL-7. Our results suggest that Ess2 plays a critical role in post-thymic T-cell survival through the Myc and IL-7 signaling pathways.
Collapse
Affiliation(s)
- Ichiro Takada
- Division of Biochemistry, Department of Biomedical Sciences, School of Medicine, Nihon University, Itabashi-ku, Tokyo, Japan.
| | - Shinya Hidano
- Department of Infectious Diseases Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Sayuri Takahashi
- Department of Urology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kaori Yanaka
- RNA Biology Laboratory, RIKEN Advanced Research Institute, Wako, Saitama, Japan
| | - Hidesato Ogawa
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Megumi Tsuchiya
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Atsushi Yokoyama
- Department of Molecular Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Shingo Sato
- Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Medical Hospital, Tokyo, Japan
| | - Hiroki Ochi
- Department of Rehabilitation for Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Saitama, Japan
| | - Tohru Nakagawa
- Department of Urology, Teikyo University, Itabashi-ku, Tokyo, Japan
| | - Takashi Kobayashi
- Department of Infectious Diseases Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Shinichi Nakagawa
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Makoto Makishima
- Division of Biochemistry, Department of Biomedical Sciences, School of Medicine, Nihon University, Itabashi-ku, Tokyo, Japan.
| |
Collapse
|
40
|
Naoun AA, Raphael I, Forsthuber TG. Immunoregulation via Cell Density and Quorum Sensing-like Mechanisms: An Underexplored Emerging Field with Potential Translational Implications. Cells 2022; 11:cells11152442. [PMID: 35954285 PMCID: PMC9368058 DOI: 10.3390/cells11152442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
Quorum sensing (QS) was historically described as a mechanism by which bacteria detect and optimize their population density via gene regulation based on dynamic environmental cues. Recently, it was proposed that QS or similar mechanisms may have broader applications across different species and cell types. Indeed, emerging evidence shows that the mammalian immune system can also elicit coordinated responses on a population level to regulate cell density and function, thus suggesting that QS-like mechanisms may also be a beneficial trait of the immune system. In this review, we explore and discuss potential QS-like mechanisms deployed by the immune system to coordinate cellular-level responses, such as T cell responses mediated via the common gamma chain (γc) receptor cytokines and the aryl hydrocarbon receptors (AhRs). We present evidence regarding a novel role of QS as a multifunctional mechanism coordinating CD4+ and CD8+ T cell behavior during steady state and in response to infection, inflammatory diseases, and cancer. Successful clinical therapies such as adoptive cell transfer for cancer treatment may be re-evaluated to harness the effects of the QS mechanism(s) and enhance treatment responsiveness. Moreover, we discuss how signaling threshold perturbations through QS-like mediators may result in disturbances of the complex crosstalk between immune cell populations, undesired T cell responses, and induction of autoimmune pathology. Finally, we discuss the potential therapeutic role of modulating immune-system-related QS as a promising avenue to treat human diseases.
Collapse
Affiliation(s)
- Adrian A. Naoun
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Itay Raphael
- Department of Neurological Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15217, USA
- Correspondence: (I.R.); (T.G.F.)
| | - Thomas G. Forsthuber
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX 78249, USA
- Correspondence: (I.R.); (T.G.F.)
