1
|
Shirley S, Ichise H, Di Natale V, Jin J, Wu C, Zou R, Zhang W, Fang Y, Zhang Y, Chen M, Peng S, Basu U, Que J, Huang Y. A vasculature-resident innate lymphoid cell population in mouse lungs. Nat Commun 2025; 16:3718. [PMID: 40253407 PMCID: PMC12009297 DOI: 10.1038/s41467-025-58982-1] [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: 09/29/2024] [Accepted: 04/08/2025] [Indexed: 04/21/2025] Open
Abstract
Tissue-resident immune cells such as innate lymphoid cells (ILC) are known to reside in the parenchymal compartments of tissues and modulate local immune protection. Here we use intravascular cell labeling, parabiosis and multiplex 3D imaging to identify a population of group 3 ILCs in mice that are present within the intravascular space of lung blood vessels (vILC3). vILC3s are distributed broadly in alveolar capillary beds from which inhaled pathogens enter the lung parenchyma. By contrast, conventional ILC3s in tissue parenchyma are enriched in lymphoid clusters in proximity to large veins. In a mouse model of pneumonia, Pseudomonas aeruginosa infection results in rapid vILC3 expansion and production of chemokines including CCL4. Blocking CCL4 in vivo attenuates neutrophil recruitment to the lung at the early stage of infection, resulting in prolonged inflammation and delayed bacterial clearance. Our findings thus define the intravascular space as a site of ILC residence in mice, and reveal a unique immune cell population that interfaces with tissue alarmins and the circulating immune system for timely host defense.
Collapse
Affiliation(s)
- Simon Shirley
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Hiroshi Ichise
- Lymphocyte Biology Section, Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Vincenzo Di Natale
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Jiacheng Jin
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Christine Wu
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Raymond Zou
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Wanwei Zhang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Yinshan Fang
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, USA
| | - Yingyu Zhang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Miao Chen
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Sophia Peng
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Uttiya Basu
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Jianwen Que
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, USA.
| | - Yuefeng Huang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
2
|
Shin HS, Kim H, Kwon SG, Lee H, Lee JO, Kim YS. Tumor cells ectopically expressing the membrane-bound form of IL-7 develop an antitumor immune response efficiently in a colon carcinoma model. Mol Cells 2025; 48:100175. [PMID: 39743142 PMCID: PMC11873615 DOI: 10.1016/j.mocell.2024.100175] [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: 09/26/2024] [Revised: 12/20/2024] [Accepted: 12/25/2024] [Indexed: 01/04/2025] Open
Abstract
Various approaches employing cytokines and cytokine gene-modified tumor cells have been explored to induce antitumor responses, yet their widespread application has been limited due to efficacy concerns and adverse effects. In this study, interleukin-7 was engineered for expression both as a natural secretory form (sIL-7) and as a membrane-bound form fused with the B7.1 type I transmembrane protein (mbIL-7/B7) on CT26 colon cancer cells. Analysis of the resulting cell clones demonstrated that ectopically expressed sIL-7 and mbIL-7/B7 both retained similar capacities to induce the expansion and activation of CD8+ T cells and to enhance antitumor responses in vitro. While the sIL-7 or mbIL-7/B7 clones showed similar growth in culture, the mbIL-7/B7 clone exhibited lower tumorigenicity in mice compared with the sIL-7 clone or wild-type CT26 cells. Specifically, the mbIL-7/B7 clone failed to form tumors in approximately 60% of the mice injected with it. Moreover, 80% of mice that rejected the mbIL-7/B7 clone developed long-term systemic immunity against CT26 cells. Analysis of immune cells within the tumor masses revealed significant increases in CD4+ T cells, CD8+ T cells, and dendritic cells in tumors formed by the mbIL-7/B7 clone compared to those formed by the sIL-7 clone. These findings suggest that the membrane-bound form of IL-7 with B7.1 is more effective than the secretory form in establishing antitumor immunity within the tumor microenvironment. Our strategy of expressing the mbIL-7/B7 chimera holds promise as a novel approach for tumor therapy, particularly in cases requiring IL-7 supplementation.
