1
|
Luo S, Notaro A, Lin L. ATLAS-seq: a microfluidic single-cell TCR screen for antigen-reactive TCRs. Nat Commun 2025; 16:216. [PMID: 39746936 PMCID: PMC11696065 DOI: 10.1038/s41467-024-54675-3] [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: 09/26/2023] [Accepted: 11/18/2024] [Indexed: 01/04/2025] Open
Abstract
Discovering antigen-reactive T cell receptors (TCRs) is central to developing effective engineered T cell immunotherapies. However, the conventional technologies for isolating antigen-reactive TCRs (i.e., major histocompatibility complex (MHC) multimer staining) focus on high-affinity interactions between the TCR and MHC-antigen complex, and may fail to identify TCRs with high efficacy for activating T cells. Here, we develop a microfluidic single-cell screening method for antigen-reactive T cells named ATLAS-seq (Aptamer-based T Lymphocyte Activity Screening and SEQuencing). This technology isolates and characterizes activated T cells via an aptamer-based fluorescent molecular sensor, which monitors the cytotoxic cytokine IFNγ secretion from single T cells upon antigen stimulation, followed by single-cell RNA and single-cell TCR sequencing. We use ATLAS-seq to screen TCRs reactive to cytomegalovirus (CMV) or prostate specific antigen (PSA) from peripheral blood mononuclear cells (PBMCs). ATLAS-seq identifies distinct TCR clonotype populations with higher T cell activation levels compared to TCRs recovered by MHC multimer staining. Select TCR clonotypes from ATLAS-seq are more efficient in target cell killing than those from MHC multimer staining. Collectively, ATLAS-seq provides an efficient and broadly applicable technology to screen antigen-reactive TCRs for engineered T cell immunotherapy.
Collapse
Affiliation(s)
- Siwei Luo
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Computational and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amber Notaro
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Computational and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lan Lin
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Center for Computational and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Ishibashi A, Li Y, Hisatomi Y, Ohta N, Uegaki Y, Tanemura A, Ohashi R, Kitamura K, Saga K, Yoshimura Y, Inubushi S, Ishida K, Iwabuchi S, Hashimoto S, Kiyohara E, Yagita H, Kaneda Y, Nimura K. Local treatment of HVJ-E with T cell costimulatory molecule stimulation elicits systemic anti-tumor effects. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200893. [PMID: 39534532 PMCID: PMC11555341 DOI: 10.1016/j.omton.2024.200893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
The tumor-infiltrating lymphocyte (TIL) is a crucial factor in controlling tumor growth. A therapeutic method activating TIL is desired for treating patients with metastatic tumors. Here, we show that treating a local tumor with a combination therapy of UV-irradiated hemagglutinating virus of Japan envelope (HVJ-E) plus agonist antibodies, including OX40, against T cell costimulatory molecules induces systemic anti-tumor effects in a T cell-dependent manner in multiple cancer cell lines. Transcriptome and T cell receptor repertoire analyses revealed that HVJ-E + anti-OX40 antibody treatment activates CD4 and CD8 T cells and promotes T cell trafficking between tumors. These systemic anti-tumor effects required an association between Nkg2d and Nkg2d ligands. Our findings provide insights into how systemic anti-tumor effects are induced and may help the development of therapeutic strategies for eliciting such effects.
Collapse
Affiliation(s)
- Airi Ishibashi
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yue Li
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
- Division of Gene Therapy Science, Gunma University Initiative for Advanced Research, Gunma University, Maebashi, Gunma 371-8511, Japan
| | - Yuuta Hisatomi
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Noriko Ohta
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
- Division of Gene Therapy Science, Gunma University Initiative for Advanced Research, Gunma University, Maebashi, Gunma 371-8511, Japan
| | - Yuko Uegaki
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Riuko Ohashi
- Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
- Histopathology Core Facility, Center for Research Promotion, Niigata University School of Medicine, Niigata 951-8510, Japan
| | - Koji Kitamura
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
- Department of Otorhinolaryngology-Head and Neck Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Kotaro Saga
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yasuhide Yoshimura
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Satoko Inubushi
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Kyoso Ishida
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Sadahiro Iwabuchi
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Shinichi Hashimoto
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Eiji Kiyohara
- Department of Dermatology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Yasufumi Kaneda
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Keisuke Nimura
- Division of Gene Therapy Science, Department of Genome Biology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
- Division of Gene Therapy Science, Gunma University Initiative for Advanced Research, Gunma University, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
3
|
Vegesana K, Thomas PG. Cracking the code of adaptive immunity: The role of computational tools. Cell Syst 2024; 15:1156-1167. [PMID: 39701033 DOI: 10.1016/j.cels.2024.11.009] [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: 04/15/2024] [Revised: 06/14/2024] [Accepted: 11/14/2024] [Indexed: 12/21/2024]
Abstract
In recent years, the advances in high-throughput and deep sequencing have generated a diverse amount of adaptive immune repertoire data. This surge in data has seen a proportional increase in computational methods aimed to characterize T cell receptor (TCR) repertoires. In this perspective, we will provide a brief commentary on the various domains of TCR repertoire analysis, their respective computational methods, and the ongoing challenges. Given the breadth of methods and applications of TCR analysis, we will focus our perspective on sequence-based computational methods.
Collapse
Affiliation(s)
- Kasi Vegesana
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul G Thomas
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
4
|
Mistri SK, Hilton BM, Horrigan KJ, Andretta ES, Savard R, Dienz O, Hampel KJ, Gerrard DL, Rose JT, Sidiropoulos N, Majumdar D, Boyson JE. SLAM/SAP signaling regulates discrete γδ T cell developmental checkpoints and shapes the innate-like γδ TCR repertoire. eLife 2024; 13:RP97229. [PMID: 39656519 PMCID: PMC11630817 DOI: 10.7554/elife.97229] [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] [Indexed: 12/12/2024] Open
Abstract
During thymic development, most γδ T cells acquire innate-like characteristics that are critical for their function in tumor surveillance, infectious disease, and tissue repair. The mechanisms, however, that regulate γδ T cell developmental programming remain unclear. Recently, we demonstrated that the SLAM/SAP signaling pathway regulates the development and function of multiple innate-like γδ T cell subsets. Here, we used a single-cell proteogenomics approach to identify SAP-dependent developmental checkpoints and to define the SAP-dependent γδ TCR repertoire in mice. SAP deficiency resulted in both a significant loss of an immature Gzma+Blk+Etv5+Tox2+ γδT17 precursor population and a significant increase in Cd4+Cd8+Rorc+Ptcra+Rag1+ thymic γδ T cells. SAP-dependent diversion of embryonic day 17 thymic γδ T cell clonotypes into the αβ T cell developmental pathway was associated with a decreased frequency of mature clonotypes in neonatal thymus, and an altered γδ TCR repertoire in the periphery. Finally, we identify TRGV4/TRAV13-4(DV7)-expressing T cells as a novel, SAP-dependent Vγ4 γδT1 subset. Together, the data support a model in which SAP-dependent γδ/αβ T cell lineage commitment regulates γδ T cell developmental programming and shapes the γδ TCR repertoire.
Collapse
MESH Headings
- Animals
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Mice
- Signal Transduction
- Signaling Lymphocytic Activation Molecule Associated Protein/metabolism
- Signaling Lymphocytic Activation Molecule Associated Protein/genetics
- Immunity, Innate
- Mice, Inbred C57BL
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Cell Differentiation
- Intraepithelial Lymphocytes/immunology
- Intraepithelial Lymphocytes/metabolism
- Signaling Lymphocytic Activation Molecule Family
Collapse
Affiliation(s)
- Somen K Mistri
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Brianna M Hilton
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Katherine J Horrigan
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Emma S Andretta
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Remi Savard
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Oliver Dienz
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Kenneth J Hampel
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical CenterBurlingtonUnited States
| | - Diana L Gerrard
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical CenterBurlingtonUnited States
| | - Joshua T Rose
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical CenterBurlingtonUnited States
| | - Nikoletta Sidiropoulos
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical CenterBurlingtonUnited States
| | - Dev Majumdar
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Jonathan E Boyson
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| |
Collapse
|
5
|
Weng Z, Mai Z, Yuan J, Liu Q, Deng F, Yang H, Ling Y, Xie X, Lin X, Lin T, Chen J, Wei X, Luo K, Fu J, Wen J. Evolution of genome and immunogenome in esophageal squamous cell carcinomas driven by neoadjuvant chemoradiotherapy. Int J Cancer 2024; 155:2021-2035. [PMID: 39081132 DOI: 10.1002/ijc.35118] [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: 03/25/2024] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 10/04/2024]
Abstract
Neoadjuvant chemoradiotherapy (NCRT) followed by surgery is a standard treatment for locally advanced esophageal squamous cell carcinomas (ESCCs). However, the evolution of genome and immunogenome in ESCCs driven by NCRT remains incompletely elucidated. We performed whole-exome sequencing of 51 ESCC tumors collected before and after NCRT, 36 of which were subjected to transcriptome sequencing. Clonal analysis identified clonal extinction in 13 ESCC patients wherein all pre-NCRT clones disappeared after NCRT, and clonal persistence in 9 patients wherein clones endured following NCRT. The clone-persistent patients showed higher pre-NCRT genomic intratumoral heterogeneity and worse prognosis than the clone-extinct ones. In contrast to the clone-extinct patients, the clone-persistent patients demonstrated a high proportion of subclonal neoantigens within pre-treatment specimens. Transcriptome analysis revealed increased immune infiltrations and up-regulated immune-related pathways after NCRT, especially in the clone-extinct patients. The number of T cell receptor-neoantigen interactions was higher in the clone-extinct patients than in the clone-persistent ones. The decrease in T cell repertoire evenness positively correlated to the decreased number of clonal neoantigens after NCRT, especially in the clone-extinct patients. In conclusion, we identified two prognosis-related clonal dynamic modes driven by NCRT in ESCCs. This study extended our knowledge of the ESCC genome and immunogenome evolutions driven by NCRT.
Collapse
Affiliation(s)
- Zelin Weng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zihang Mai
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianye Yuan
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Qianwen Liu
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fangqi Deng
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hong Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yihong Ling
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiuying Xie
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaodan Lin
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ting Lin
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiyang Chen
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaoli Wei
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kongjia Luo
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianhua Fu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing Wen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
6
|
Hui L, Wu F, Xu Y, Yang G, Luo Q, Li Y, Ma L, Yao X, Li J. The T-cell receptor β chain CDR3 insights of bovine liver immune repertoire under heat stress. Anim Biosci 2024; 37:2178-2188. [PMID: 38938039 PMCID: PMC11541024 DOI: 10.5713/ab.24.0152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/08/2024] [Accepted: 05/26/2024] [Indexed: 06/29/2024] Open
Abstract
OBJECTIVE The liver plays a dual role in regulating temperature and immune responses. Examining the influence of heat stress (HS) on liver T cells contributes significantly to understanding the intricate interplay between the immune system and hepatic tissues under thermal stress. This study focused on investigating the characteristics of the T-cell receptor (TCR) β chain CDR3 repertoire in bovine liver samples under both HS and pairfed (PF) environmental conditions. METHODS Sequencing data from six samples sourced from the GEO database underwent annotation. Utilizing immunarch and VDJtool software, the study conducted comprehensive analyses encompassing basic evaluation, clonality assessment, immune repertoire comparison, diversity estimation, gene usage profiling, VJ gene segment pairing scrutiny, clonal tracking, and Kmers analysis. RESULTS All four TCR chains, namely α, β, γ, and δ, were detected, with the α chains exhibiting the highest detection frequency, followed closely by the β chains. The prevalence of αβ TCRs in bovine liver samples underscored their crucial role in governing hepatic tissue's physiological functions. The TCR β CDR3 repertoire showcased substantial inter-individual variability, featuring diverse clonotypes exhibiting distinct amino acid lengths. Intriguingly, HS cattle displayed heightened diversity and clonality, suggesting potential peripheral T cell migration into the liver under environmental conditions. Notably, differential VJ gene pairings were observed in HS cattle compared to the PF, despite individual variations in V and J gene utilization. Additionally, while most high-frequency amino acid 5-mers remained consistent between the HS and PF, GELHF, and YDYHF were notably prevalent in the HS group. Across all samples, a prevalent trend of high-frequency 5mers skewed towards polar and hydrophobic amino acids was evident. CONCLUSION This study elucidates the characteristics of liver TCR β chain CDR3 repertoire under HS conditions, enhancing our understanding of HS implications.
Collapse
Affiliation(s)
- Linhu Hui
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563000,
China
| | - Fengli Wu
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563000,
China
| | - Yuanyuan Xu
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563000,
China
| | - Guangjun Yang
- Xiangyun County Livestock Workstation, Xiangyun 671000,
China
| | - Qiaorong Luo
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201,
China
| | - Yangyang Li
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563000,
China
| | - Long Ma
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563000,
China
| | - Xinsheng Yao
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563000,
China
| | - Jun Li
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi 563000,
China
| |
Collapse
|
7
|
Mahdy AKH, Lokes E, Schöpfel V, Kriukova V, Britanova OV, Steiert TA, Franke A, ElAbd H. Bulk T cell repertoire sequencing (TCR-Seq) is a powerful technology for understanding inflammation-mediated diseases. J Autoimmun 2024; 149:103337. [PMID: 39571301 DOI: 10.1016/j.jaut.2024.103337] [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: 04/19/2024] [Revised: 10/12/2024] [Accepted: 11/09/2024] [Indexed: 12/15/2024]
Abstract
Multiple alterations in the T cell repertoire were identified in many chronic inflammatory diseases such as inflammatory bowel disease, multiple sclerosis, and rheumatoid arthritis, suggesting that T cells might, directly or indirectly, be implicated in these pathologies. This has sparked a deep interest in characterizing disease-associated T cell clonotypes as well as in identifying and quantifying their contribution to the pathophysiology of different autoimmune and inflammation-mediated diseases. Bulk T cell repertoire sequencing (TCR-Seq) has emerged as a powerful method to profile the T cell repertoire of a sample in a high throughput fashion. Given the increasing utilization of TCR-Seq, we aimed here to provide a comprehensive, up-to-date review of the method, its extensions, and its ability to investigate chronic and autoimmune diseases. Specifically, we started by introducing the immunological basis of TCR repertoire generation and features, followed by discussing different experimental approach to perform TCR-Seq, then we describe different methods and frameworks for analyzing the generated datasets. Subsequently, different experimental techniques for investigating the antigenicity of T cell clonotypes are described. Lastly, we discuss recent studies that utilized TCR-Seq to understand different inflammation-mediated diseases, discuss fallbacks of the technology and potential future directions to overcome current limitations.
Collapse
Affiliation(s)
- Aya K H Mahdy
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany
| | - Evgeniya Lokes
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany
| | - Valentina Schöpfel
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany
| | - Valeriia Kriukova
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany
| | - Olga V Britanova
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany
| | - Tim A Steiert
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany.
| | - Hesham ElAbd
- Institute of Clinical Molecular Biology, Kiel University & University Medical Centre Schleswig-Holstein, Kiel, 24105, Germany.
| |
Collapse
|
8
|
Lutz R, Grünschläger F, Simon M, Awwad MHS, Bauer M, Yousefian S, Beumer N, Jopp-Saile L, Sedlmeier A, Solé-Boldo L, Avanesyan B, Vonficht D, Stelmach P, Steinbuss G, Boch T, Steiger S, Baertsch MA, Prokoph N, Rippe K, Durie BGM, Wickenhauser C, Trumpp A, Müller-Tidow C, Hübschmann D, Weinhold N, Raab MS, Brors B, Goldschmidt H, Imbusch CD, Hundemer M, Haas S. Multiple myeloma long-term survivors exhibit sustained immune alterations decades after first-line therapy. Nat Commun 2024; 15:10396. [PMID: 39613747 DOI: 10.1038/s41467-024-54543-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/14/2024] [Indexed: 12/01/2024] Open
Abstract
The long-term consequences of cancer and its therapy on the patients' immune system years after cancer-free survival remain poorly understood. Here, we present an in-depth characterization of the bone marrow immune ecosystem of multiple myeloma long-term survivors, from initial diagnosis up to 17 years following a single therapy line and cancer-free survival. Using comparative single-cell analyses combined with molecular, genomic, and functional approaches, we demonstrate that multiple myeloma long-term survivors exhibit pronounced alterations in their bone marrow microenvironment associated with impaired immunity. These immunological alterations were frequently linked to an inflammatory immune circuit fueled by the long-term persistence or resurgence of residual myeloma cells. Notably, even in the complete absence of any detectable residual disease for decades, sustained changes in the immune system were observed, suggesting an irreversible 'immunological scarring' caused by the initial exposure to the cancer and therapy. Collectively, our study provides key insights into the molecular and cellular bone marrow ecosystem of long-term survivors of multiple myeloma, revealing both reversible and irreversible alterations in the immune compartment.