| |
Collapse
|
41
|
Galarza-Muñoz G, Kennedy-Boone D, Schott G, Bradrick SS, Garcia-Blanco MA. Antisense modulation of IL7R splicing to control sIL7R expression in human CD4 + T cells. RNA (NEW YORK, N.Y.) 2022; 28:1058-1073. [PMID: 35613883 PMCID: PMC9297843 DOI: 10.1261/rna.079137.122] [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: 02/22/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
The interleukin 7 receptor (IL7R) is strongly associated with increased risk to develop multiple sclerosis (MS), an autoimmune disease of the central nervous system, and this association is likely driven by up-regulation of the soluble isoform of IL7R (sIL7R). Expression of sIL7R is determined by exclusion of the alternative exon 6 from IL7R transcripts, and our previous work revealed that the MS risk allele of the SNP rs6897932 within this exon enhances the expression of sIL7R by promoting exclusion of exon 6. sIL7R potentiates the activity of IL7, leading to enhanced expansion of T cells and increased disability in the experimental autoimmune encephalomyelitis (EAE) murine model of MS. This role in modulating T cell-driven immunity positions sIL7R as an attractive therapeutic target whose expression could be reduced for treatment of MS or increased for treatment of cancers. In this study, we identified novel antisense oligonucleotides (ASOs) that effectively control the inclusion (anti-sIL7R ASOs) or exclusion (pro-sIL7R ASOs) of this exon in a dose-dependent fashion. These ASOs provided excellent control of exon 6 splicing and sIL7R secretion in human primary CD4+ T cells. Supporting their potential for therapeutic targeting, we showed that lead anti-sIL7R ASOs correct the enhanced exon 6 exclusion imposed by the MS risk allele of rs6897932, whereas lead pro-sIL7R ASOs phenocopy it. The data presented here form the foundation for future preclinical studies that will test the therapeutic potential of these ASOs in MS and immuno-oncology.
Collapse
Affiliation(s)
- Gaddiel Galarza-Muñoz
- Autoimmunity BioSolutions, Galveston, Texas 77550, USA
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77550, USA
| | - Debbie Kennedy-Boone
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
| | - Geraldine Schott
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
| | - Shelton S Bradrick
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas 77550, USA
| | - Mariano A Garcia-Blanco
- Autoimmunity BioSolutions, Galveston, Texas 77550, USA
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas 77550, USA
| |
Collapse
|
42
|
Lagou MK, Anastasiadou DP, Karagiannis GS. A Proposed Link Between Acute Thymic Involution and Late Adverse Effects of Chemotherapy. Front Immunol 2022; 13:933547. [PMID: 35844592 PMCID: PMC9283860 DOI: 10.3389/fimmu.2022.933547] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Epidemiologic data suggest that cancer survivors tend to develop a protuberant number of adverse late effects, including second primary malignancies (SPM), as a result of cytotoxic chemotherapy. Besides the genotoxic potential of these drugs that directly inflict mutational burden on genomic DNA, the precise mechanisms contributing to SPM development are poorly understood. Cancer is nowadays perceived as a complex process that goes beyond the concept of genetic disease and includes tumor cell interactions with complex stromal and immune cell microenvironments. The cancer immunoediting theory offers an explanation for the development of nascent neoplastic cells. Briefly, the theory suggests that newly emerging tumor cells are mostly eliminated by an effective tissue immunosurveillance, but certain tumor variants may occasionally escape innate and adaptive mechanisms of immunological destruction, entering an equilibrium phase, where immunologic tumor cell death "equals" new tumor cell birth. Subsequent microenvironmental pressures and accumulation of helpful mutations in certain variants may lead to escape from the equilibrium phase, and eventually cause an overt neoplasm. Cancer immunoediting functions as a dedicated sentinel under the auspice of a highly competent immune system. This perspective offers the fresh insight that chemotherapy-induced thymic involution, which is characterized by the extensive obliteration of the sensitive thymic epithelial cell (TEC) compartment, can cause long-term defects in thymopoiesis and in establishment of diverse T cell receptor repertoires and peripheral T cell pools of cancer survivors. Such delayed recovery of T cell adaptive immunity may result in prolonged hijacking of the cancer immunoediting mechanisms, and lead to development of persistent and mortal infections, inflammatory disorders, organ-specific autoimmunity lesions, and SPMs. Acknowledging that chemotherapy-induced thymic involution is a potential risk factor for the emergence of SPM demarcates new avenues for the rationalized development of pharmacologic interventions to promote thymic regeneration in patients receiving cytoreductive chemotherapies.