Collapse
Affiliation(s)
- Hee-Su Shin
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Hyejin Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Korea
| | - Soon-Gyu Kwon
- Department of Life Sciences and Institute of Membrane Proteins, POSTECH, Pohang 37673, Korea
| | - Hayyoung Lee
- Department of Life Sciences and Postech Biotech Center, POSTECH, Pohang 37673, Korea
| | - Jie-Oh Lee
- Department of Life Sciences and Institute of Membrane Proteins, POSTECH, Pohang 37673, Korea.
| | - Young Sang Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Korea.
| |
Collapse
|
3
|
Bachoo S, Gudgeon N, Mann R, Stavrou V, Bishop EL, Kelly A, Uribe AH, Loeliger J, Frick C, Maddocks ODK, Lavender P, Hess C, Dimeloe S. IL-7 promotes integrated glucose and amino acid sensing during homeostatic CD4 + T cell proliferation. Cell Rep 2025; 44:115199. [PMID: 39799568 DOI: 10.1016/j.celrep.2024.115199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/15/2024] [Accepted: 12/20/2024] [Indexed: 01/15/2025] Open
Abstract
Interleukin (IL)-7 promotes T cell expansion during lymphopenia. We studied the metabolic basis in CD4+ T cells, observing increased glucose usage for nucleotide synthesis and oxidation in the tricarboxylic acid (TCA) cycle. Unlike other TCA metabolites, glucose-derived citrate does not accumulate upon IL-7 exposure, indicating diversion into other processes. In agreement, IL-7 promotes glucose-dependent histone acetylation and chromatin accessibility, notable at the loci of the amino acid-sensing Ragulator complex. Consistently, the expression of its subunit late endosomal/lysosomal adaptor, MAPK and mTOR activator 5 (LAMTOR5) is promoted by IL-7 in a glucose-dependent manner, and glucose availability determines amino acid-dependent mechanistic target of rapamycin (mTOR) activation, confirming integrated nutrient sensing. LAMTOR5 deletion impairs IL-7-mediated T cell expansion, establishing that glycolysis in the absence of Ragulator activation is insufficient to support this. Clinically, CD4+ T cells from stem cell transplant recipients demonstrate coordinated upregulation of glycolytic and TCA cycle enzymes, amino acid-sensing machinery, and mTOR targets, highlighting the potential to therapeutically target this pathway to fine-tune lymphopenia-induced T cell proliferation.
Collapse
Affiliation(s)
- Seema Bachoo
- School of Infection, Inflammation and Immunology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Nancy Gudgeon
- School of Infection, Inflammation and Immunology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Rebecca Mann
- School of Infection, Inflammation and Immunology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Victoria Stavrou
- School of Infection, Inflammation and Immunology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Emma L Bishop
- School of Infection, Inflammation and Immunology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Audrey Kelly
- School of Immunology & Microbial Sciences, MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - Alejandro Huerta Uribe
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Jordan Loeliger
- Immunobiology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Corina Frick
- Immunobiology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Oliver D K Maddocks
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Paul Lavender
- School of Immunology & Microbial Sciences, MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - Christoph Hess
- Immunobiology, Department of Biomedicine, University of Basel, Basel, Switzerland; Department of Medicine, CITIID, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Sarah Dimeloe
- School of Infection, Inflammation and Immunology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
4
|
Wang Y, Li D, Liu Y, Chen S, Dong Z. Adaptive immune cells antagonize ILC2 homeostasis via SLAMF3 and SLAMF5. SCIENCE ADVANCES 2025; 11:eadp9894. [PMID: 39792675 PMCID: PMC11721569 DOI: 10.1126/sciadv.adp9894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025]
Abstract
Type 2 innate lymphoid cells (ILC2s) mainly reside in tissues with few lymphoid cells. How their tissue residency is regulated remains poorly understood. This study explores the inhibitory role of SLAM-family receptors (SFRs) on adaptive immune cells in ILC2 maintenance. We observed an increase in the population of ILC2s in Rag1-deficient mice. Homotypic engagement of SFRs between ILC2s and adaptive immune cells was identified as a potential mechanism. SFR deficiency led to an increase in ILC2s. Conditional deletion of SFRs on T and/or B cells led to an increased ILC2 abundance. Mechanistically, as ILC precursors differentiate into ILC2s, SFRs, primarily SLAMF3 and SLAMF5, are inhibitory, which impair IL-7-induced PI3K activation and enhance apoptosis via SHP-1. These findings reveal a mechanism by which adaptive immune cells negatively regulate the homeostasis of ILC2s and contribute to our understanding of the complex interplay between innate and adaptive immune cells in the regulation of immune responses.