Collapse
Affiliation(s)
- Raphael Lutz
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- Oncology Center Speyer, Speyer, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Florian Grünschläger
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Malte Simon
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
| | - Mohamed H S Awwad
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marcus Bauer
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-, Wittenberg, Germany
| | - Schayan Yousefian
- Berlin Institute of Health (BIH) at Charité Universitätsmedizin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité Universitätsmedizin, Berlin, Germany
| | - Niklas Beumer
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lea Jopp-Saile
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Anastasia Sedlmeier
- Computational Oncology, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Llorenç Solé-Boldo
- Berlin Institute of Health (BIH) at Charité Universitätsmedizin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité Universitätsmedizin, Berlin, Germany
| | - Bogdan Avanesyan
- Berlin Institute of Health (BIH) at Charité Universitätsmedizin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité Universitätsmedizin, Berlin, Germany
| | - Dominik Vonficht
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Patrick Stelmach
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Georg Steinbuss
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Tobias Boch
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Simon Steiger
- Division of Chromatin Networks, German Cancer Research Center (DKFZ) and BioQuant, Heidelberg, Germany
| | - Marc-Andrea Baertsch
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nina Prokoph
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karsten Rippe
- Division of Chromatin Networks, German Cancer Research Center (DKFZ) and BioQuant, Heidelberg, Germany
| | | | - Claudia Wickenhauser
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-, Wittenberg, Germany
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit EMBL and University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Hübschmann
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Computational Oncology, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Innovation and Service Unit for Bioinformatics and Precision Medicine (BPM), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Niels Weinhold
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marc S Raab
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Medical Faculty and Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany.
| | - Hartmut Goldschmidt
- Department of Medicine V, Hematology, Oncology and Rheumatology, GMMG Studygroup, Heidelberg University Hospital, Heidelberg, Germany.
| | - Charles D Imbusch
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany.
- Research Center for Immunotherapy, University Medical Center Mainz, Mainz, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Mainz, Germany.
| | - Michael Hundemer
- Department of Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Simon Haas
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- Berlin Institute of Health (BIH) at Charité Universitätsmedizin, Berlin, Germany.
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- Charité Universitätsmedizin, Berlin, Germany.
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
9
|
Cramer A, Yang T, Riemann L, Almeida V, Kammeyer C, Abu YE, Gluschke E, Kleiner S, León-Lara X, Janssen A, Hofmann A, Horke A, von Kaisenberg C, Förster R, Beerbaum P, Boehne M, Ravens S. Early-life thymectomy leads to an increase of granzyme-producing γδ T cells in children with congenital heart disease. Nat Commun 2024; 15:9841. [PMID: 39537635 PMCID: PMC11561289 DOI: 10.1038/s41467-024-51673-3] [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: 11/16/2023] [Accepted: 08/14/2024] [Indexed: 11/16/2024] Open
Abstract
Congenital heart disease (CHD) is the most common birth defect in newborns, often requiring cardiac surgery with concomitant thymectomy that is known to increase disease susceptibility later in life. Studies of γδ T cells, which are one of the dominant T cells in the early fetal human thymus, are rare. Here, we provide a comprehensive analysis of the γδ T cell compartment via flow cytometry and next-generation sequencing in children and infants with CHD, who underwent cardiac surgery shortly after birth. A perturbation of the γδ T cell repertoire is evident, and Vδ1 T cell numbers are reduced. However, those cells that are present, do retain cytotoxicity. In contrast, GZMA+CD28+CD161hi innate effector Vγ9Vδ2 T cells are found in higher proportions. TCR-seq identifies an increase in TRDJ3+ γδ T cell clones in children with CHD, but not in a confirmatory group of neonates prior to CHD surgery, which overall points to a persistence of fetal-derived effector γδ T cells in children with CHD.
Collapse
MESH Headings
- Humans
- Heart Defects, Congenital/surgery
- Heart Defects, Congenital/immunology
- Infant
- Thymectomy
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Infant, Newborn
- Male
- Female
- Granzymes/metabolism
- Granzymes/genetics
- Child
- Thymus Gland/immunology
- Child, Preschool
- T-Lymphocytes/immunology
- Flow Cytometry
Collapse
Affiliation(s)
- Alexa Cramer
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Tao Yang
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Lennart Riemann
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Vicente Almeida
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christoph Kammeyer
- Department of Pediatric Cardiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Yusuf E Abu
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Elisa Gluschke
- Department of Pediatric Cardiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Svea Kleiner
- Department of Pediatric Cardiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Ximena León-Lara
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Anika Janssen
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Alejandro Hofmann
- Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | - Alexander Horke
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Constantin von Kaisenberg
- Department of Obstetrics, Gynecology and Reproductive Medicine, Hannover Medical School (MHH), Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625, Hannover, Germany
| | - Philipp Beerbaum
- Department of Pediatric Cardiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Martin Boehne
- Department of Pediatric Cardiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Sarina Ravens
- Institute of Immunology, Hannover Medical School, Hannover, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
10
|
Yue T, Chen SY, Shen WK, Zhang ZY, Cheng L, Guo AY. TCRosetta: An Integrated Analysis and Annotation Platform for T-cell Receptor Sequences. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae013. [PMID: 39436242 DOI: 10.1093/gpbjnl/qzae013] [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: 03/05/2023] [Revised: 12/23/2023] [Accepted: 01/08/2024] [Indexed: 10/23/2024]
Abstract
T cells and T-cell receptors (TCRs) are essential components of the adaptive immune system. Characterization of the TCR repertoire offers a promising and highly informative source for understanding the functions of T cells in the immune response and immunotherapy. Although TCR repertoire studies have attracted much attention, there are few online servers available for TCR repertoire analysis, especially for TCR sequence annotation or advanced analyses. Therefore, we developed TCRosetta, a comprehensive online server that integrates analytical methods for TCR repertoire analysis and visualization. TCRosetta combines general feature analysis, large-scale sequence clustering, network construction, peptide-TCR binding prediction, generation probability calculation, and k-mer motif analysis for TCR sequences, making TCR data analysis as simple as possible. The TCRosetta server accepts multiple input data formats and can analyze ∼ 20,000 TCR sequences in less than 3 min. TCRosetta is the most comprehensive web server available for TCR repertoire analysis and is freely available at https://guolab.wchscu.cn/TCRosetta/.
Collapse
Affiliation(s)
- Tao Yue
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Si-Yi Chen
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wen-Kang Shen
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhan-Ye Zhang
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Liming Cheng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - An-Yuan Guo
- Center for Artificial Intelligence Biology, Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Cao C, Xu M, Wei Y, Peng T, Lin S, Liu X, Xu Y, Chu T, Liu S, Wu P, Hu B, Ding W, Li L, Ma D, Wu P. CXCR4 orchestrates the TOX-programmed exhausted phenotype of CD8 + T cells via JAK2/STAT3 pathway. CELL GENOMICS 2024; 4:100659. [PMID: 39317187 PMCID: PMC11602566 DOI: 10.1016/j.xgen.2024.100659] [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: 02/22/2024] [Revised: 06/17/2024] [Accepted: 08/22/2024] [Indexed: 09/26/2024]
Abstract
Evidence from clinical trials suggests that CXCR4 antagonists enhance immunotherapy effectiveness in several cancers. However, the specific mechanisms through which CXCR4 contributes to immune cell phenotypes are not fully understood. Here, we employed single-cell transcriptomic analysis and identified CXCR4 as a marker gene in T cells, with CD8+PD-1high exhausted T (Tex) cells exhibiting high CXCR4 expression. By blocking CXCR4, the Tex phenotype was attenuated in vivo. Mechanistically, CXCR4-blocking T cells mitigated the Tex phenotype by regulating the JAK2-STAT3 pathway. Single-cell RNA/TCR/ATAC-seq confirmed that Cxcr4-deficient CD8+ T cells epigenetically mitigated the transition from functional to exhausted phenotypes. Notably, clinical sample analysis revealed that CXCR4+CD8+ T cells showed higher expression in patients with a non-complete pathological response. Collectively, these findings demonstrate the mechanism by which CXCR4 orchestrates CD8+ Tex cells and provide a rationale for combining CXCR4 antagonists with immunotherapy in clinical trials.
Collapse
Affiliation(s)
- Canhui Cao
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Miaochun Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ye Wei
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ting Peng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shitong Lin
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaojie Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yashi Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Tian Chu
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shiyi Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ping Wu
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Bai Hu
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wencheng Ding
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Li Li
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ding Ma
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Peng Wu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
12
|
Giannoni E, Sanchez Sanchez G, Verdebout I, Papadopoulou M, Rezwani M, Ahmed R, Ladell K, Miners KL, McLaren JE, Fraser DJ, Price DA, Eberl M, Agyeman PKA, Schlapbach LJ, Vermijlen D. Sepsis shapes the human γδ TCR repertoire in an age- and pathogen-dependent manner. Eur J Immunol 2024; 54:e2451190. [PMID: 39072722 DOI: 10.1002/eji.202451190] [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: 04/15/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
Sepsis affects 25 million children per year globally, leading to 2.9 million deaths and substantial disability in survivors. Extensive characterization of interactions between the host and bacteria in children is required to design novel preventive and therapeutic strategies tailored to this age group. Vγ9Vδ2 T cells are the first T cells generated in humans. These cells are defined by the expression of Vγ9Vδ2 T-cell receptors (TCRs, using the TRGV9 and TRDV2 gene segments), which react strongly against the prototypical bacterial phosphoantigen HMBPP. We investigated this reactivity by analyzing the TCR δ (TRD) repertoire in the blood of 76 children (0-16 years) with blood culture-proven bacterial sepsis caused by HMBPP-positive Escherichia coli or by HMBPP-negative Staphylococcus aureus or by HMBPP-negative Streptococcus pneumoniae. Strikingly, we found that S. aureus, and to a lesser extent E. coli but not S. pneumoniae, shaped the TRDV2 repertoire in young children (<2 years) but not in older children or adults. This dichotomy was due to the selective expansion of a fetal TRDV2 repertoire. Thus, young children possess fetal-derived Vγ9Vδ2 T cells that are highly responsive toward specific bacterial pathogens.
Collapse
Affiliation(s)
- Eric Giannoni
- Clinic of Neonatology, Department Mother-Woman-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Guillem Sanchez Sanchez
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Isoline Verdebout
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Maria Papadopoulou
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Moosa Rezwani
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Raya Ahmed
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Kristin Ladell
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Kelly L Miners
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - James E McLaren
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Donald J Fraser
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
- Wales Kidney Research Unit, Heath Park Campus, Cardiff, UK
- Directorate of Nephrology and Transplantation, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - David A Price
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Matthias Eberl
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Philipp K A Agyeman
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Luregn J Schlapbach
- Department of Intensive Care and Neonatology, and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- Child Health Research Centre, University of Queensland, Brisbane, Australia
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
13
|
Castro JP, Shindyapina AV, Barbieri A, Ying K, Strelkova OS, Paulo JA, Tyshkovskiy A, Meinl R, Kerepesi C, Petrashen AP, Mariotti M, Meer MV, Hu Y, Karamyshev A, Losyev G, Galhardo M, Logarinho E, Indzhykulian AA, Gygi SP, Sedivy JM, Manis JP, Gladyshev VN. Age-associated clonal B cells drive B cell lymphoma in mice. NATURE AGING 2024; 4:1403-1417. [PMID: 39117982 DOI: 10.1038/s43587-024-00671-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 06/19/2024] [Indexed: 08/10/2024]
Abstract
Although cancer is an age-related disease, how the processes of aging contribute to cancer progression is not well understood. In this study, we uncovered how mouse B cell lymphoma develops as a consequence of a naturally aged system. We show here that this malignancy is associated with an age-associated clonal B cell (ACBC) population that likely originates from age-associated B cells. Driven by c-Myc activation, promoter hypermethylation and somatic mutations, IgM+ ACBCs clonally expand independently of germinal centers and show increased biological age. ACBCs become self-sufficient and support malignancy when transferred into young recipients. Inhibition of mTOR or c-Myc in old mice attenuates pre-malignant changes in B cells during aging. Although the etiology of mouse and human B cell lymphomas is considered distinct, epigenetic changes in transformed mouse B cells are enriched for changes observed in human B cell lymphomas. Together, our findings characterize the spontaneous progression of cancer during aging through both cell-intrinsic and microenvironmental changes and suggest interventions for its prevention.
Collapse
Affiliation(s)
- José P Castro
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Aging and Aneuploidy Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | | | | | - Kejun Ying
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Olga S Strelkova
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - João A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Rico Meinl
- Retro Biosciences, Redwood City, CA, USA
| | - Csaba Kerepesi
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Institute for Computer Science and Control (SZTAKI), Loránd Eötvös Research Network, Budapest, Hungary
| | - Anna P Petrashen
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Marco Mariotti
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Barcelona, Spain
| | - Margarita V Meer
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- San Diego Institute of Sciences, Altos Labs, San Diego, CA, USA
| | - Yan Hu
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Grigoriy Losyev
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mafalda Galhardo
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Elsa Logarinho
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Artur A Indzhykulian
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - John M Sedivy
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - John P Manis
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vadim N Gladyshev
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Huang J, Mao L, Lei Q, Guo AY. Bioinformatics tools and resources for cancer and application. Chin Med J (Engl) 2024; 137:2052-2064. [PMID: 39075637 PMCID: PMC11374212 DOI: 10.1097/cm9.0000000000003254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Indexed: 07/31/2024] Open
Abstract
ABSTRACT Tumor bioinformatics plays an important role in cancer research and precision medicine. The primary focus of traditional cancer research has been molecular and clinical studies of a number of fundamental pathways and genes. In recent years, driven by breakthroughs in high-throughput technologies, large-scale cancer omics data have accumulated rapidly. How to effectively utilize and share these data is particularly important. To address this crucial task, many computational tools and databases have been developed over the past few years. To help researchers quickly learn and understand the functions of these tools, in this review, we summarize publicly available bioinformatics tools and resources for pan-cancer multi-omics analysis, regulatory analysis of tumorigenesis, tumor treatment and prognosis, immune infiltration analysis, immune repertoire analysis, cancer driver gene and driver mutation analysis, and cancer single-cell analysis, which may further help researchers find more suitable tools for their research.
Collapse
Affiliation(s)
- Jin Huang
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lingzi Mao
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Qian Lei
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - An-Yuan Guo
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
15
|
Wei YC, Pospiech M, Meng Y, Alachkar H. Development and characterization of human T-cell receptor (TCR) alpha and beta clones' library as biological standards and resources for TCR sequencing and engineering. Biol Methods Protoc 2024; 9:bpae064. [PMID: 39507623 PMCID: PMC11540440 DOI: 10.1093/biomethods/bpae064] [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: 04/22/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 11/08/2024] Open
Abstract
Characterization of T-cell receptors (TCRs) repertoire was revolutionized by next-generation sequencing technologies; however, standardization using biological controls to facilitate precision of current alignment and assembly tools remains a challenge. Additionally, availability of TCR libraries for off-the-shelf cloning and engineering TCR-specific T cells is a valuable resource for TCR-based immunotherapies. We established nine human TCR α and β clones that were evaluated using the 5'-rapid amplification of cDNA ends-like RNA-based TCR sequencing on the Illumina platform. TCR sequences were extracted and aligned using MiXCR, TRUST4, and CATT to validate their sensitivity and specificity and to validate library preparation methods. The correlation between actual and expected TCR ratios within libraries confirmed accuracy of the approach. Our findings established the development of biological standards and library of TCR clones to be leveraged in TCR sequencing and engineering. The remaining human TCR clones' libraries for a more diverse biological control will be generated.
Collapse
Affiliation(s)
- Yu-Chun Wei
- Department of Clinical Pharmacy, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, 90089, United States
| | - Mateusz Pospiech
- Department of Clinical Pharmacy, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, 90089, United States
| | - Yiting Meng
- Department of Clinical Pharmacy, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, 90089, United States
| | - Houda Alachkar
- Department of Clinical Pharmacy, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, 90089, United States
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90089, United States
| |
Collapse
|
16
|
Marín-Benesiu F, Chica-Redecillas L, Arenas-Rodríguez V, de Santiago E, Martínez-Diz S, López-Torres G, Cortés-Valverde AI, Romero-Cachinero C, Entrala-Bernal C, Fernandez-Rosado FJ, Martínez-González LJ, Alvarez-Cubero MJ. The T-cell repertoire of Spanish patients with COVID-19 as a strategy to link T-cell characteristics to the severity of the disease. Hum Genomics 2024; 18:94. [PMID: 39227859 PMCID: PMC11373388 DOI: 10.1186/s40246-024-00654-0] [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: 04/22/2024] [Accepted: 08/06/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND The architecture and dynamics of T cell populations are critical in orchestrating the immune response to SARS-CoV-2. In our study, we used T Cell Receptor sequencing (TCRseq) to investigate TCR repertoires in 173 post-infection COVID-19 patients. METHODS The cohort included 98 mild and 75 severe cases with a median age of 53. We amplified and sequenced the TCR β chain Complementary Determining Region 3 (CDR3b) and performed bioinformatic analyses to assess repertoire diversity, clonality, and V/J allelic usage between age, sex and severity groups. CDR3b amino acid sequence inference was performed by clustering structural motifs and filtering validated reactive CDR3b to COVID-19. RESULTS Our results revealed a pronounced decrease in diversity and an increase in clonal expansion in the TCR repertoires of severe COVID-19 patients younger than 55 years old. These results reflect the observed trends in patients older than 55 years old (both mild and severe). In addition, we identified a significant reduction in the usage of key V alleles (TRBV14, TRBV19, TRBV15 and TRBV6-4) associated with disease severity. Notably, severe patients under 55 years old had allelic patterns that resemble those over 55 years old, accompanied by a skewed frequency of COVID-19-related motifs. CONCLUSIONS Present results suggest that severe patients younger than 55 may have a compromised TCR repertoire contributing to a worse disease outcome.