Collapse
Affiliation(s)
- Maria K. Lagou
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Tumor Microenvironment and Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, United States
| | - Dimitra P. Anastasiadou
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Tumor Microenvironment and Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, United States
| | - George S. Karagiannis
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Tumor Microenvironment and Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, United States
- Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein Cancer Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
43
|
Kiapour N, Wu B, Wang Y, Seyedsadr M, Kapoor S, Zhang X, Elzoheiry M, Kasimoglu E, Wan Y, Markovic-Plese S. Therapeutic Effect of Anti-CD52 Monoclonal Antibody in Multiple Sclerosis and Its Animal Models Is Mediated via T Regulatory Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:49-56. [PMID: 35750335 PMCID: PMC9458467 DOI: 10.4049/jimmunol.2100176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/27/2022] [Indexed: 06/15/2023]
Abstract
The objective of this study is to determine the mechanism of action of anti-CD52 mAb treatment in patients with relapsing-remitting multiple sclerosis (RRMS). Experimental autoimmune encephalomyelitis (EAE), an animal model of the disease, was used to address the role of T regulatory cells (Tregs) in the anti-CD52 mAb-induced suppression of the disease. In vitro studies on PBMCs from RRMS patients and matched healthy controls determined the effect of IL-7 on the expansion of CD4+CD25+CD127- Tregs and induction of their suppressive phenotype. This study using EAE animal models of MS has shown that mouse anti-CD52 mAb suppression of clinical disease was augmented by coadministration of IL-7 and partially reversed by anti-IL-7 mAb. In vitro human studies showed that IL-7 induced expansion of CD4+CD25+CD127- Tregs and increased their FOXP3, GITIR, CD46, CTLA-4, granzyme B, and perforin expression. Anti-CD52 mAb treatment of mice with relapsing-remitting EAE induced expansion of Foxp3+CD4+ Tregs and the suppression of IL-17A+CD4+ and IFN-γ+CD4+ cells in peripheral immune organs and CNS infiltrates. The effect was detected immediately after the treatment and maintained over long-term follow-up. Foxp3+CD4+ Treg-mediated suppression of IL-17A+CD4+ and IFN-γ+CD4+ cells in the spinal cord infiltrates was reversed after inducible Foxp3 depletion. Our results demonstrated that the therapeutic effect of U.S. Food and Drug Administration-approved anti-CD52 mAb is dependent on the presence of Tregs.
Collapse
Affiliation(s)
- Nazanin Kiapour
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Bing Wu
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Frontier Science Center for Immunology and Metabolism of Medical Research Institute, Wuhan University, Wuhan, China
| | - Yan Wang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA; and
| | | | - Sahil Kapoor
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Xin Zhang
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Duke Molecular Physiology Institute, Department of Orthopedic Surgery, Duke University, Durham, NC
| | - Manal Elzoheiry
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA; and
| | - Ezgi Kasimoglu
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA; and
| | - Yisong Wan
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Silva Markovic-Plese
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, NC;
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA; and
| |
Collapse
|
44
|
Sun X, Nguyen T, Achour A, Ko A, Cifello J, Ling C, Sharma J, Hiroi T, Zhang Y, Chia CW, Wood Iii W, Wu WW, Zukley L, Phue JN, Becker KG, Shen RF, Ferrucci L, Weng NP. Longitudinal analysis reveals age-related changes in the T cell receptor repertoire of human T cell subsets. J Clin Invest 2022; 132:158122. [PMID: 35708913 PMCID: PMC9433102 DOI: 10.1172/jci158122] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
A diverse T cell receptor (TCR) repertoire is essential for protection against a variety of pathogens, and TCR repertoire size is believed to decline with age. However, the precise size of human TCR repertoires, in both total and subsets of T cells, as well as their changes with age, are not fully characterized. We conducted a longitudinal analysis of the human blood TCRα and TCRβ repertoire of CD4+ and CD8+ T cell subsets using a unique molecular identifier–based (UMI-based) RNA-seq method. Thorough analysis of 1.9 × 108 T cells yielded the lower estimate of TCR repertoire richness in an adult at 3.8 × 108. Alterations of the TCR repertoire with age were observed in all 4 subsets of T cells. The greatest reduction was observed in naive CD8+ T cells, while the greatest clonal expansion was in memory CD8+ T cells, and the highest increased retention of TCR sequences was in memory CD8+ T cells. Our results demonstrated that age-related TCR repertoire attrition is subset specific and more profound for CD8+ than CD4+ T cells, suggesting that aging has a more profound effect on cytotoxic as opposed to helper T cell functions. This may explain the increased susceptibility of older adults to novel infections.
Collapse
Affiliation(s)
- Xiaoping Sun
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Thomas Nguyen
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Achouak Achour
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Annette Ko
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Jeffrey Cifello
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Chen Ling
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Jay Sharma
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Toyoko Hiroi
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| | - Yongqing Zhang
- Gene expression and Genomics Unit, Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, United States of America
| | - Chee W Chia
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, United States of America
| | - William Wood Iii
- Gene expression and Genomics Unit, Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, United States of America
| | - Wells W Wu
- Facility for Biotechnology Resources, Food and Drug Administration, Silver Spring, United States of America
| | - Linda Zukley
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, United States of America
| | - Je-Nie Phue
- Facility for Biotechnology Resources, Food and Drug Administration, Silver Spring, United States of America
| | - Kevin G Becker
- Gene expression and Genomics Unit, Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, United States of America
| | - Rong-Fong Shen
- Facility for Biotechnology Resources, Food and Drug Administration, Silver Spring, United States of America
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, United States of America
| | - Nan-Ping Weng
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, NIH, Baltimore, United States of America
| |
Collapse
|
45
|
The generation and application of antigen-specific T cell therapies for cancer and viral-associated disease. Mol Ther 2022; 30:2130-2152. [PMID: 35149193 PMCID: PMC9171249 DOI: 10.1016/j.ymthe.2022.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/27/2021] [Accepted: 02/03/2022] [Indexed: 11/23/2022] Open
Abstract
Immunotherapy with antigen-specific T cells is a promising, targeted therapeutic option for patients with cancer as well as for immunocompromised patients with virus infections. In this review, we characterize and compare current manufacturing protocols for the generation of T cells specific to viral and non-viral tumor-associated antigens. Specifically, we discuss: (1) the different methodologies to expand virus-specific T cell and non-viral tumor-associated antigen-specific T cell products, (2) an overview of the immunological principles involved when developing such manufacturing protocols, and (3) proposed standardized methodologies for the generation of polyclonal, polyfunctional antigen-specific T cells irrespective of donor source. Ex vivo expanded cells have been safely administered to treat numerous patients with virus-associated malignancies, hematologic malignancies, and solid tumors. Hence, we have performed a comprehensive review of the clinical trial results evaluating the safety, feasibility, and efficacy of these products in the clinic. In summary, this review seeks to provide new insights regarding antigen-specific T cell technology to benefit a rapidly expanding T cell therapy field.