Collapse
MESH Headings
- Humans
- COVID-19/genetics
- COVID-19/immunology
- COVID-19/virology
- Male
- Middle Aged
- Female
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- SARS-CoV-2/pathogenicity
- Severity of Illness Index
- Adult
- Aged
- Complementarity Determining Regions/genetics
- Complementarity Determining Regions/immunology
- Spain
- T-Lymphocytes/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Alleles
Collapse
Affiliation(s)
- Fernando Marín-Benesiu
- Department of Biochemistry, Molecular Biology III and Inmunology, Faculty of Medicine, University of Granada, Parque Tecnológico de la Salud, Avd. de la Investigación nº 11, Tower C. 11th floor, Granada, 18071, Spain
- Centre for Genomics and Oncological Research: Pfizer, Andalusian Regional Government, GENYO, University of Granada, Parque Tecnológico de la Salud, Granada, Spain
| | - Lucia Chica-Redecillas
- Department of Biochemistry, Molecular Biology III and Inmunology, Faculty of Medicine, University of Granada, Parque Tecnológico de la Salud, Avd. de la Investigación nº 11, Tower C. 11th floor, Granada, 18071, Spain
- Centre for Genomics and Oncological Research: Pfizer, Andalusian Regional Government, GENYO, University of Granada, Parque Tecnológico de la Salud, Granada, Spain
| | - Verónica Arenas-Rodríguez
- Centre for Genomics and Oncological Research: Pfizer, Andalusian Regional Government, GENYO, University of Granada, Parque Tecnológico de la Salud, Granada, Spain
| | - Esperanza de Santiago
- Centre for Genomics and Oncological Research: Pfizer, Andalusian Regional Government, GENYO, University of Granada, Parque Tecnológico de la Salud, Granada, Spain
| | - Silvia Martínez-Diz
- Preventive Medicine and Public Health Service, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | | | | | | | - Carmen Entrala-Bernal
- LORGEN G.P, Ciencias de la Salud - Business Innovation Centre (BIC), Granada, PT, Spain
| | | | - Luis Javier Martínez-González
- Department of Biochemistry, Molecular Biology III and Inmunology, Faculty of Medicine, University of Granada, Parque Tecnológico de la Salud, Avd. de la Investigación nº 11, Tower C. 11th floor, Granada, 18071, Spain.
- Centre for Genomics and Oncological Research: Pfizer, Andalusian Regional Government, GENYO, University of Granada, Parque Tecnológico de la Salud, Granada, Spain.
| | - Maria Jesus Alvarez-Cubero
- Department of Biochemistry, Molecular Biology III and Inmunology, Faculty of Medicine, University of Granada, Parque Tecnológico de la Salud, Avd. de la Investigación nº 11, Tower C. 11th floor, Granada, 18071, Spain
- Centre for Genomics and Oncological Research: Pfizer, Andalusian Regional Government, GENYO, University of Granada, Parque Tecnológico de la Salud, Granada, Spain
- Ibs Granada, Biosanitary Research Institute of Granada, Granada, Spain
| |
Collapse
|
17
|
Serdyuk YV, Zornikova KV, Dianov DV, Ivanova NO, Davydova VD, Fefelova EI, Nenasheva TA, Sheetikov SA, Bogolyubova AV. T-Cell Receptors Cross-Reactive to Coronaviral Epitopes Homologous to the SPR Peptide. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1631-1642. [PMID: 39418521 DOI: 10.1134/s0006297924090098] [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: 05/15/2024] [Revised: 07/26/2024] [Accepted: 08/09/2024] [Indexed: 10/19/2024]
Abstract
The COVID-19 pandemic caused by the rapid spread of the novel coronavirus SARS-CoV-2, has promoted an interest in studying the T-cell immune response. It was found that the polyclonal and cross-reactive T-cell response against seasonal coronaviruses and other SARS-CoV-2 strains reduced disease severity. We investigated the immunodominant T-cell epitope SPRWYFYYYL from the nucleocapsid protein of SARS-CoV-2. The immune response to this epitope is characterized by the formation of highly homologous (convergent) receptors that have been found in the T-cell receptor (TCR) repertoires of different individuals. This epitope belongs to a group of highly conserved peptides that are rarely mutated in novel SARS-CoV-2 strains and are homologous to the epitopes of seasonal coronaviruses. It has been suggested that the cross-reactive response to homologous peptides contributes to the reduction of COVID-19 severity. However, some investigators have questioned this hypothesis, suggesting that the low affinity of the cross-reactive receptors reduces the strength of the immune response. The aim of this study was to evaluate the effect of amino acid substitutions in the SPR epitope on its binding affinity to specific TCRs. For this, we performed antigen-dependent cellular expansions were performed using samples from four COVID-19-transfected donors and sequenced their TCR repertoires. The resulting SPR-specific repertoire of β-chains in TCRs had a greater sequence diversity than the repertoire of α-chains. However, the TCR repertoires of all four donors contained public receptors, three of which were cloned and used to generate the Jurkat E6-1 TPR cell line. Only one of these receptors was activated by the SPR peptide and recognized with the same affinity by its mutant homologue LPRWYFYYY from seasonal coronaviruses. This indicates that the presence of the mutation did not affect the strength of the immune response, which may explain why the cross-reactive response to the SPR epitope is so frequent and contributes positively to COVID-19 infection.
Collapse
Affiliation(s)
- Yana V Serdyuk
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Ksenia V Zornikova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Dmitry V Dianov
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Nataliia O Ivanova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Vassa D Davydova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Ekaterina I Fefelova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Tatiana A Nenasheva
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Saveliy A Sheetikov
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Apollinariya V Bogolyubova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia.
| |
Collapse
|
18
|
Schattgen SA, Turner JS, Ghonim MA, Crawford JC, Schmitz AJ, Kim H, Zhou JQ, Awad W, Mettelman RC, Kim W, McIntire KM, Haile A, Klebert MK, Suessen T, Middleton WD, Teefey SA, Presti RM, Ellebedy AH, Thomas PG. Influenza vaccination stimulates maturation of the human T follicular helper cell response. Nat Immunol 2024; 25:1742-1753. [PMID: 39164477 PMCID: PMC11362011 DOI: 10.1038/s41590-024-01926-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 07/11/2024] [Indexed: 08/22/2024]
Abstract
The differentiation and specificity of human CD4+ T follicular helper cells (TFH cells) after influenza vaccination have been poorly defined. Here we profiled blood and draining lymph node (LN) samples from human volunteers for over 2 years after two influenza vaccines were administered 1 year apart to define the evolution of the CD4+ TFH cell response. The first vaccination induced an increase in the frequency of circulating TFH (cTFH) and LN TFH cells at week 1 postvaccination. This increase was transient for cTFH cells, whereas the LN TFH cells further expanded during week 2 and remained elevated in frequency for at least 3 months. We observed several distinct subsets of TFH cells in the LN, including pre-TFH cells, memory TFH cells, germinal center (GC) TFH cells and interleukin-10+ TFH cell subsets beginning at baseline and at all time points postvaccination. The shift toward a GC TFH cell phenotype occurred with faster kinetics after the second vaccine compared to the first vaccine. We identified several influenza-specific TFH cell clonal lineages, including multiple responses targeting internal influenza virus proteins, and found that each TFH cell state was attainable within a clonal lineage. Thus, human TFH cells form a durable and dynamic multitissue network.
Collapse
Affiliation(s)
- Stefan A Schattgen
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jackson S Turner
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mohamed A Ghonim
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jeremy Chase Crawford
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Aaron J Schmitz
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hyunjin Kim
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Julian Q Zhou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Walid Awad
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Robert C Mettelman
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Wooseob Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Microbiology, Korea University College of Medicine, Seoul, Korea
| | - Katherine M McIntire
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Alem Haile
- Clinical Trials Unit, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael K Klebert
- Clinical Trials Unit, Washington University School of Medicine, St. Louis, MO, USA
| | - Teresa Suessen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - William D Middleton
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sharlene A Teefey
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel M Presti
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, USA.
| | - Paul G Thomas
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
19
|
Zhuo Z, Wang J, Zhang Y, Meng G. Integrative alternative splicing analysis reveals new prognosis signature in B-cell acute lymphoblastic leukemia. Int J Biol Sci 2024; 20:4496-4512. [PMID: 39247833 PMCID: PMC11380455 DOI: 10.7150/ijbs.98899] [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: 05/26/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
The dysregulation of alternative splicing (AS) is increasingly recognized as a pivotal player in the pathogenesis, progression, and treatment resistance of B-cell acute lymphoblastic leukemia (B-ALL). Despite its significance, the clinical implications of AS events in B-ALL remain largely unexplored. This study developed a prognostic model based on 18 AS events (18-AS), derived from a meticulous integration of bioinformatics methodologies and advanced machine learning algorithms. The 18-AS signature observed in B-ALL distinctly categorized patients into different groups with significant differences in immune infiltration, V(D)J rearrangement, drug sensitivity, and immunotherapy outcomes. Patients classified within the high 18-AS group exhibited lower immune infiltration scores, poorer chemo- and immune-therapy responses, and worse overall survival, underscoring the model's potential in refining therapeutic strategies. To validate the clinical applicability of the 18-AS, we established an SF-AS regulatory network and identified candidate drugs. More importantly, we conducted in vitro cell proliferation assays to confirm our analysis, demonstrating that the High-18AS cell line (SUP-B15) exhibited significantly enhanced sensitivity to Dasatinib, Dovitinib, and Midostaurin compared to the Low-18AS cell line (REH). These findings reveal AS events as novel prognostic biomarkers and therapeutic targets, advancing personalized treatment strategies in B-ALL management.
Collapse
Affiliation(s)
- Zhiyi Zhuo
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 197 Ruijin Er Road, Shanghai 200025, P. R. China
- Department of Geriatrics and Medical Center on Aging, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, First Affiliated Hospital of Xinjiang Medical University, Xinjiang, P. R. China
| | - Junfei Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 197 Ruijin Er Road, Shanghai 200025, P. R. China
- Department of Geriatrics and Medical Center on Aging, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, First Affiliated Hospital of Xinjiang Medical University, Xinjiang, P. R. China
| | - Yonglei Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 197 Ruijin Er Road, Shanghai 200025, P. R. China
- Department of Geriatrics and Medical Center on Aging, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, First Affiliated Hospital of Xinjiang Medical University, Xinjiang, P. R. China
| | - Guoyu Meng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 197 Ruijin Er Road, Shanghai 200025, P. R. China
- Department of Geriatrics and Medical Center on Aging, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, First Affiliated Hospital of Xinjiang Medical University, Xinjiang, P. R. China
| |
Collapse
|
20
|
Patin EC, Nenclares P, Chan Wah Hak C, Dillon MT, Patrikeev A, McLaughlin M, Grove L, Foo S, Soliman H, Barata JP, Marsden J, Baldock H, Gkantalis J, Roulstone V, Kyula J, Burley A, Hubbard L, Pedersen M, Smith SA, Clancy-Thompson E, Melcher AA, Ono M, Rullan A, Harrington KJ. Sculpting the tumour microenvironment by combining radiotherapy and ATR inhibition for curative-intent adjuvant immunotherapy. Nat Commun 2024; 15:6923. [PMID: 39134540 PMCID: PMC11319479 DOI: 10.1038/s41467-024-51236-6] [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: 01/05/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
The combination of radiotherapy/chemoradiotherapy and immune checkpoint blockade can result in poor outcomes in patients with locally advanced head and neck squamous cell carcinoma (HNSCC). Here, we show that combining ATR inhibition (ATRi) with radiotherapy (RT) increases the frequency of activated NKG2A+PD-1+ T cells in animal models of HNSCC. Compared with the ATRi/RT treatment regimen alone, the addition of simultaneous NKG2A and PD-L1 blockade to ATRi/RT, in the adjuvant, post-radiotherapy setting induces a robust antitumour response driven by higher infiltration and activation of cytotoxic T cells in the tumour microenvironment. The efficacy of this combination relies on CD40/CD40L costimulation and infiltration of activated, proliferating memory CD8+ and CD4+ T cells with persistent or new T cell receptor (TCR) signalling, respectively. We also observe increased richness in the TCR repertoire and emergence of numerous and large TCR clonotypes that cluster based on antigen specificity in response to NKG2A/PD-L1/ATRi/RT. Collectively, our data point towards potential combination approaches for the treatment of HNSCC.
Collapse
Affiliation(s)
- Emmanuel C Patin
- Targeted Therapy Team, The Institute of Cancer Research, London, UK.
| | - Pablo Nenclares
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Charleen Chan Wah Hak
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Magnus T Dillon
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Anton Patrikeev
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | - Lorna Grove
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Shane Foo
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | | | | | - Holly Baldock
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Jim Gkantalis
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | - Joan Kyula
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Amy Burley
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Lisa Hubbard
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Malin Pedersen
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | | | - Alan A Melcher
- Translational Immunotherapy Team, The Institute of Cancer Research, London, UK
| | - Masahiro Ono
- Department of Life Sciences, Imperial College London, London, UK
| | - Antonio Rullan
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Kevin J Harrington
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| |
Collapse
|
21
|
Aziz F, Hisatsune J, Ono HK, Kajimura J, Yu L, Masuda K, Kitagawa H, Sato'o Y, Yahara K, Yamaoka M, Nakane A, Kawasaki H, Obata S, Fukushima-Nomura A, Ito Y, Aung MS, Amagai M, Salasia SIO, Ohge H, Kusunoki Y, Sugai M. Genomic analysis and identification of a novel superantigen, SargEY, in Staphylococcus argenteus isolated from atopic dermatitis lesions. mSphere 2024; 9:e0050524. [PMID: 38990001 PMCID: PMC11288046 DOI: 10.1128/msphere.00505-24] [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: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/12/2024] Open
Abstract
During surveillance of Staphylococcus aureus in lesions from patients with atopic dermatitis (AD), we isolated Staphylococcus argenteus, a species registered in 2011 as a new member of the genus Staphylococcus and previously considered a lineage of S. aureus. Genome sequence comparisons between S. argenteus isolates and representative S. aureus clinical isolates from various origins revealed that the S. argenteus genome from AD patients closely resembles that of S. aureus causing skin infections. We previously reported that 17%-22% of S. aureus isolated from skin infections produce staphylococcal enterotoxin Y (SEY), which predominantly induces T-cell proliferation via the T-cell receptor (TCR) Vα pathway. Complete genome sequencing of S. argenteus isolates revealed a gene encoding a protein similar to superantigen SEY, designated as SargEY, on its chromosome. Population structure analysis of S. argenteus revealed that these isolates are ST2250 lineage, which was the only lineage positive for the SEY-like gene among S. argenteus. Recombinant SargEY demonstrated immunological cross-reactivity with anti-SEY serum. SargEY could induce proliferation of human CD4+ and CD8+ T cells, as well as production of TNF-α and IFN-γ. SargEY showed emetic activity in a marmoset monkey model. SargEY and SET (a phylogenetically close but uncharacterized SE) revealed their dependency on TCR Vα in inducing human T-cell proliferation. Additionally, TCR sequencing revealed other previously undescribed Vα repertoires induced by SEH. SargEY and SEY may play roles in exacerbating the respective toxin-producing strains in AD. IMPORTANCE Staphylococcus aureus is frequently isolated from active lesions of atopic dermatitis (AD) patients. We reported that 17%-22% of S. aureus isolated from AD patients produced a novel superantigen staphylococcal enterotoxin Y (SEY). Unlike many S. aureus superantigens that activate T cells via T-cell receptor (TCR) Vß, SEY activates T cells via TCR Vα and stimulates cytokine secretion. Staphylococcus argenteus was isolated from AD patients during the surveillance for S. aureus. Phylogenetic comparison of the genome indicated that the isolate was very similar to S. aureus causing skin infections. The isolate encoded a SEY-like protein, designated SargEY, which, like SEY, activated T cells via the TCR Vα. ST2250 is the only lineage positive for SargEY gene. ST2250 S. argenteus harboring a superantigen SargEY gene may be a novel staphylococcal clone that infects human skin and is involved in the exacerbation of AD.