Collapse
|
46
|
Tsuchida T, Wada T, Mizugaki A, Oda Y, Kayano K, Yamakawa K, Tanaka S. Protocol for a Sepsis Model Utilizing Fecal Suspension in Mice: Fecal Suspension Intraperitoneal Injection Model. Front Med (Lausanne) 2022; 9:765805. [PMID: 35646946 PMCID: PMC9134078 DOI: 10.3389/fmed.2022.765805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
Background Various animal models of sepsis have been developed to optimize sepsis treatment. However, therapeutic agents that were successful in animal models were rarely effective in human clinical trials. The cecal ligation and puncture (CLP) model is currently the gold standard for sepsis studies. However, its limitations include the high variability among researchers and the difficulty in comparing animals with different cecum shapes and sizes. In this study, we established a protocol for the creation of a simple and accessible sepsis rodent model using fecal suspensions that minimized differences in technical effects among researchers and individual differences in animals. Methods A mouse model of sepsis using fecal suspension intraperitoneal injection (FSI) was created using fresh stool excreted within 24 h. The collected fresh stool was dissolved in saline solution and filtered. The obtained fecal suspension was injected intraperitoneally into the mice. Moreover, fecal suspensions with different concentrations were prepared, and the survival rates were compared among the fecal suspensions for each concentration. To assess the validity of the FSI as a sepsis model, CLP and FSI with similar mortality rates were compared pathologically, physiologically, immunologically, and bacteriologically. Histopathological comparison was evaluated by hematoxylin-eosin and Gram staining of the parenchymal organs. Physiological evaluation was performed by comparing the respiratory rate, body temperature, and blood gas analysis results. Immunological assessment was performed using multiplex analysis. Bacteriological comparisons were performed by culturing ascites fluid. Results The FSI model increased mortality in proportion to the fecal suspension concentration. The mortality rate was reduced with antibiotic administration. In various comparative experiments conducted using the FSI and CLP models, both models showed findings consistent with sepsis. Furthermore, the FSI model showed less variability among the individuals in each test. Conclusion This is the first detailed and accurate report of a protocol for creating a sepsis model using fecal suspension. The FSI model is a minimally invasive and accessible sepsis rodent model. Its clinical validity as a sepsis model was proven via histological, physiological, microbiological, and immunological evaluation methods. The FSI model minimizes individual differences between mice and helps to conduct accurate studies after the onset of sepsis.
Collapse
Affiliation(s)
- Takumi Tsuchida
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Takeshi Wada
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- *Correspondence: Takeshi Wada,
| | - Asumi Mizugaki
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshitaka Oda
- Department of Cancer Pathology, Faculty of Medicine, WPI-ICReDD, Hokkaido University, Sapporo, Japan
| | - Katsuhide Kayano
- Department of Emergency Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Kazuma Yamakawa
- Department of Emergency Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, WPI-ICReDD, Hokkaido University, Sapporo, Japan
| |
Collapse
|
47
|
Wei F, Cheng XX, Xue JZ, Xue SA. Emerging Strategies in TCR-Engineered T Cells. Front Immunol 2022; 13:850358. [PMID: 35432319 PMCID: PMC9006933 DOI: 10.3389/fimmu.2022.850358] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy of cancer has made tremendous progress in recent years, as demonstrated by the remarkable clinical responses obtained from adoptive cell transfer (ACT) of patient-derived tumor infiltrating lymphocytes, chimeric antigen receptor (CAR)-modified T cells (CAR-T) and T cell receptor (TCR)-engineered T cells (TCR-T). TCR-T uses specific TCRS optimized for tumor engagement and can recognize epitopes derived from both cell-surface and intracellular targets, including tumor-associated antigens, cancer germline antigens, viral oncoproteins, and tumor-specific neoantigens (neoAgs) that are largely sequestered in the cytoplasm and nucleus of tumor cells. Moreover, as TCRS are naturally developed for sensitive antigen detection, they are able to recognize epitopes at far lower concentrations than required for CAR-T activation. Therefore, TCR-T holds great promise for the treatment of human cancers. In this focused review, we summarize basic, translational, and clinical insights into the challenges and opportunities of TCR-T. We review emerging strategies used in current ACT, point out limitations, and propose possible solutions. We highlight the importance of targeting tumor-specific neoAgs and outline a strategy of combining neoAg vaccines, checkpoint blockade therapy, and adoptive transfer of neoAg-specific TCR-T to produce a truly tumor-specific therapy, which is able to penetrate into solid tumors and resist the immunosuppressive tumor microenvironment. We believe such a combination approach should lead to a significant improvement in cancer immunotherapies, especially for solid tumors, and may provide a general strategy for the eradication of multiple cancers.
Collapse
Affiliation(s)
- Fang Wei
- Genetic Engineering Laboratory, School of Biological & Environmental Engineering, Xi'An University, Xi'An, China
| | - Xiao-Xia Cheng
- Genetic Engineering Laboratory, School of Biological & Environmental Engineering, Xi'An University, Xi'An, China
| | - John Zhao Xue
- Genetic Engineering Laboratory, School of Biological & Environmental Engineering, Xi'An University, Xi'An, China
| | - Shao-An Xue
- Genetic Engineering Laboratory, School of Biological & Environmental Engineering, Xi'An University, Xi'An, China
| |
Collapse
|
48
|
NFAT Factors Are Dispensable for the Development but Are Critical for the Maintenance of Foxp3+ Regulatory T Cells. Cells 2022; 11:cells11091397. [PMID: 35563702 PMCID: PMC9104130 DOI: 10.3390/cells11091397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 11/16/2022] Open
Abstract
The transcription factors of the nuclear factor of activated T cell (NFAT) family play a crucial role in multiple aspects of T cell function. It has recently been reported that NFATs play an important role in the suppressive function of CD4+CD25+Foxp3+ regulatory T (Treg) cells. In this study, we have investigated the role of NFATs in the thymic development of Treg cells in mice. We show that NFAT factors are dispensable for the development of Foxp3+ Treg cells in the thymus but are critical for the maintenance of both the phenotype and survival of Treg cells in the thymus as well as in peripheral lymphoid organs. Specifically, the homeostasis of CD4+CD25+Foxp3+ but not the CD4+CD25−Foxp3+ fraction is severely perturbed when NFAT signaling is blocked, leading to a strongly reduced Treg population. We underscored this intriguing effect of NFAT on CD4+CD25+Foxp3+ Treg cells to the disruption of survival signals provided by interleukin 2 (IL-2). Accordingly, blocking Treg cell death by abolishing the activity of pro-apoptotic Bcl-2 family member Bim, compensated for the survival defects induced due to a lack of NFAT-IL-2-IL-2R signaling. Inhibition of NFAT activity led to a strong reduction in the number of Foxp3+ Treg cells; however, it did not influence the level of Foxp3 expression on an individual cell basis. In addition, we show a differential effect of IL-2 and IL-7 signaling on Foxp3+ Treg versus CD4+CD25− T cell development, again underlining the dispensability of NFAT signaling in the development, but not in the maintenance of Foxp3+ Treg cells.
Collapse
|
49
|
Ahmad R, Haque M. Surviving the Storm: Cytokine Biosignature in SARS-CoV-2 Severity Prediction. Vaccines (Basel) 2022; 10:vaccines10040614. [PMID: 35455363 PMCID: PMC9026643 DOI: 10.3390/vaccines10040614] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The world has been stricken mentally, physically, and economically by the COVID-19 virus. However, while SARS-CoV-2 viral infection results in mild flu-like symptoms in most patients, a number of those infected develop severe illness. These patients require hospitalization and intensive care. The severe disease can spiral downwards with eventual severe damage to the lungs and failure of multiple organs, leading to the individual’s demise. It is necessary to identify those who are developing a severe form of illness to provide early management. Therefore, it is crucial to learn about the mechanisms and chemical mediators that lead to critical conditions in SARS-CoV-2 infection. This paper reviews studies regarding the individual chemical mediators, pathways, and means that contribute to worsening health conditions in SARS-CoV-2 infection. Abstract A significant part of the world population has been affected by the devastating SARS-CoV-2 infection. It has deleterious effects on mental and physical health and global economic conditions. Evidence suggests that the pathogenesis of SARS-CoV-2 infection may result in immunopathology such as neutrophilia, lymphopenia, decreased response of type I interferon, monocyte, and macrophage dysregulation. Even though most individuals infected with the SARS-CoV-2 virus suffer mild symptoms similar to flu, severe illness develops in some cases, including dysfunction of multiple organs. Excessive production of different inflammatory cytokines leads to a cytokine storm in COVID-19 infection. The large quantities of inflammatory cytokines trigger several inflammation pathways through tissue cell and immune cell receptors. Such mechanisms eventually lead to complications such as acute respiratory distress syndrome, intravascular coagulation, capillary leak syndrome, failure of multiple organs, and, in severe cases, death. Thus, to devise an effective management plan for SARS-CoV-2 infection, it is necessary to comprehend the start and pathways of signaling for the SARS-CoV-2 infection-induced cytokine storm. This article discusses the current findings of SARS-CoV-2 related to immunopathology, the different paths of signaling and other cytokines that result in a cytokine storm, and biomarkers that can act as early signs of warning for severe illness. A detailed understanding of the cytokine storm may aid in the development of effective means for controlling the disease’s immunopathology. In addition, noting the biomarkers and pathophysiology of severe SARS-CoV-2 infection as early warning signs can help prevent severe complications.