Collapse
Affiliation(s)
- Fatkhanuddin Aziz
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima City, Hiroshima, Japan
- Veterinary Technology Program, Department of Bioresources Technology and Veterinary, Vocational College, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Junzo Hisatsune
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima City, Hiroshima, Japan
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| | - Hisaya K. Ono
- Laboratory of Zoonoses, Kitasato University School of Veterinary Medicine, Towada, Aomori, Japan
| | - Junko Kajimura
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima City, Hiroshima, Japan
| | - Liansheng Yu
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| | - Kanako Masuda
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima City, Hiroshima, Japan
| | - Hiroki Kitagawa
- Department of Infectious Diseases, Hiroshima University Hospital, Hiroshima City, Hiroshima, Japan
| | - Yusuke Sato'o
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke-shi, Tochigi, Japan
| | - Koji Yahara
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| | - Mika Yamaoka
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima City, Hiroshima, Japan
| | - Akio Nakane
- Department of Microbiology and Immunology, Hirosaki University, Hirosaki, Aomori, Japan
| | - Hiroshi Kawasaki
- Department of Dermatology, Keio University School of Medicine, Shijuku-ku, Tokyo, Japan
- Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Laboratory for Developmental Genetics, RIKEN, Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Shoko Obata
- Department of Dermatology, Keio University School of Medicine, Shijuku-ku, Tokyo, Japan
| | | | - Yoshihiro Ito
- Department of Dermatology, Keio University School of Medicine, Shijuku-ku, Tokyo, Japan
| | - Meiji Soe Aung
- Department of Hygiene, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Shijuku-ku, Tokyo, Japan
- Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Siti Isrina Oktavia Salasia
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Hiroki Ohge
- Department of Infectious Diseases, Hiroshima University Hospital, Hiroshima City, Hiroshima, Japan
| | - Yoichiro Kusunoki
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima City, Hiroshima, Japan
| | - Motoyuki Sugai
- Department of Bacteriology, Graduate School of Biomedical and Health Sciences, Hiroshima City, Hiroshima, Japan
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| |
Collapse
|
22
|
Yeh AC, Koyama M, Waltner OG, Minnie SA, Boiko JR, Shabaneh TB, Takahashi S, Zhang P, Ensbey KS, Schmidt CR, Legg SRW, Sekiguchi T, Nelson E, Bhise SS, Stevens AR, Goodpaster T, Chakka S, Furlan SN, Markey KA, Bleakley ME, Elson CO, Bradley PH, Hill GR. Microbiota dictate T cell clonal selection to augment graft-versus-host disease after stem cell transplantation. Immunity 2024; 57:1648-1664.e9. [PMID: 38876098 PMCID: PMC11236519 DOI: 10.1016/j.immuni.2024.05.018] [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: 06/26/2023] [Revised: 02/09/2024] [Accepted: 05/20/2024] [Indexed: 06/16/2024]
Abstract
Allogeneic T cell expansion is the primary determinant of graft-versus-host disease (GVHD), and current dogma dictates that this is driven by histocompatibility antigen disparities between donor and recipient. This paradigm represents a closed genetic system within which donor T cells interact with peptide-major histocompatibility complexes (MHCs), though clonal interrogation remains challenging due to the sparseness of the T cell repertoire. We developed a Bayesian model using donor and recipient T cell receptor (TCR) frequencies in murine stem cell transplant systems to define limited common expansion of T cell clones across genetically identical donor-recipient pairs. A subset of donor CD4+ T cell clonotypes differentially expanded in identical recipients and were microbiota dependent. Microbiota-specific T cells augmented GVHD lethality and could target microbial antigens presented by gastrointestinal epithelium during an alloreactive response. The microbiota serves as a source of cognate antigens that contribute to clonotypic T cell expansion and the induction of GVHD independent of donor-recipient genetics.
Collapse
MESH Headings
- Graft vs Host Disease/immunology
- Graft vs Host Disease/microbiology
- Animals
- Mice
- Mice, Inbred C57BL
- CD4-Positive T-Lymphocytes/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Microbiota/immunology
- Clonal Selection, Antigen-Mediated
- Transplantation, Homologous
- Bayes Theorem
- Stem Cell Transplantation/adverse effects
- Mice, Inbred BALB C
- Gastrointestinal Microbiome/immunology
- Hematopoietic Stem Cell Transplantation/adverse effects
Collapse
Affiliation(s)
- Albert C Yeh
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Motoko Koyama
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Olivia G Waltner
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Simone A Minnie
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Julie R Boiko
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tamer B Shabaneh
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Shuichiro Takahashi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ping Zhang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kathleen S Ensbey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Christine R Schmidt
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Samuel R W Legg
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tomoko Sekiguchi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ethan Nelson
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Shruti S Bhise
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Andrew R Stevens
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tracy Goodpaster
- Experimental Histopathology Core, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Saranya Chakka
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Scott N Furlan
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kate A Markey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Marie E Bleakley
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Division of Hematology, Oncology, and Bone Marrow Transplantation, Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Charles O Elson
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Philip H Bradley
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Biochemistry and Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Geoffrey R Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
23
|
Lisi S, Malerba F, Quaranta P, Florio R, Vitaloni O, Monaca E, Bruni Ercole B, Bitonti AR, Del Perugia O, Mignanelli M, Perrera P, Sabbatella R, Raimondi F, Piazza CR, Moles A, Alfano C, Pistello M, Cattaneo A. Selection and characterization of human scFvs targeting the SARS-CoV-2 nucleocapsid protein isolated from antibody libraries of COVID-19 patients. Sci Rep 2024; 14:15864. [PMID: 38982108 PMCID: PMC11233501 DOI: 10.1038/s41598-024-66558-0] [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: 01/18/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024] Open
Abstract
In 2019, the novel SARS-CoV-2 coronavirus emerged in China, causing the pneumonia named COVID-19. At the beginning, all research efforts were focused on the spike (S) glycoprotein. However, it became evident that the nucleocapsid (N) protein is pivotal in viral replication, genome packaging and evasion of the immune system, is highly immunogenic, which makes it another compelling target for antibody development alongside the spike protein. This study focused on the construction of single chain fragments variable (scFvs) libraries from SARS-CoV-2-infected patients to establish a valuable, immortalized and extensive antibodies source. We used the Intracellular Antibody Capture Technology to select a panel of scFvs against the SARS-CoV-2 N protein. The whole panel of scFv was expressed and characterized both as intrabodies and recombinant proteins. ScFvs were then divided into 2 subgroups: those that exhibited high binding activity to N protein when expressed in yeast or in mammalian cells as intrabodies, and those purified as recombinant proteins, displaying affinity for recombinant N protein in the nanomolar range. This panel of scFvs against the N protein represents a novel platform for research and potential diagnostic applications.
Collapse
Affiliation(s)
- Simonetta Lisi
- Bio@SNS Laboratory, Scuola Normale Superiore, 56126, Pisa, Italy
| | - Francesca Malerba
- Fondazione EBRI (European Brain Research Institute) Rita Levi-Montalcini, 00161, Rome, Italy
| | - Paola Quaranta
- Retrovirus Centre, Department of Translational Research, University of Pisa, 56126, Pisa, Italy
- Virology Operative Unit, Pisa University Hospital, 56124, Pisa, Italy
| | - Rita Florio
- Fondazione EBRI (European Brain Research Institute) Rita Levi-Montalcini, 00161, Rome, Italy
| | - Ottavia Vitaloni
- Bio@SNS Laboratory, Scuola Normale Superiore, 56126, Pisa, Italy
| | - Elisa Monaca
- Structural Biology and Biophysics Unit, Fondazione Ri.MED, 90133, Palermo, Italy
| | - Bruno Bruni Ercole
- Fondazione EBRI (European Brain Research Institute) Rita Levi-Montalcini, 00161, Rome, Italy
| | | | - Olga Del Perugia
- Bio@SNS Laboratory, Scuola Normale Superiore, 56126, Pisa, Italy
| | | | - Paola Perrera
- Retrovirus Centre, Department of Translational Research, University of Pisa, 56126, Pisa, Italy
| | - Raffaele Sabbatella
- Structural Biology and Biophysics Unit, Fondazione Ri.MED, 90133, Palermo, Italy
| | | | - Carmen Rita Piazza
- Retrovirus Centre, Department of Translational Research, University of Pisa, 56126, Pisa, Italy
- Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Anna Moles
- Genomnia Srl, 20091, Bresso, MI, Italy
- Institute of Biochemistry and Cell Biology, CNR, 80131, Napoli, Italy
| | - Caterina Alfano
- Structural Biology and Biophysics Unit, Fondazione Ri.MED, 90133, Palermo, Italy
| | - Mauro Pistello
- Retrovirus Centre, Department of Translational Research, University of Pisa, 56126, Pisa, Italy
- Virology Operative Unit, Pisa University Hospital, 56124, Pisa, Italy
| | - Antonino Cattaneo
- Bio@SNS Laboratory, Scuola Normale Superiore, 56126, Pisa, Italy.
- Fondazione EBRI (European Brain Research Institute) Rita Levi-Montalcini, 00161, Rome, Italy.
| |
Collapse
|
24
|
Sorel N, Díaz-Pascual F, Bessot B, Sadek H, Mollet C, Chouteau M, Zahn M, Gil-Farina I, Tajer P, van Eggermond M, Berghuis D, Lankester AC, André I, Gabriel R, Cavazzana M, Pike-Overzet K, Staal FJT, Lagresle-Peyrou C. Restoration of T and B Cell Differentiation after RAG1 Gene Transfer in Human RAG1 Defective Hematopoietic Stem Cells. Biomedicines 2024; 12:1495. [PMID: 39062069 PMCID: PMC11275127 DOI: 10.3390/biomedicines12071495] [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: 04/12/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Recombinase-activating gene (RAG)-deficient SCID patients lack B and T lymphocytes due to the inability to rearrange immunoglobulin and T cell receptor genes. The two RAG genes act as a required dimer to initiate gene recombination. Gene therapy is a valid treatment alternative for RAG-SCID patients who lack a suitable bone marrow donor, but developing such therapy for RAG1/2 has proven challenging. Using a clinically approved lentiviral vector with a codon-optimized RAG1 gene, we report here preclinical studies using CD34+ cells from four RAG1-SCID patients. We used in vitro T cell developmental assays and in vivo assays in xenografted NSG mice. The RAG1-SCID patient CD34+ cells transduced with the RAG1 vector and transplanted into NSG mice led to restored human B and T cell development. Together with favorable safety data on integration sites, these results substantiate an ongoing phase I/II clinical trial for RAG1-SCID.
Collapse
Affiliation(s)
- Nataël Sorel
- Human Lymphohematopoiesis Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, 75015 Paris, France (I.A.)
| | | | - Boris Bessot
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, 75015 Paris, France
| | - Hanem Sadek
- Human Lymphohematopoiesis Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, 75015 Paris, France (I.A.)
| | - Chloé Mollet
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, 75015 Paris, France
| | - Myriam Chouteau
- Human Lymphohematopoiesis Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, 75015 Paris, France (I.A.)
| | - Marco Zahn
- ProtaGene CGT GmbH, Im Neuenheimer Feld 582, 69120 Heidelberg, Germany
| | - Irene Gil-Farina
- ProtaGene CGT GmbH, Im Neuenheimer Feld 582, 69120 Heidelberg, Germany
| | - Parisa Tajer
- Department of Immunohematology and Blood Transfusion, L3-Q Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marja van Eggermond
- Department of Immunohematology and Blood Transfusion, L3-Q Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Dagmar Berghuis
- Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (D.B.); (A.C.L.)
| | - Arjan C. Lankester
- Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (D.B.); (A.C.L.)
| | - Isabelle André
- Human Lymphohematopoiesis Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, 75015 Paris, France (I.A.)
| | - Richard Gabriel
- ProtaGene CGT GmbH, Im Neuenheimer Feld 582, 69120 Heidelberg, Germany
| | - Marina Cavazzana
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, 75015 Paris, France
- Biotherapy Department, Necker-Enfants Malades Hospital, AP-HP, 75015 Paris, France;
- Imagine Institute UMR1163, Université Paris Cité, Sorbonne Paris Cité, 75015 Paris, France
| | - Kasrin Pike-Overzet
- Biotherapy Department, Necker-Enfants Malades Hospital, AP-HP, 75015 Paris, France;
| | - Frank J. T. Staal
- Department of Immunohematology and Blood Transfusion, L3-Q Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (D.B.); (A.C.L.)
| | - Chantal Lagresle-Peyrou
- Human Lymphohematopoiesis Laboratory, Université Paris Cité, Imagine Institute, INSERM UMR 1163, 75015 Paris, France (I.A.)
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, 75015 Paris, France
| |
Collapse
|
25
|
Mistri SK, Hilton BM, Horrigan KJ, Andretta ES, Savard R, Dienz O, Hampel KJ, Gerrard DL, Rose JT, Sidiropoulos N, Majumdar D, Boyson JE. SLAM/SAP signaling regulates discrete γδ T cell developmental checkpoints and shapes the innate-like γδ TCR repertoire. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.575073. [PMID: 38260519 PMCID: PMC10802474 DOI: 10.1101/2024.01.10.575073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
During thymic development, most γδ T cells acquire innate-like characteristics that are critical for their function in tumor surveillance, infectious disease, and tissue repair. The mechanisms, however, that regulate γδ T cell developmental programming remain unclear. Recently, we demonstrated that the SLAM-SAP signaling pathway regulates the development and function of multiple innate-like γδ T cell subsets. Here, we used a single-cell proteogenomics approach to identify SAP-dependent developmental checkpoints and to define the SAP-dependent γδ TCR repertoire. SAP deficiency resulted in both a significant loss of an immature Gzma + Blk + Etv5 + Tox2 + γδT17 precursor population, and a significant increase in Cd4 + Cd8+ Rorc + Ptcra + Rag1 + thymic γδ T cells. SAP-dependent diversion of embryonic day 17 thymic γδ T cell clonotypes into the αβ T cell developmental pathway was associated with a decreased frequency of mature clonotypes in neonatal thymus, and an altered γδ TCR repertoire in the periphery. Finally, we identify TRGV4/TRAV13-4(DV7)-expressing T cells as a novel, SAP-dependent Vγ4 γδT1 subset. Together, the data suggest that SAP-dependent γδ/αβ T cell lineage commitment regulates γδ T cell developmental programming and shapes the γδ TCR repertoire.
Collapse
Affiliation(s)
- Somen K Mistri
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Brianna M. Hilton
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Katherine J. Horrigan
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Emma S. Andretta
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Remi Savard
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Oliver Dienz
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Kenneth J Hampel
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical Center, Burlington, Vermont 05405, USA
| | - Diana L. Gerrard
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical Center, Burlington, Vermont 05405, USA
| | - Joshua T. Rose
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical Center, Burlington, Vermont 05405, USA
| | - Nikoletta Sidiropoulos
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical Center, Burlington, Vermont 05405, USA
| | - Devdoot Majumdar
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Jonathan E. Boyson
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| |
Collapse
|
26
|
Zhou G, Li T, Du J, Wu M, Lin D, Pu W, Zhang J, Gu Z. Harnessing HetHydrogel: A Universal Platform to Dropletize Single-Cell Multiomics. SMALL METHODS 2024; 8:e2301631. [PMID: 38419597 DOI: 10.1002/smtd.202301631] [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: 11/25/2023] [Revised: 01/12/2024] [Indexed: 03/02/2024]
Abstract
A universal platform is developed for dropletizing single cell plate-based multiomic assays, consisting of three main pillars: a miniaturized open Heterogeneous Hydrogel reactor (abbreviated HetHydrogel) for multi-step biochemistry, its tunable permeability that allows Tn5 tagmentation, and single cell droplet barcoding. Through optimizing the HetHydrogel manufacturing procedure, the chemical composition, and cell permeation conditions, simultaneous high-throughput mitochondrial DNA genotyping and chromatin profiling at the single-cell level are demonstrated using a mixed-species experiment. This platform offers a powerful way to investigate the genotype-phenotype relationships of various mtDNA mutations in biological processes. The HetHydrogel platform is believed to have the potential to democratize droplet technologies, upgrading a whole range of plate-based single cell assays to high throughput format.