Collapse
Affiliation(s)
- Rahnuma Ahmad
- Department of Physiology, Medical College for Women and Hospital, Plot No 4 Road 8/9, Sector-1, Dhaka 1230, Bangladesh;
| | - Mainul Haque
- Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kem Perdana Sungai Besi, Kuala Lumpur 57000, Malaysia
- Correspondence: or
| |
Collapse
|
50
|
Papadopetraki A, Maridaki M, Zagouri F, Dimopoulos MA, Koutsilieris M, Philippou A. Physical Exercise Restrains Cancer Progression through Muscle-Derived Factors. Cancers (Basel) 2022; 14:cancers14081892. [PMID: 35454797 PMCID: PMC9024747 DOI: 10.3390/cancers14081892] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/02/2022] [Accepted: 04/07/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The benefits of physical exercise against cancer onset and progression, as well as the adverse effects of physical inactivity have changed the way that we utilize exercise for cancer patients. Nevertheless, although guidelines of various scientific societies and organizations propose exercise as a complementary intervention during cancer therapies, the exact cellular and molecular mechanisms by which exercise acts against cancer have not yet been elucidated. In the present review, we analyze the factors which either are secreted from skeletal muscle or are regulated by exercise and can restrain cancer evolution. We also describe the exercise-induced factors that counteract severe side effects of cancer treatment, as well as the ways that muscle-derived factors are delivered to the target cells. Abstract A growing body of in vitro and in vivo studies suggests that physical activity offers important benefits against cancer, in terms of both prevention and treatment. However, the exact mechanisms implicated in the anticancer effects of exercise remain to be further elucidated. Muscle-secreted factors in response to contraction have been proposed to mediate the physical exercise-induced beneficial effects and be responsible for the inter-tissue communications. Specifically, myokines and microRNAs (miRNAs) constitute the most studied components of the skeletal muscle secretome that appear to affect the malignancy, either directly by possessing antioncogenic properties, or indirectly by mobilizing the antitumor immune responses. Moreover, some of these factors are capable of mitigating serious, disease-associated adverse effects that deteriorate patients’ quality of life and prognosis. The present review summarizes the myokines and miRNAs that may have potent anticancer properties and the expression of which is induced by physical exercise, while the mechanisms of secretion and intercellular transportation of these factors are also discussed.
Collapse
Affiliation(s)
- Argyro Papadopetraki
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.P.); (M.K.)
| | - Maria Maridaki
- Faculty of Physical Education and Sport Science, National and Kapodistrian University of Athens, 17237 Dafne, Greece;
| | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece; (F.Z.); (M.-A.D.)
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece; (F.Z.); (M.-A.D.)
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.P.); (M.K.)
| | - Anastassios Philippou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.P.); (M.K.)
- Correspondence: ; Tel./Fax: +30-210-7462690
| |
Collapse
|