Collapse
Affiliation(s)
- Guoqiang Zhou
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511458, China
| | - Ting Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Jingjing Du
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511458, China
| | - Mengying Wu
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511458, China
| | - Deng Lin
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511458, China
| | - Weilin Pu
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511458, China
| | - Jingwei Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200438, China
- Zhejiang Lab, Hangzhou, 310000, China
| | - Zhenglong Gu
- Center for Mitochondrial Genetics and Health, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511458, China
| |
Collapse
|
27
|
Beebe KD, Eisner JR, Guo J, Shibata Y, Davison JM, Uronis J, Farhangfar C, Farhangfar F, Mooney J, Milburn MV, White RL, Amin A, Milla ME, Foureau DM. The Immunogenomic Landscape of Peripheral High-Dose IL-2 Pharmacodynamics in Patients with Metastatic Renal Cell Carcinoma: A Benchmark for Next-Generation IL-2-Based Immunotherapies. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:29-39. [PMID: 38767437 DOI: 10.4049/jimmunol.2300736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/24/2024] [Indexed: 05/22/2024]
Abstract
High-dose (HD) IL-2 was the first immuno-oncology agent approved for treating advanced renal cell carcinoma and metastatic melanoma, but its use was limited because of substantial toxicities. Multiple next-generation IL-2 agents are being developed to improve tolerability. However, a knowledge gap still exists for the genomic markers that define the target pharmacology for HD IL-2 itself. In this retrospective observational study, we collected PBMC samples from 23 patients with metastatic renal cell carcinoma who were treated with HD IL-2 between 2009 and 2015. We previously reported the results of flow cytometry analyses. In this study, we report the results of our RNA-sequencing immunogenomic survey, which was performed on bulk PBMC samples from immediately before (day 1), during (day 3), and after treatment (day 5) in cycle 1 and/or cycle 2 of the first course of HD IL-2. As part of a detailed analysis of immunogenomic response to HD IL-2 treatment, we analyzed the changes in individual genes and immune gene signatures. By day 3, most lymphoid cell types had transiently decreased, whereas myeloid transcripts increased. Although most genes and/or signatures generally returned to pretreatment expression levels by day 5, certain ones representative of B cell, NK cell, and T cell proliferation and effector functions continued to increase, along with B cell (but not T cell) oligoclonal expansion. Regulatory T cells progressively expanded during and after treatment. They showed strong negative correlation with myeloid effector cells. This detailed RNA-sequencing immunogenomic survey of IL-2 pharmacology complements results of prior flow cytometry analyses. These data provide valuable pharmacological context for assessing PBMC gene expression data from patients dosed with IL-2-related compounds that are currently in development.
Collapse
Affiliation(s)
| | | | - John Guo
- GeneCentric Therapeutics, Inc., Durham, NC
| | | | | | | | | | | | | | | | | | - Asim Amin
- Levine Cancer Institute, Atrium Health, Charlotte, NC
| | | | | |
Collapse
|
28
|
Yared N, Papadopoulou M, Barennes P, Pham HP, Quiniou V, Netzer S, Kaminski H, Burguet L, Demeste A, Colas P, Mora-Charrot L, Rousseau B, Izotte J, Zouine A, Gauthereau X, Vermijlen D, Déchanet-Merville J, Capone M. Long-lived central memory γδ T cells confer protection against murine cytomegalovirus reinfection. PLoS Pathog 2024; 20:e1010785. [PMID: 38976755 PMCID: PMC11257398 DOI: 10.1371/journal.ppat.1010785] [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: 08/04/2022] [Revised: 07/18/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024] Open
Abstract
The involvement of γδ TCR-bearing lymphocytes in immunological memory has gained increasing interest due to their functional duality between adaptive and innate immunity. γδ T effector memory (TEM) and central memory (TCM) subsets have been identified, but their respective roles in memory responses are poorly understood. In the present study, we used subsequent mouse cytomegalovirus (MCMV) infections of αβ T cell deficient mice in order to analyze the memory potential of γδ T cells. As for CMV-specific αβ T cells, MCMV induced the accumulation of cytolytic, KLRG1+CX3CR1+ γδ TEM that principally localized in infected organ vasculature. Typifying T cell memory, γδ T cell expansion in organs and blood was higher after secondary viral challenge than after primary infection. Viral control upon MCMV reinfection was prevented when masking γδ T-cell receptor, and was associated with a preferential amplification of private and unfocused TCR δ chain repertoire composed of a combination of clonotypes expanded post-primary infection and, more unexpectedly, of novel expanded clonotypes. Finally, long-term-primed γδ TCM cells, but not γδ TEM cells, protected T cell-deficient hosts against MCMV-induced death upon adoptive transfer, probably through their ability to survive and to generate TEM in the recipient host. This better survival potential of TCM cells was confirmed by a detailed scRNASeq analysis of the two γδ T cell memory subsets which also revealed their similarity to classically adaptive αβ CD8 T cells. Overall, our study uncovered memory properties of long-lived TCM γδ T cells that confer protection in a chronic infection, highlighting the interest of this T cell subset in vaccination approaches.
Collapse
Affiliation(s)
- Nathalie Yared
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Maria Papadopoulou
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Université Libre de Bruxelles Center for Research in Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | | | | | - Sonia Netzer
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Hanna Kaminski
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Laure Burguet
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Amandine Demeste
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Pacôme Colas
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Lea Mora-Charrot
- Bordeaux University, Service Commun des Animaleries, Bordeaux, France
| | - Benoit Rousseau
- Bordeaux University, Service Commun des Animaleries, Bordeaux, France
| | - Julien Izotte
- Bordeaux University, Service Commun des Animaleries, Bordeaux, France
| | - Atika Zouine
- Bordeaux University, Centre National de la Recherche Scientifique, Institut national de la santé et de la recherche médicale, FACSility, TBM Core, Bordeaux, France
| | - Xavier Gauthereau
- Bordeaux University, Centre National de la Recherche Scientifique, Institut national de la santé et de la recherche médicale, OneCell, RT-PCR and Single Cell Libraries, TBM Core, Bordeaux, France
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Université Libre de Bruxelles Center for Research in Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO department, Walloon ExceLlence Research Institute, Wavre, Belgium
| | - Julie Déchanet-Merville
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Myriam Capone
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| |
Collapse
|
29
|
León-Lara X, Fichtner AS, Willers M, Yang T, Schaper K, Riemann L, Schöning J, Harms A, Almeida V, Schimrock A, Janssen A, Ospina-Quintero L, von Kaisenberg C, Förster R, Eberl M, Richter MF, Pirr S, Viemann D, Ravens S. γδ T cell profiling in a cohort of preterm infants reveals elevated frequencies of CD83+ γδ T cells in sepsis. J Exp Med 2024; 221:e20231987. [PMID: 38753245 PMCID: PMC11098939 DOI: 10.1084/jem.20231987] [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: 10/30/2023] [Revised: 03/06/2024] [Accepted: 04/22/2024] [Indexed: 05/19/2024] Open
Abstract
Preterm infants are at high risk of developing neonatal sepsis. γδ T cells are thought to be an important set of effector cells in neonates. Here, γδ T cells were investigated in a longitudinal cohort of preterm neonates using next-generation sequencing, flow cytometry, and functional assays. During the first year of life, the Vγ9Vδ2 T cell subset showed dynamic phenotypic changes and elevated levels of fetal-derived Vγ9Vδ2 T cells were evident in infants with sepsis. Single-cell transcriptomics identified HLA-DRhiCD83+ γδ T cells in neonatal sepsis, which expressed genes related to antigen presentation. In vitro assays showed that CD83 was expressed on activated Vγ9Vδ2 T cells in preterm and term neonates, but not in adults. In contrast, activation of adult Vγ9Vδ2 T cells enhanced CD86 expression, which was presumably the key receptor to induce CD4 T cell proliferation. Together, we provide a map of the maturation of γδ T cells after preterm birth and highlight their phenotypic diversity in infections.
Collapse
MESH Headings
- Adult
- Female
- Humans
- Infant
- Infant, Newborn
- Male
- Antigens, CD/metabolism
- Antigens, CD/genetics
- CD83 Antigen
- Cohort Studies
- Infant, Premature/immunology
- Lymphocyte Activation/immunology
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/genetics
- Neonatal Sepsis/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
Collapse
Affiliation(s)
- Ximena León-Lara
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | - Maike Willers
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Tao Yang
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | - Lennart Riemann
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Jennifer Schöning
- Translational Pediatrics, Department of Pediatrics, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Anna Harms
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Vicente Almeida
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Anja Schimrock
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Anika Janssen
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | - Constantin von Kaisenberg
- Department of Obstetrics, Gynecology, and Reproductive Medicine, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Matthias Eberl
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | | | - Sabine Pirr
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Dorothee Viemann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Translational Pediatrics, Department of Pediatrics, University Hospital Wuerzburg, Wuerzburg, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- PRIMAL (Priming IMmunity at the Beginning of Life) Consortium, Lübeck, Germany
- Center for Infection Research, University Würzburg, Würzburg, Germany
| | - Sarina Ravens
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
30
|
Piacentini L, Vavassori C, Werba PJ, Saccu C, Spirito R, Colombo GI. Deciphering Abdominal Aortic Diseases Through T-Cell Clonal Repertoire of Perivascular Adipose Tissue. J Am Heart Assoc 2024; 13:e034096. [PMID: 38888318 PMCID: PMC11255777 DOI: 10.1161/jaha.123.034096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/17/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Recent studies suggest that immune-mediated inflammation of perivascular adipose tissue of abdominal aortic aneurysms (AAAs) contributes to disease development and progression. Whether the perivascular adipose tissue of AAA is characterized by a specific adaptive immune signature remains unknown. METHODS AND RESULTS To investigate this hypothesis, we sequenced the T-cell receptor β-chain in the perivascular adipose tissue of patients with AAA and compared it with patients with aortic occlusive disease, who share the former anatomical site of the lesion and risk factors but differ in pathogenic mechanisms. Our results demonstrate that patients with AAA have a lower repertoire diversity than those with aortic occlusive disease and significant differences in variable/joining gene segment usage. Furthermore, we identified a set of 7 public T-cell receptor β-chain clonotypes that distinguished AAA and aortic occlusive disease with very high accuracy. We also found that the T-cell receptor β-chain repertoire differentially characterizes small and large AAAs (aortic diameter<55 mm and ≥55 mm, respectively). CONCLUSIONS This work supports the hypothesis that T cell-mediated immunity is fundamental in AAA pathogenesis and opens up new clinical perspectives.
Collapse
MESH Headings
- Humans
- Aortic Aneurysm, Abdominal/immunology
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/pathology
- Male
- Aged
- Female
- T-Lymphocytes/immunology
- Adipose Tissue/pathology
- Adipose Tissue/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Middle Aged
- Aorta, Abdominal/pathology
- Aorta, Abdominal/immunology
Collapse
Affiliation(s)
- Luca Piacentini
- Bioinformatics and Artificial Intelligence Unit, Centro Cardiologico MonzinoIRCCSMilanItaly
- Immunology and Functional Genomics Unit, Centro Cardiologico MonzinoIRCCSMilanItaly
| | - Chiara Vavassori
- Immunology and Functional Genomics Unit, Centro Cardiologico MonzinoIRCCSMilanItaly
| | - Pablo J. Werba
- Atherosclerosis Prevention Unit, Centro Cardiologico MonzinoIRCCSMilanItaly
| | - Claudio Saccu
- Department of Cardiovascular Surgery of the University of Milan, Centro Cardiologico MonzinoIRCCSMilanItaly
| | - Rita Spirito
- Department of Cardiovascular Surgery of the University of Milan, Centro Cardiologico MonzinoIRCCSMilanItaly
| | - Gualtiero I. Colombo
- Immunology and Functional Genomics Unit, Centro Cardiologico MonzinoIRCCSMilanItaly
| |
Collapse
|
31
|
Simpson J, Starke CE, Ortiz AM, Ransier A, Darko S, Llewellyn-Lacey S, Fennessey CM, Keele BF, Douek DC, Price DA, Brenchley JM. Immunotoxin-mediated depletion of Gag-specific CD8+ T cells undermines natural control of SIV. JCI Insight 2024; 9:e174168. [PMID: 38885329 PMCID: PMC11383179 DOI: 10.1172/jci.insight.174168] [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: 07/25/2023] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
Antibody-mediated depletion studies have demonstrated that CD8+ T cells are required for effective immune control of SIV. However, this approach is potentially confounded by several factors, including reactive CD4+ T cell proliferation, and provides no information on epitope specificity, a likely determinant of CD8+ T cell efficacy. We circumvented these limitations by selectively depleting CD8+ T cells specific for the Gag epitope CTPYDINQM (CM9) via the administration of immunotoxin-conjugated tetrameric complexes of CM9/Mamu-A*01. Immunotoxin administration effectively depleted circulating but not tissue-localized CM9-specific CD8+ T cells, akin to the bulk depletion pattern observed with antibodies directed against CD8. However, we found no evidence to indicate that circulating CM9-specific CD8+ T cells suppressed viral replication in Mamu-A*01+ rhesus macaques during acute or chronic progressive infection with a pathogenic strain of SIV. This observation extended to macaques with established infection during and after continuous antiretroviral therapy. In contrast, natural controller macaques experienced dramatic increases in plasma viremia after immunotoxin administration, highlighting the importance of CD8+ T cell-mediated immunity against CM9. Collectively, these data showed that CM9-specific CD8+ T cells were necessary but not sufficient for robust immune control of SIV in a nonhuman primate model and, more generally, validated an approach that could inform the design of next-generation vaccines against HIV-1.
Collapse
Affiliation(s)
- Jennifer Simpson
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Carly E Starke
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Alexandra M Ortiz
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Amy Ransier
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Sam Darko
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Sian Llewellyn-Lacey
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
| | - Christine M Fennessey
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, NIH, Frederick, Maryland, USA
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, NIH, Frederick, Maryland, USA
| | - Daniel C Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
| | - Jason M Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
32
|
Lundgren S, Myllymäki M, Järvinen T, Keränen MAI, Theodoropoulos J, Smolander J, Kim D, Salmenniemi U, Walldin G, Savola P, Kelkka T, Rajala H, Hellström-Lindberg E, Itälä-Remes M, Kankainen M, Mustjoki S. Somatic mutations associate with clonal expansion of CD8 + T cells. SCIENCE ADVANCES 2024; 10:eadj0787. [PMID: 38848368 PMCID: PMC11160466 DOI: 10.1126/sciadv.adj0787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 05/06/2024] [Indexed: 06/09/2024]
Abstract
Somatic mutations in T cells can cause cancer but also have implications for immunological diseases and cell therapies. The mutation spectrum in nonmalignant T cells is unclear. Here, we examined somatic mutations in CD4+ and CD8+ T cells from 90 patients with hematological and immunological disorders and used T cell receptor (TCR) and single-cell sequencing to link mutations with T cell expansions and phenotypes. CD8+ cells had a higher mutation burden than CD4+ cells. Notably, the biggest variant allele frequency (VAF) of non-synonymous variants was higher than synonymous variants in CD8+ T cells, indicating non-random occurrence. The non-synonymous VAF in CD8+ T cells strongly correlated with the TCR frequency, but not age. We identified mutations in pathways essential for T cell function and often affected lymphoid neoplasia. Single-cell sequencing revealed cytotoxic TEMRA phenotypes of mutated T cells. Our findings suggest that somatic mutations contribute to CD8+ T cell expansions without malignant transformation.
Collapse
Affiliation(s)
- Sofie Lundgren
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Mikko Myllymäki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Timo Järvinen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko A. I. Keränen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Jason Theodoropoulos
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Johannes Smolander
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Daehong Kim
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Urpu Salmenniemi
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Stem Cell Transplantation Unit, Turku University Hospital, Turku, Finland
| | - Gunilla Walldin
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Paula Savola
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Department of Clinical Chemistry, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Tiina Kelkka
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Hanna Rajala
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Eva Hellström-Lindberg
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Maija Itälä-Remes
- Stem Cell Transplantation Unit, Turku University Hospital, Turku, Finland
| | - Matti Kankainen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| |
Collapse
|
33
|
Caduff N, Rieble L, Böni M, McHugh D, Roshan R, Miley W, Labo N, Barman S, Trivett M, Bosma DMT, Rühl J, Goebels N, Whitby D, Münz C. KSHV infection of B cells primes protective T cell responses in humanized mice. Nat Commun 2024; 15:4841. [PMID: 38844783 PMCID: PMC11156630 DOI: 10.1038/s41467-024-49209-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: 07/03/2023] [Accepted: 05/24/2024] [Indexed: 06/09/2024] Open
Abstract
Kaposi sarcoma associated herpesvirus (KSHV) is associated with around 1% of all human tumors, including the B cell malignancy primary effusion lymphoma (PEL), in which co-infection with the Epstein Barr virus (EBV) can almost always be found in malignant cells. Here, we demonstrate that KSHV/EBV co-infection of mice with reconstituted human immune systems (humanized mice) leads to IgM responses against both latent and lytic KSHV antigens, and expansion of central and effector memory CD4+ and CD8+ T cells. Among these, KSHV/EBV dual-infection allows for the priming of CD8+ T cells that are specific for the lytic KSHV antigen K6 and able to kill KSHV/EBV infected B cells. This suggests that K6 may represent a vaccine antigen for the control of KSHV and its associated pathologies in high seroprevalence regions, such as Sub-Saharan Africa.
Collapse
Affiliation(s)
- Nicole Caduff
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
- Genentech Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Lisa Rieble
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Michelle Böni
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Donal McHugh
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
- Pfizer, Medical Department, Schärenmoosstrasse 99, 8052, Zürich, Switzerland
| | - Romin Roshan
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Wendell Miley
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Nazzarena Labo
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Sumanta Barman
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Matthew Trivett
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Douwe M T Bosma
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Julia Rühl
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Norbert Goebels
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Denise Whitby
- Viral Oncology Section, AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
34
|
Guo J, Wang C, Luo N, Wu Y, Huang W, Zhu J, Shi W, Ding J, Ge Y, Liu C, Lu Z, Bast RC, Ai G, Yang W, Wang R, Li C, Chen R, Liu S, Jin H, Zhao B, Cheng Z. IL-2-free tumor-infiltrating lymphocyte therapy with PD-1 blockade demonstrates potent efficacy in advanced gynecologic cancer. BMC Med 2024; 22:207. [PMID: 38769543 PMCID: PMC11106999 DOI: 10.1186/s12916-024-03420-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Tumor-infiltrating lymphocyte (TIL) therapy has been restricted by intensive lymphodepletion and high-dose intravenous interleukin-2 (IL-2) administration. To address these limitations, we conducted preclinical and clinical studies to evaluate the safety, antitumor activity, and pharmacokinetics of an innovative modified regimen in patients with advanced gynecologic cancer. METHODS Patient-derived xenografts (PDX) were established from a local recurrent cervical cancer patient. TILs were expanded ex vivo from minced tumors without feeder cells in the modified TIL therapy regimen. Patients underwent low-dose cyclophosphamide lymphodepletion followed by TIL infusion without intravenous IL-2. The primary endpoint was safety; the secondary endpoints included objective response rate, duration of response, and T cell persistence. RESULTS In matched patient-derived xenografts (PDX) models, homologous TILs efficiently reduced tumor size (p < 0.0001) and underwent IL-2 absence in vivo. In the clinical section, all enrolled patients received TIL infusion using a modified TIL therapy regimen successfully with a manageable safety profile. Five (36%, 95% CI 16.3-61.2) out of 14 evaluable patients experienced objective responses, and three complete responses were ongoing at 19.5, 15.4, and 5.2 months, respectively. Responders had longer overall survival (OS) than non-responders (p = 0.036). Infused TILs showed continuous proliferation and long-term persistence in all patients and showed greater proliferation in responders which was indicated by the Morisita overlap index (MOI) of TCR clonotypes between infused TILs and peripheral T cells on day 14 (p = 0.004) and day 30 (p = 0.004). Higher alteration of the CD8+/CD4+ ratio on day 14 indicated a longer OS (p = 0.010). CONCLUSIONS Our modified TIL therapy regimen demonstrated manageable safety, and TILs could survive and proliferate without IL-2 intravenous administration, showing potent efficacy in patients with advanced gynecologic cancer. TRIAL REGISTRATION NCT04766320, Jan 04, 2021.
Collapse
Affiliation(s)
- Jing Guo
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunyan Wang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ning Luo
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuliang Wu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Huang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jihui Zhu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Weihui Shi
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinye Ding
- Tongji University School of Medicine, Shanghai, China
| | - Yao Ge
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunhong Liu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhen Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert C Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guihai Ai
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Weihong Yang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rui Wang
- Department of Military Health Statistics, Naval Medical University, Shanghai, China
| | - Caixia Li
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rong Chen
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shupeng Liu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Gynecologic Minimally Invasive Surgery Research Center, Tongji University School of Medicine, Shanghai, China
| | - Huajun Jin
- Shanghai Juncell Therapeutics, Shanghai, China
| | - Binghui Zhao
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongping Cheng
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
- Gynecologic Minimally Invasive Surgery Research Center, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
35
|
Sanchez Sanchez G, Emmrich S, Georga M, Papadaki A, Kossida S, Seluanov A, Gorbunova V, Vermijlen D. Invariant γδTCR natural killer-like effector T cells in the naked mole-rat. Nat Commun 2024; 15:4248. [PMID: 38762584 PMCID: PMC11102460 DOI: 10.1038/s41467-024-48652-z] [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: 07/20/2023] [Accepted: 05/03/2024] [Indexed: 05/20/2024] Open
Abstract
The naked mole-rat (Heterocephalus glaber) is a long-lived rodent species showing resistance to the development of cancer. Although naked mole-rats have been reported to lack natural killer (NK) cells, γδ T cell-based immunity has been suggested in this species, which could represent an important arm of the immune system for antitumor responses. Here, we investigate the biology of these unconventional T cells in peripheral tissues (blood, spleen) and thymus of the naked mole-rat at different ages by TCR repertoire profiling and single-cell gene expression analysis. Using our own TCR annotation in the naked mole-rat genome, we report that the γδ TCR repertoire is dominated by a public invariant Vγ4-2/Vδ1-4 TCR, containing the complementary-determining-region-3 (CDR3)γ CTYWDSNYAKKLF / CDR3δ CALWELRTGGITAQLVF that are likely generated by short-homology-repeat-driven DNA rearrangements. This invariant TCR is specifically found in γδ T cells expressing genes associated with NK cytotoxicity and is generated in both the thoracic and cervical thymus of the naked mole-rat until adult life. Our results indicate that invariant Vγ4-2/Vδ1-4 NK-like effector T cells in the naked mole-rat can contribute to tumor immunosurveillance by γδ TCR-mediated recognition of a common molecular signal.
Collapse
MESH Headings
- Animals
- Mole Rats/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Thymus Gland/immunology
- Thymus Gland/cytology
- Killer Cells, Natural/immunology
- Spleen/immunology
- Complementarity Determining Regions/genetics
- Natural Killer T-Cells/immunology
Collapse
Affiliation(s)
- Guillem Sanchez Sanchez
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Stephan Emmrich
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Maria Georga
- IMGT®, the international ImMunoGenetics information system®, Institut de Génétique Humaine (IGH), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier (UM), Montpellier, France
| | - Ariadni Papadaki
- IMGT®, the international ImMunoGenetics information system®, Institut de Génétique Humaine (IGH), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier (UM), Montpellier, France
| | - Sofia Kossida
- IMGT®, the international ImMunoGenetics information system®, Institut de Génétique Humaine (IGH), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier (UM), Montpellier, France
| | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, NY, USA
- Department of Medicine, University of Rochester Medical Center and Medicine, University of Rochester, Rochester, NY, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY, USA
- Department of Medicine, University of Rochester Medical Center and Medicine, University of Rochester, Rochester, NY, USA
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium.
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Gosselies, Belgium.
- ULB Center for Research in Immunology (U-CRI), Université Libre de Bruxelles (ULB), Gosselies, Belgium.
- WELBIO Department, WEL Research Institute, Wavre, Belgium.
| |
Collapse
|
36
|
Peng X, Zhu X, Liu X, Huang Y, Zhu B. Increase in HIV reservoir and T cell immune response after CoronaVac vaccination in people living with HIV. Heliyon 2024; 10:e30394. [PMID: 38720759 PMCID: PMC11076980 DOI: 10.1016/j.heliyon.2024.e30394] [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: 01/16/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction CoronaVac, an inactivated vaccine developed by Sinovac Life Sciences, has been widely used for protection against Coronavirus Disease 2019 (COVID-19). This study investigates its effect on the HIV reservoir and T cell repertoires in people living with HIV (PLWHs). Methods Blood samples were collected from fifteen PLWHs who were administered at least two doses of CoronaVac between April 2021 and February 2022. The levels of cell-associated HIV RNA (CA HIV RNA) and HIV DNA, as well as the T cell receptor (TCR) repertoire profiles, TCR clustering and TCRβ annotation, were studied. Results A significant increase was observed in CA HIV RNA at 2 weeks (431.5 ± 164.2 copies/106 cells, P = 0.039) and 12 weeks (330.2 ± 105.9 copies/106 cells, P = 0.019) after the second dose, when compared to the baseline (0 weeks) (73.6 ± 23.7 copies/106 cells). Various diversity indices of the TCRβ repertoire, including Shannon index, Pielou's evenness index, and Hvj Index, revealed a slight increase (P < 0.05) following CoronaVac vaccination. The proportion of overlapping TCRβ clonotypes increased from baseline (31.9 %) to 2 weeks (32.5 %) and 12 weeks (40.4 %) after the second dose. We also found that the breadth and depth of COVID-19-specific T cells increased from baseline (0.003 and 0.0035) to 12 weeks (0.0066 and 0.0058) post the second dose. Conclusions Our study demonstrated an initial increase in HIV reservoir and TCR repertoire diversity, as well as an expansion in the depth and breadth of COVID-19-specific T-cell clones among CoronaVac-vaccinated PLWHs. These findings provide important insights into the effects of COVID-19 vaccination in PLWHs.
Collapse
Affiliation(s)
- Xiaorong Peng
- The Department of Infectious Diseases, 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, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Xueling Zhu
- The Department of Infectious Diseases, 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, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Xiang Liu
- The Department of Infectious Diseases, 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, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Ying Huang
- The Department of Infectious Diseases, 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, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Biao Zhu
- The Department of Infectious Diseases, 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, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| |
Collapse
|
37
|
Baker AM, Nageswaran G, Nenclares P, Ronel T, Smith K, Kimberley C, Laclé MM, Bhide S, Harrington KJ, Melcher A, Rodriguez-Justo M, Chain B, Graham TA. FUME-TCRseq Enables Sensitive and Accurate Sequencing of the T-cell Receptor from Limited Input of Degraded RNA. Cancer Res 2024; 84:1560-1569. [PMID: 38479434 PMCID: PMC11094417 DOI: 10.1158/0008-5472.can-23-3340] [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: 10/24/2023] [Revised: 01/19/2024] [Accepted: 02/27/2024] [Indexed: 05/16/2024]
Abstract
Genomic analysis of the T-cell receptor (TCR) reveals the strength, breadth, and clonal dynamics of the adaptive immune response to pathogens or cancer. The diversity of the TCR repertoire, however, means that sequencing is technically challenging, particularly for samples with low-quality, degraded nucleic acids. Here, we developed and validated FUME-TCRseq, a robust and sensitive RNA-based TCR sequencing methodology that is suitable for formalin-fixed paraffin-embedded samples and low amounts of input material. FUME-TCRseq incorporates unique molecular identifiers into each molecule of cDNA, allowing correction for sequencing errors and PCR bias. Using RNA extracted from colorectal and head and neck cancers to benchmark the accuracy and sensitivity of FUME-TCRseq against existing methods demonstrated excellent concordance between the datasets. Furthermore, FUME-TCRseq detected more clonotypes than a commercial RNA-based alternative, with shorter library preparation time and significantly lower cost. The high sensitivity and the ability to sequence RNA of poor quality and limited amount enabled quantitative analysis of small numbers of cells from archival tissue sections, which is not possible with other methods. Spatially resolved FUME-TCRseq analysis of colorectal cancers using macrodissected archival samples revealed the shifting T-cell landscapes at the transition to an invasive phenotype and between tumor subclones containing distinct driver alterations. In summary, FUME-TCRseq represents an accurate, sensitive, and low-cost tool for the characterization of T-cell repertoires, particularly in samples with low-quality RNA that have not been accessible using existing methodology. SIGNIFICANCE FUME-TCRseq is a TCR sequencing methodology that supports sensitive and spatially resolved detection of TCR clones in archival clinical specimens, which can facilitate longitudinal tracking of immune responses through disease course and treatment.
Collapse
Affiliation(s)
- Ann-Marie Baker
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Gayathri Nageswaran
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Pablo Nenclares
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, United Kingdom
| | - Tahel Ronel
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Kane Smith
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Christopher Kimberley
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Miangela M. Laclé
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Shreerang Bhide
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, United Kingdom
- Head and Neck Unit, The Royal Marsden Hospital NHS Trust, London, United Kingdom
| | - Kevin J. Harrington
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, United Kingdom
| | - Alan Melcher
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, United Kingdom
- Division of Breast Cancer Research, Institute of Cancer Research, London, United Kingdom
| | | | - Benny Chain
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Trevor A. Graham
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
38
|
Gupta T, Antanaviciute A, Hyun-Jung Lee C, Ottakandathil Babu R, Aulicino A, Christoforidou Z, Siejka-Zielinska P, O'Brien-Ball C, Chen H, Fawkner-Corbett D, Geros AS, Bridges E, McGregor C, Cianci N, Fryer E, Alham NK, Jagielowicz M, Santos AM, Fellermeyer M, Davis SJ, Parikh K, Cheung V, Al-Hillawi L, Sasson S, Slevin S, Brain O, Fernandes RA, Koohy H, Simmons A. Tracking in situ checkpoint inhibitor-bound target T cells in patients with checkpoint-induced colitis. Cancer Cell 2024; 42:797-814.e15. [PMID: 38744246 DOI: 10.1016/j.ccell.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 02/09/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
The success of checkpoint inhibitors (CPIs) for cancer has been tempered by immune-related adverse effects including colitis. CPI-induced colitis is hallmarked by expansion of resident mucosal IFNγ cytotoxic CD8+ T cells, but how these arise is unclear. Here, we track CPI-bound T cells in intestinal tissue using multimodal single-cell and subcellular spatial transcriptomics (ST). Target occupancy was increased in inflamed tissue, with drug-bound T cells located in distinct microdomains distinguished by specific intercellular signaling and transcriptional gradients. CPI-bound cells were largely CD4+ T cells, including enrichment in CPI-bound peripheral helper, follicular helper, and regulatory T cells. IFNγ CD8+ T cells emerged from both tissue-resident memory (TRM) and peripheral populations, displayed more restricted target occupancy profiles, and co-localized with damaged epithelial microdomains lacking effective regulatory cues. Our multimodal analysis identifies causal pathways and constitutes a resource to inform novel preventive strategies.
Collapse
Affiliation(s)
- Tarun Gupta
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Agne Antanaviciute
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; MRC WIMM Centre For Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK.
| | - Chloe Hyun-Jung Lee
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; MRC WIMM Centre For Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Rosana Ottakandathil Babu
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; MRC WIMM Centre For Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Anna Aulicino
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Zoe Christoforidou
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Paulina Siejka-Zielinska
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Caitlin O'Brien-Ball
- Chinese Academy of Medical Sciences (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7BN, UK
| | - Hannah Chen
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - David Fawkner-Corbett
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Academic Paediatric Surgery Unit (APSU), Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Ana Sousa Geros
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Esther Bridges
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Colleen McGregor
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Nicole Cianci
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Eve Fryer
- Pathology, Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Nasullah Khalid Alham
- Nuffield Department of Surgical Sciences and Oxford NIHR Biomedical Research Centre (BRC), University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Marta Jagielowicz
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Ana Mafalda Santos
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Martin Fellermeyer
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Simon J Davis
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Kaushal Parikh
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| | - Vincent Cheung
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Lulia Al-Hillawi
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Sarah Sasson
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Stephanie Slevin
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Oliver Brain
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Ricardo A Fernandes
- Chinese Academy of Medical Sciences (CAMS) Oxford Institute (COI), University of Oxford, Oxford OX3 7BN, UK
| | - Hashem Koohy
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; MRC WIMM Centre For Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK.
| | - Alison Simmons
- Medical Research Council (MRC) Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK; Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford OX3 9DU, UK.
| |
Collapse
|
39
|
Upadhye A, Meza Landeros KE, Ramírez-Suástegui C, Schmiedel BJ, Woo E, Chee SJ, Malicki D, Coufal NG, Gonda D, Levy ML, Greenbaum JA, Seumois G, Crawford J, Roberts WD, Schoenberger SP, Cheroutre H, Ottensmeier CH, Vijayanand P, Ganesan AP. Intra-tumoral T cells in pediatric brain tumors display clonal expansion and effector properties. NATURE CANCER 2024; 5:791-807. [PMID: 38228835 DOI: 10.1038/s43018-023-00706-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 12/11/2023] [Indexed: 01/18/2024]
Abstract
Brain tumors in children are a devastating disease in a high proportion of patients. Owing to inconsistent results in clinical trials in unstratified patients, the role of immunotherapy remains unclear. We performed an in-depth survey of the single-cell transcriptomes and clonal relationship of intra-tumoral T cells from children with brain tumors. Our results demonstrate that a large fraction of T cells in the tumor tissue are clonally expanded with the potential to recognize tumor antigens. Such clonally expanded T cells display enrichment of transcripts linked to effector function, tissue residency, immune checkpoints and signatures of neoantigen-specific T cells and immunotherapy response. We identify neoantigens in pediatric brain tumors and show that neoantigen-specific T cell gene signatures are linked to better survival outcomes. Notably, among the patients in our cohort, we observe substantial heterogeneity in the degree of clonal expansion and magnitude of T cell response. Our findings suggest that characterization of intra-tumoral T cell responses may enable selection of patients for immunotherapy, an approach that requires prospective validation in clinical trials.
Collapse
Affiliation(s)
- Aditi Upadhye
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Kevin E Meza Landeros
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Center for Genomic Sciences, National Autonomous University of Mexico, Cuernavaca, Mexico
| | | | | | - Edwin Woo
- Southampton University Hospitals NHS Trust, Southampton, UK
| | - Serena J Chee
- Department of Respiratory Medicine, Liverpool Heart and Chest Hospital NHS Foundation Trust, Liverpool, UK
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Denise Malicki
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Rady Children's Hospital, San Diego, CA, USA
| | - Nicole G Coufal
- Rady Children's Hospital, San Diego, CA, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - David Gonda
- Rady Children's Hospital, San Diego, CA, USA
- Department of Neurological Surgery, University of California San Diego, La Jolla, CA, USA
| | - Michael L Levy
- Rady Children's Hospital, San Diego, CA, USA
- Department of Neurological Surgery, University of California San Diego, La Jolla, CA, USA
| | | | | | - John Crawford
- Rady Children's Hospital, San Diego, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
- Children's Hospital Orange County, Irvine, CA, USA
| | - William D Roberts
- Rady Children's Hospital, San Diego, CA, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | | | | | - Christian H Ottensmeier
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
- Clatterbridge Cancer Center NHS Foundation Trust, Liverpool, UK
| | - Pandurangan Vijayanand
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Anusha-Preethi Ganesan
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Rady Children's Hospital, San Diego, CA, USA.
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
40
|
Chen PL, Hung SI, Chung WH, Chen CB, Kuo CN, Lin YK, Chiu CY. T-cell receptor diversity and allergen sensitivity in childhood asthma and atopic dermatitis. Pediatr Allergy Immunol 2024; 35:e14143. [PMID: 38745384 DOI: 10.1111/pai.14143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Childhood allergies of asthma and atopic dermatitis (AD) involve an overactive T-cell immune response triggered by allergens. However, the impact of T-cell receptor (TCR) repertoires on allergen sensitization and their role in mediating different phenotypes of asthma and AD in early childhood remains unclear. METHODS A total of 78 children, comprising 26 with asthma alone, 26 with AD alone, and 26 healthy controls (HC), were enrolled. TCR repertoire profiles were determined using a unique molecular identifier system for next-generation sequencing. Integrative analyses of their associations with allergen-specific IgE levels and allergies were performed. RESULTS The diversity in TCR alpha variable region (TRAV) genes of TCR repertoires and complementarity determining region 3 (CDR3) clonality in TRAV/TRBV (beta) genes were significantly higher in children with AD compared with those with asthma and HC (p < .05). Compared with HC, the expression of TRAV13-1 and TRAV4 genes was significantly higher in both asthma and AD (p < .05), with a significant positive correlation with mite-specific IgE levels (p < .01). In contrast, TRBV7-9 gene expression was significantly lower in both asthma and AD (p < .01), with this gene showing a significant negative correlation with mite-specific IgE levels (p < .01). Furthermore, significantly higher TRAV8-3 gene expression, positively correlated with food-specific IgE levels, was found in children with AD compared with those with asthma (p < .05). CONCLUSION Integrated TCR repertoires analysis provides clinical insights into the diverse TCR genes linked to antigen specificity, offering potential for precision immunotherapy in childhood allergies.
Collapse
Affiliation(s)
- Pei-Ling Chen
- Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, and Chang Gung University, Taoyuan, Taiwan
| | - Shuen-Iu Hung
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Hung Chung
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei and Keelung, Taiwan
| | - Chun-Bing Chen
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei and Keelung, Taiwan
| | - Chieh-Ni Kuo
- Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, and Chang Gung University, Taoyuan, Taiwan
| | - Yin-Ku Lin
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Keelung, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Yung Chiu
- Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, and Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
41
|
Broquet A, Gourain V, Goronflot T, Le Mabecque V, Sinha D, Ashayeripanah M, Jacqueline C, Martin P, Davieau M, Boutin L, Poulain C, Martin FP, Fourgeux C, Petrier M, Cannevet M, Leclercq T, Guillonneau M, Chaumette T, Laurent T, Harly C, Scotet E, Legentil L, Ferrières V, Corgnac S, Mami-Chouaib F, Mosnier JF, Mauduit N, McWilliam HEG, Villadangos JA, Gourraud PA, Asehnoune K, Poschmann J, Roquilly A. Sepsis-trained macrophages promote antitumoral tissue-resident T cells. Nat Immunol 2024; 25:802-819. [PMID: 38684922 DOI: 10.1038/s41590-024-01819-8] [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: 02/09/2023] [Accepted: 03/14/2024] [Indexed: 05/02/2024]
Abstract
Sepsis induces immune alterations, which last for months after the resolution of illness. The effect of this immunological reprogramming on the risk of developing cancer is unclear. Here we use a national claims database to show that sepsis survivors had a lower cumulative incidence of cancers than matched nonsevere infection survivors. We identify a chemokine network released from sepsis-trained resident macrophages that triggers tissue residency of T cells via CCR2 and CXCR6 stimulations as the immune mechanism responsible for this decreased risk of de novo tumor development after sepsis cure. While nonseptic inflammation does not provoke this network, laminarin injection could therapeutically reproduce the protective sepsis effect. This chemokine network and CXCR6 tissue-resident T cell accumulation were detected in humans with sepsis and were associated with prolonged survival in humans with cancer. These findings identify a therapeutically relevant antitumor consequence of sepsis-induced trained immunity.
Collapse
Affiliation(s)
- Alexis Broquet
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| | - Victor Gourain
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
| | - Thomas Goronflot
- CHU Nantes, Pôle Hospitalo-Universitaire 11: Santé Publique, Clinique des Données, INSERM, Nantes Université, CIC 1413, Nantes, France
| | - Virginie Le Mabecque
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
| | - Debajyoti Sinha
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
| | - Mitra Ashayeripanah
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Cédric Jacqueline
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
| | - Pierre Martin
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
| | - Marion Davieau
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| | - Lea Boutin
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
| | - Cecile Poulain
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| | - Florian P Martin
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| | - Cynthia Fourgeux
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
| | - Melanie Petrier
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
| | - Manon Cannevet
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| | - Thomas Leclercq
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
| | - Maeva Guillonneau
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
- Olgram SAS, Bréhan, France
| | - Tanguy Chaumette
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
| | - Thomas Laurent
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
| | | | | | - Laurent Legentil
- Ecole Nationale Supérieure de Chimie de Rennes, Université de Rennes, ISCR - UMR CNRS 6226, Rennes, France
| | - Vincent Ferrières
- Ecole Nationale Supérieure de Chimie de Rennes, Université de Rennes, ISCR - UMR CNRS 6226, Rennes, France
| | - Stephanie Corgnac
- INSERM UMR 1186, Integrative Tumour Immunology and Immunotherapy, Gustave Roussy, Faculty de Médecine, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumour Immunology and Immunotherapy, Gustave Roussy, Faculty de Médecine, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | | | | | - Hamish E G McWilliam
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jose A Villadangos
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Pierre Antoine Gourraud
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
- CHU Nantes, Pôle Hospitalo-Universitaire 11: Santé Publique, Clinique des Données, INSERM, Nantes Université, CIC 1413, Nantes, France
| | - Karim Asehnoune
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France
| | - Jeremie Poschmann
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France.
| | - Antoine Roquilly
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology UMR 1064, Nantes, France.
- CHU Nantes, INSERM, Nantes Université, Anesthesie Reanimation, CIC 1413, Nantes, France.
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
42
|
Irac SE, Soon MSF, Borcherding N, Tuong ZK. Single-cell immune repertoire analysis. Nat Methods 2024; 21:777-792. [PMID: 38637691 DOI: 10.1038/s41592-024-02243-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/12/2024] [Indexed: 04/20/2024]
Abstract
Single-cell T cell and B cell antigen receptor-sequencing data analysis can potentially perform in-depth assessments of adaptive immune cells that inform on understanding immune cell development to tracking clonal expansion in disease and therapy. However, it has been extremely challenging to analyze and interpret T cells and B cells and their adaptive immune receptor repertoires at the single-cell level due to not only the complexity of the data but also the underlying biology. In this Review, we delve into the computational breakthroughs that have transformed the analysis of single-cell T cell and B cell antigen receptor-sequencing data.
Collapse
Affiliation(s)
- Sergio E Irac
- Cancer Immunoregulation and Immunotherapy, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Megan Sioe Fei Soon
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Omniscope, Palo Alto, CA, USA
| | - Zewen Kelvin Tuong
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
43
|
Peng H, Wu X, Cui X, Liu S, Liang Y, Cai X, Shi M, Zhong R, Li C, Liu J, Wu D, Gao Z, Lu X, Luo H, He J, Liang W. Molecular and immune characterization of Chinese early-stage non-squamous non-small cell lung cancer: a multi-omics cohort study. Transl Lung Cancer Res 2024; 13:763-784. [PMID: 38736486 PMCID: PMC11082711 DOI: 10.21037/tlcr-23-800] [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: 12/02/2023] [Accepted: 03/15/2024] [Indexed: 05/14/2024]
Abstract
Background Albeit considered with superior survival, around 30% of the early-stage non-squamous non-small cell lung cancer (Ns-NSCLC) patients relapse within 5 years, suggesting unique biology. However, the biological characteristics of early-stage Ns-NSCLC, especially in the Chinese population, are still unclear. Methods Multi-omics interrogation of early-stage Ns-NSCLC (stage I-III), paired blood samples and normal lung tissues (n=76) by whole-exome sequencing (WES), RNA sequencing, and T-cell receptor (TCR) sequencing were conducted. Results An average of 128 exonic mutations were identified, and the most frequently mutant gene was EGFR (55%), followed by TP53 (37%) and TTN (26%). Mutations in MUC17, ABCA2, PDE4DIP, and MYO18B predicted significantly unfavorable disease-free survival (DFS). Moreover, cytobands amplifications in 8q24.3, 14q13.1, 14q11.2, and deletion in 3p21.1 were highlighted in recurrent cases. Higher incidence of human leukocyte antigen loss of heterozygosity (HLA-LOH), higher tumor mutational burden (TMB) and tumor neoantigen burden (TNB) were identified in ever-smokers than never-smokers. HLA-LOH also correlated with higher TMB, TNB, intratumoral heterogeneity (ITH), and whole chromosomal instability (wCIN) scores. Interestingly, higher ITH was an independent predictor of better DFS in early-stage Ns-NSCLC. Up-regulation of immune-related genes, including CRABP2, ULBP2, IL31RA, and IL1A, independently portended a dismal prognosis. Enhanced TCR diversity of peripheral blood mononuclear cells (PBMCs) predicted better prognosis, indicative of a noninvasive method for relapse surveillance. Eventually, seven machine-learning (ML) algorithms were employed to evaluate the predictive accuracy of clinical, genomic, transcriptomic, and TCR repertoire data on DFS, showing that clinical and RNA features combination in the random forest (RF) algorithm, with area under the curve (AUC) of 97.5% and 83.3% in the training and testing cohort, respectively, significantly outperformed other methods. Conclusions This study comprehensively profiled the genomic, transcriptomic, and TCR repertoire spectrums of Chinese early-stage Ns-NSCLC, shedding light on biological underpinnings and candidate biomarkers for prognosis development.
Collapse
Affiliation(s)
- Haoxin Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, Nanshan School, Guangzhou Medical University, Guangzhou, China
| | - Xiangrong Wu
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, Nanshan School, Guangzhou Medical University, Guangzhou, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoli Cui
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd., Shenzhen, China
| | - Shaopeng Liu
- Department of Computer Science, Guangdong Polytechnic Normal University, Guangzhou, China
- Department of Artificial Intelligence Research, Pazhou Lab, Guangzhou, China
| | - Yueting Liang
- Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiuyu Cai
- Department of General Internal Medicine, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Cener for Cancer Medicine, Guangzhou, China
| | - Mengping Shi
- Department of Computer Science, Guangdong Polytechnic Normal University, Guangzhou, China
| | - Ran Zhong
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Caichen Li
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jun Liu
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dongfang Wu
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd., Shenzhen, China
| | - Zhibo Gao
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd., Shenzhen, China
| | - Xu Lu
- Department of Computer Science, Guangdong Polytechnic Normal University, Guangzhou, China
- Department of Artificial Intelligence Research, Pazhou Lab, Guangzhou, China
| | - Haitao Luo
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd., Shenzhen, China
| | - Jianxing He
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenhua Liang
- Department of Thoracic Oncology and Surgery, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Medical Oncology, The First People’s Hospital of Zhaoqing, Zhaoqing, China
| |
Collapse
|
44
|
Li Z, Liu J, Zhang B, Yue J, Shi X, Cui K, Liu Z, Chang Z, Sun Z, Li M, Yang Y, Ma Z, Li L, Zhang C, Sun P, Zhong J, Zhao L. Neoadjuvant tislelizumab plus stereotactic body radiotherapy and adjuvant tislelizumab in early-stage resectable hepatocellular carcinoma: the Notable-HCC phase 1b trial. Nat Commun 2024; 15:3260. [PMID: 38627377 PMCID: PMC11021407 DOI: 10.1038/s41467-024-47420-3] [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: 12/29/2023] [Accepted: 03/29/2024] [Indexed: 04/19/2024] Open
Abstract
Notable-HCC (NCT05185531) is a phase 1b trial, aiming to evaluate the safety and preliminary effectiveness of neoadjuvant PD-1 blockade plus stereotactic body radiotherapy (SBRT) in early-stage resectable hepatocellular carcinoma (HCC). Twenty patients with HCC of BCLC stage 0-A received 3 × Gy SBRT and two cycles of tislelizumab, an anti-PD-1 monoclonal antibody before the curative HCC resection. Primary endpoints were the surgery delay, radiographic and pathological tumor response after the neoadjuvant therapy, safety and tolerability. During the neoadjuvant therapy, treatment-related adverse events (TRAEs) of grade 1-2 occurred in all 20 patients (100%), eight patients (40%) had grade 3 TRAEs, no grade 4 to 5 TRAE occurred, and all resolved without corticosteroids treatment. Per mRECIST, the objective response rate was 63.2% (12/19), with 3 complete response; the disease control rate was 100%. Two (10.5%) patients achieved complete pathological response. No surgery delay occurred. The neoadjuvant therapy did not increase the surgical difficulty or the incidence of complications. Secondary endpoints of disease-free survival and overall survival were not mature at the time of the analysis. Our pilot trial shows that neoadjuvant therapy with anti-PD-1 + SBRT is safe and promotes tumor responses in early-stage resectable HCC.
Collapse
Affiliation(s)
- Zhongchao Li
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
| | - Jing Liu
- Department of Abdominal Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
| | - Bo Zhang
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
| | - Jinbo Yue
- Department of Abdominal Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
| | - Xuetao Shi
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
| | - Kai Cui
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
| | - Zhaogang Liu
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
| | - Zhibin Chang
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, 6699 Qingdao Road, Huaiyin District, Jinan, China
| | - Zhicheng Sun
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, 6699 Qingdao Road, Huaiyin District, Jinan, China
| | - Mingming Li
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, 6699 Qingdao Road, Huaiyin District, Jinan, China
| | - Yue Yang
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, 6699 Qingdao Road, Huaiyin District, Jinan, China
| | - Zhao Ma
- The Fourth People's Hospital of Jinan, Jinan, China
| | - Lei Li
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
| | - Chengsheng Zhang
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
| | - Pengfei Sun
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
| | - Jingtao Zhong
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China
| | - Lei Zhao
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong First Medical University, 440 Jiyan Road, Huaiyin District, Jinan, China.
- Shandong First Medical University and Shandong Academy of Medical Sciences, 6699 Qingdao Road, Huaiyin District, Jinan, China.
- The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
45
|
Barnkob MB, Michaels YS, André V, Macklin PS, Gileadi U, Valvo S, Rei M, Kulicke C, Chen JL, Jain V, Woodcock VK, Colin-York H, Hadjinicolaou AV, Kong Y, Mayya V, Mazet JM, Mead GJ, Bull JA, Rijal P, Pugh CW, Townsend AR, Gérard A, Olsen LR, Fritzsche M, Fulga TA, Dustin ML, Jones EY, Cerundolo V. Semmaphorin 3 A causes immune suppression by inducing cytoskeletal paralysis in tumour-specific CD8 + T cells. Nat Commun 2024; 15:3173. [PMID: 38609390 PMCID: PMC11017241 DOI: 10.1038/s41467-024-47424-z] [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: 11/02/2021] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Semaphorin-3A (SEMA3A) functions as a chemorepulsive signal during development and can affect T cells by altering their filamentous actin (F-actin) cytoskeleton. The exact extent of these effects on tumour-specific T cells are not completely understood. Here we demonstrate that Neuropilin-1 (NRP1) and Plexin-A1 and Plexin-A4 are upregulated on stimulated CD8+ T cells, allowing tumour-derived SEMA3A to inhibit T cell migration and assembly of the immunological synapse. Deletion of NRP1 in both CD4+ and CD8+ T cells enhance CD8+ T-cell infiltration into tumours and restricted tumour growth in animal models. Conversely, over-expression of SEMA3A inhibit CD8+ T-cell infiltration. We further show that SEMA3A affects CD8+ T cell F-actin, leading to inhibition of immune synapse formation and motility. Examining a clear cell renal cell carcinoma patient cohort, we find that SEMA3A expression is associated with reduced survival, and that T-cells appear trapped in SEMA3A rich regions. Our study establishes SEMA3A as an inhibitor of effector CD8+ T cell tumour infiltration, suggesting that blocking NRP1 could improve T cell function in tumours.
Collapse
Affiliation(s)
- Mike B Barnkob
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK.
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Department of Clinical Immunology, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| | - Yale S Michaels
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
- Paul Albrechtsen Research Institute, CancerCare Manitoba, 675 Mcdermot Ave, Winnipeg, MB, R3E 0V9, Canada
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Bannatyne Ave, Winnipeg, MB, R3E 3N4, Canada
| | - Violaine André
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
| | - Philip S Macklin
- Nuffield Department of Medicine, University of Oxford, Nuffield Department of Medicine Research Building, Roosevelt Drive, Oxford, OX3 7FZ, UK
| | - Uzi Gileadi
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
| | - Salvatore Valvo
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Dr, Oxford, OX3 7FY, UK
| | - Margarida Rei
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Corinna Kulicke
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
- Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, OR, US
| | - Ji-Li Chen
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
| | - Vitul Jain
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Victoria K Woodcock
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
| | - Huw Colin-York
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
| | - Andreas V Hadjinicolaou
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
- Division of Gastroenterology & Hepatology, Department of Medicine, Cambridge University Hospitals, University of Cambridge, Cambridge, England
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, England
| | - Youxin Kong
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Viveka Mayya
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Dr, Oxford, OX3 7FY, UK
| | - Julie M Mazet
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Dr, Oxford, OX3 7FY, UK
| | - Gracie-Jennah Mead
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Dr, Oxford, OX3 7FY, UK
| | - Joshua A Bull
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
| | - Pramila Rijal
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
| | - Christopher W Pugh
- Nuffield Department of Medicine, University of Oxford, Nuffield Department of Medicine Research Building, Roosevelt Drive, Oxford, OX3 7FZ, UK
| | - Alain R Townsend
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
| | - Audrey Gérard
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Dr, Oxford, OX3 7FY, UK
| | - Lars R Olsen
- Department of Health Technology, Technical University of Denmark, Ørsteds Plads, Building 345C, 2800 Kgs, Lyngby, Denmark
| | - Marco Fritzsche
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Dr, Oxford, OX3 7FY, UK
| | - Tudor A Fulga
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Dr, Oxford, OX3 7FY, UK
| | - E Yvonne Jones
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK.
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford, OX3 9DS, UK
| |
Collapse
|
46
|
Nakonechnaya TO, Moltedo B, Putintseva EV, Leyn S, Bolotin DA, Britanova OV, Shugay M, Chudakov DM. Convergence, plasticity, and tissue residence of regulatory T cell response via TCR repertoire prism. eLife 2024; 12:RP89382. [PMID: 38591522 PMCID: PMC11003740 DOI: 10.7554/elife.89382] [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] [Indexed: 04/10/2024] Open
Abstract
Suppressive function of regulatory T cells (Treg) is dependent on signaling of their antigen receptors triggered by cognate self, dietary, or microbial peptides presented on MHC II. However, it remains largely unknown whether distinct or shared repertoires of Treg TCRs are mobilized in response to different challenges in the same tissue or the same challenge in different tissues. Here we use a fixed TCRβ chain FoxP3-GFP mouse model to analyze conventional (eCD4) and regulatory (eTreg) effector TCRα repertoires in response to six distinct antigenic challenges to the lung and skin. This model shows highly 'digital' repertoire behavior with easy-to-track challenge-specific TCRα CDR3 clusters. For both eCD4 and eTreg subsets, we observe challenge-specific clonal expansions yielding homologous TCRα clusters within and across animals and exposure sites, which are also reflected in the draining lymph nodes but not systemically. Some CDR3 clusters are shared across cancer challenges, suggesting a response to common tumor-associated antigens. For most challenges, eCD4 and eTreg clonal response does not overlap. Such overlap is exclusively observed at the sites of certain tumor challenges, and not systematically, suggesting transient and local tumor-induced eCD4=>eTreg plasticity. This transition includes a dominant tumor-responding eCD4 CDR3 motif, as well as characteristic iNKT TCRα CDR3. In addition, we examine the homeostatic tissue residency of clonal eTreg populations by excluding the site of challenge from our analysis. We demonstrate that distinct CDR3 motifs are characteristic of eTreg cells residing in particular lymphatic tissues, regardless of the challenge. This observation reveals the tissue-resident, antigen-specific clonal Treg populations.
Collapse
Affiliation(s)
- Tatyana O Nakonechnaya
- Institute of Translational Medicine, Pirogov Russian National Research Medical UniversityMoscowRussian Federation
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
| | - Bruno Moltedo
- Howard Hughes Medical Institute and Immunology Program, Sloan Kettering Institute and Ludwig Center at Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Ekaterina V Putintseva
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
| | - Sofya Leyn
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
| | - Dmitry A Bolotin
- Institute of Translational Medicine, Pirogov Russian National Research Medical UniversityMoscowRussian Federation
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
| | - Olga V Britanova
- Institute of Translational Medicine, Pirogov Russian National Research Medical UniversityMoscowRussian Federation
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
| | - Mikhail Shugay
- Institute of Translational Medicine, Pirogov Russian National Research Medical UniversityMoscowRussian Federation
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
| | - Dmitriy M Chudakov
- Institute of Translational Medicine, Pirogov Russian National Research Medical UniversityMoscowRussian Federation
- Genomics of Adaptive Immunity Department, Shemyakin and Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
- Central European Institute of TechnologyBrnoCzech Republic
- Abu Dhabi Stem Cells CenterAbu DhabiUnited Arab Emirates
| |
Collapse
|
47
|
Lauss M, Phung B, Borch TH, Harbst K, Kaminska K, Ebbesson A, Hedenfalk I, Yuan J, Nielsen K, Ingvar C, Carneiro A, Isaksson K, Pietras K, Svane IM, Donia M, Jönsson G. Molecular patterns of resistance to immune checkpoint blockade in melanoma. Nat Commun 2024; 15:3075. [PMID: 38594286 PMCID: PMC11004175 DOI: 10.1038/s41467-024-47425-y] [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: 07/27/2023] [Accepted: 04/02/2024] [Indexed: 04/11/2024] Open
Abstract
Immune checkpoint blockade (ICB) has improved outcome for patients with metastatic melanoma but not all benefit from treatment. Several immune- and tumor intrinsic features are associated with clinical response at baseline. However, we need to further understand the molecular changes occurring during development of ICB resistance. Here, we collect biopsies from a cohort of 44 patients with melanoma after progression on anti-CTLA4 or anti-PD1 monotherapy. Genetic alterations of antigen presentation and interferon gamma signaling pathways are observed in approximately 25% of ICB resistant cases. Anti-CTLA4 resistant lesions have a sustained immune response, including immune-regulatory features, as suggested by multiplex spatial and T cell receptor (TCR) clonality analyses. One anti-PD1 resistant lesion harbors a distinct immune cell niche, however, anti-PD1 resistant tumors are generally immune poor with non-expanded TCR clones. Such immune poor microenvironments are associated with melanoma cells having a de-differentiated phenotype lacking expression of MHC-I molecules. In addition, anti-PD1 resistant tumors have reduced fractions of PD1+ CD8+ T cells as compared to ICB naïve metastases. Collectively, these data show the complexity of ICB resistance and highlight differences between anti-CTLA4 and anti-PD1 resistance that may underlie differential clinical outcomes of therapy sequence and combination.
Collapse
Affiliation(s)
- Martin Lauss
- Division of Oncology, Department of Clinical Sciences, Faculty of Medicine, Lund University, 22185, Lund, Sweden
- Lund University Cancer Center, LUCC, Lund, Sweden
| | - Bengt Phung
- Division of Oncology, Department of Clinical Sciences, Faculty of Medicine, Lund University, 22185, Lund, Sweden
- Lund University Cancer Center, LUCC, Lund, Sweden
| | - Troels Holz Borch
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Katja Harbst
- Division of Oncology, Department of Clinical Sciences, Faculty of Medicine, Lund University, 22185, Lund, Sweden
- Lund University Cancer Center, LUCC, Lund, Sweden
| | - Kamila Kaminska
- Division of Oncology, Department of Clinical Sciences, Faculty of Medicine, Lund University, 22185, Lund, Sweden
- Lund University Cancer Center, LUCC, Lund, Sweden
| | - Anna Ebbesson
- Division of Oncology, Department of Clinical Sciences, Faculty of Medicine, Lund University, 22185, Lund, Sweden
- Lund University Cancer Center, LUCC, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology, Department of Clinical Sciences, Faculty of Medicine, Lund University, 22185, Lund, Sweden
- Lund University Cancer Center, LUCC, Lund, Sweden
| | - Joan Yuan
- Division of Molecular Hematology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, 22185, Lund, Sweden
| | - Kari Nielsen
- Lund University Cancer Center, LUCC, Lund, Sweden
- Division of Dermatology, Skåne University Hospital and Department of Clinical Sciences, Faculty of Medicine, Lund University, 22185, Lund, Sweden
| | - Christian Ingvar
- Division of Surgery, Department of Clinical Sciences, Faculty of Medicine, Lund University, 22185, Lund, Sweden
| | - Ana Carneiro
- Division of Oncology, Department of Clinical Sciences, Faculty of Medicine, Lund University, 22185, Lund, Sweden
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital Comprehensive Cancer Center, 22185, Lund, Sweden
| | - Karolin Isaksson
- Lund University Cancer Center, LUCC, Lund, Sweden
- Division of Surgery, Department of Clinical Sciences, Faculty of Medicine, Lund University, 22185, Lund, Sweden
- Department of Surgery, Kristianstad Hospital, 29133, Kristianstad, Sweden
| | - Kristian Pietras
- Lund University Cancer Center, LUCC, Lund, Sweden
- Division of Translational Cancer Research, Department of Laboratory Medicine, Faculty of Medicine, Lund University, 22185, Lund, Sweden
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Marco Donia
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Göran Jönsson
- Division of Oncology, Department of Clinical Sciences, Faculty of Medicine, Lund University, 22185, Lund, Sweden.
- Lund University Cancer Center, LUCC, Lund, Sweden.
| |
Collapse
|
48
|
Pavlova AV, Zvyagin IV, Shugay M. Detecting T-cell clonal expansions and quantifying clone survival using deep profiling of immune repertoires. Front Immunol 2024; 15:1321603. [PMID: 38633256 PMCID: PMC11021634 DOI: 10.3389/fimmu.2024.1321603] [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: 10/14/2023] [Accepted: 03/12/2024] [Indexed: 04/19/2024] Open
Abstract
An individual's T-cell repertoire constantly changes under the influence of external and internal factors. Cells that do not receive a stimulatory signal die, while those that encounter and recognize a pathogen or receive a co-stimulatory signal divide, resulting in clonal expansions. T-cell clones can be traced by monitoring the presence of their unique T-cell receptor (TCR) sequence, which is assembled de novo through a process known as V(D)J rearrangement. Tracking T cells can provide valuable insights into the survival of cells after hematopoietic stem cell transplantation (HSCT) or cancer treatment response and can indicate the induction of protective immunity by vaccination. In this study, we report a bioinformatic method for quantifying the T-cell repertoire dynamics from TCR sequencing data. We demonstrate its utility by measuring the T-cell repertoire stability in healthy donors, by quantifying the effect of donor lymphocyte infusion (DLI), and by tracking the fate of the different T-cell subsets in HSCT patients and the expansion of pathogen-specific clones in vaccinated individuals.
Collapse
Affiliation(s)
- Anastasia V. Pavlova
- Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Ivan V. Zvyagin
- Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Mikhail Shugay
- Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
49
|
Sheetikov SA, Khmelevskaya AA, Zornikova KV, Zvyagin IV, Shomuradova AS, Serdyuk YV, Shakirova NT, Peshkova IO, Titov A, Romaniuk DS, Shagina IA, Chudakov DM, Kiryukhin DO, Shcherbakova OV, Khamaganova EG, Dzutseva V, Afanasiev A, Bogolyubova AV, Efimov GA. Clonal structure and the specificity of vaccine-induced T cell response to SARS-CoV-2 Spike protein. Front Immunol 2024; 15:1369436. [PMID: 38629062 PMCID: PMC11018901 DOI: 10.3389/fimmu.2024.1369436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
Adenovirus vaccines, particularly the COVID-19 Ad5-nCoV adenovirus vaccine, have emerged as promising tools in the fight against infectious diseases. In this study, we investigated the structure of the T cell response to the Spike protein of the SARS-CoV-2 virus used in the COVID-19 Ad5-nCoV adenoviral vaccine in a phase 3 clinical trial (NCT04540419). In 69 participants, we collected peripheral blood samples at four time points after vaccination or placebo injection. Sequencing of T cell receptor repertoires from Spike-stimulated T cell cultures at day 14 from 17 vaccinated revealed a more diverse CD4+ T cell repertoire compared to CD8+. Nevertheless, CD8+ clonotypes accounted for more than half of the Spike-specific repertoire. Our longitudinal analysis showed a peak T cell response at day 14, followed by a decline until month 6. Remarkably, multiple T cell clonotypes persisted for at least 6 months after vaccination, as demonstrated by ex vivo stimulation. Examination of CDR3 regions revealed homologous sequences in both CD4+ and CD8+ clonotypes, with major CD8+ clonotypes sharing high similarity with annotated sequences specific for the NYNYLYRLF peptide, suggesting potential immunodominance. In conclusion, our study demonstrates the immunogenicity of the Ad5-nCoV adenoviral vaccine and highlights its ability to induce robust and durable T cell responses. These findings provide valuable insight into the efficacy of the vaccine against COVID-19 and provide critical information for ongoing efforts to control infectious diseases.
Collapse
Affiliation(s)
- Saveliy A. Sheetikov
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alexandra A. Khmelevskaya
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Ksenia V. Zornikova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Ivan V. Zvyagin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Alina S. Shomuradova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Yana V. Serdyuk
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Naina T. Shakirova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Iuliia O. Peshkova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Aleksei Titov
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Dmitrii S. Romaniuk
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Irina A. Shagina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Dmitry M. Chudakov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Central European Institute of Technology, Masaryk University, Brno, Czechia
| | - Dmitry O. Kiryukhin
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Olga V. Shcherbakova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Ekaterina G. Khamaganova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Vitalina Dzutseva
- Novosibirsk State University, Medical School, Novosibirsk, Russia
- NPO Petrovax Pharm LLC, Moscow, Russia
| | | | | | - Grigory A. Efimov
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| |
Collapse
|
50
|
Li G, Chen Y, Liu Y, Gao Z, Jia R, Lv Z, Li Y, Wang Z, Han G. TCR β chain repertoire characteristic between healthy human CD4+ and CD8+ T cells. Biosci Rep 2024; 44:BSR20231653. [PMID: 38323526 PMCID: PMC10920061 DOI: 10.1042/bsr20231653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/03/2024] [Accepted: 02/06/2024] [Indexed: 02/08/2024] Open
Abstract
T cell is vital in the adaptive immune system, which relays on T-cell receptor (TCR) to recognize and defend against infection and tumors. T cells are mainly divided into well-known CD4+ and CD8+ T cells, which can recognize short peptide antigens presented by major histocompatibility complex (MHC) class II and MHC class I respectively in humoral and cell-mediated immunity. Due to the Human Leukocyte Antigen (HLA) diversity and restriction with peptides complexation, TCRs are quite diverse and complicated. To better elucidate the TCR in humans, the present study shows the difference between the TCR repertoire in CD4+ and CD8+ T cells from 30 healthy donors. The result showed count, clonality, diversity, frequency, and VDJ usage in CD4+ and CD8+ TCR-β repertoire is different, but CDR3 length is not. The Common Clone Cluster result showed that CD4+ and CD8+ TCR repertoires are connected separately between the bodies, which is odd considering the HLA diversity. More knowledge about TCR makes more opportunities for immunotherapy. The TCR repertoire is still a myth for discovery.
Collapse
Affiliation(s)
- Ge Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yaqiong Chen
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yinji Liu
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Zhenfang Gao
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ruiyan Jia
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Zhonglin Lv
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yuxiang Li
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Zhiding Wang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Gencheng Han
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| |
Collapse
|