Collapse
Affiliation(s)
- Yuande Wang
- Department of Allergy, the First Affiliated Hospital of Anhui Medical University and Institute of Clinical Immunology, Anhui Medical University, Hefei 230032, China
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Dan Li
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Yehai Liu
- Department of Allergy, the First Affiliated Hospital of Anhui Medical University and Institute of Clinical Immunology, Anhui Medical University, Hefei 230032, China
| | - Shasha Chen
- Department of Allergy, the First Affiliated Hospital of Anhui Medical University and Institute of Clinical Immunology, Anhui Medical University, Hefei 230032, China
- Innovative Institute of Tumor Immunity and Medicine (ITIM), Hefei 230032, China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei 230032, China
| | - Zhongjun Dong
- Department of Allergy, the First Affiliated Hospital of Anhui Medical University and Institute of Clinical Immunology, Anhui Medical University, Hefei 230032, China
- State Key Laboratory of Membrane Biology, School of Medicine and Institute for Immunology, Tsinghua University, Beijing 100084, China
- Innovative Institute of Tumor Immunity and Medicine (ITIM), Hefei 230032, China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei 230032, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
5
|
Flögel L, Kaiser E, Hans MC, Goedicke-Fritz S, Bous M, Abdul-Khaliq H, Poryo M, Zemlin M, Weber R. Immunological characterization of pleural effusions in pediatric patients. Front Immunol 2024; 15:1506073. [PMID: 39737183 PMCID: PMC11682977 DOI: 10.3389/fimmu.2024.1506073] [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: 10/04/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
Background The pleural cavity represents a unique immunological compartment that can mount inflammatory reactions during infections, after surgery and in chronic immunological diseases. The connection between systemic immune reactions in the blood and local immune reactions in pleural effusions remains unclear. This study provides the first comprehensive immunological characterization of paired blood and pleural effusion samples, utilizing combined cell and cytokine analyses in pediatric patients undergoing cardiac surgery. Methods In 30 pediatric patients (median age: 22 months) with pleural effusion after cardiac surgery for congenital heart defects, corresponding peripheral blood and pleural effusion samples were analyzed for their immune response. We used flow cytometry and multiplex immunoassays to quantify 14 T cell subpopulations and 12 T cell associated cytokines in each biosample. Results IL-6, IL-8, IL-10, TNF (p<0.0001) levels were significantly higher in pleural effusion compared to plasma. In contrast, IFN-γ, GM-CSF, IL-17A levels were lower in pleural effusion than in plasma (p ≤ 0.0005). In comparison to peripheral blood, there was a significantly higher proportion of T helper cells 1 (Th1, p=0.0023), T helper cells 17 (Th17, p=0.0334) and memory effector cytotoxic T cells (CD3+CD8+CD45RO+CD62L-, p=0.0449) in pleural effusion and the same trend was observed for memory effector Th cells (CD3+CD4+CD45RO+CD62L-, p=0.0633) and double-negative T cells (CD3+CD4-CD8-) (p=0.1085). Naïve Th cells (CD3+CD4+CD45RO-CD62L+) and naïve cytotoxic T cells (CD3+CD8+CD45RO-CD62L+) were slightly reduced in pleural effusion compared to peripheral blood (not significant). Conclusion Immunological factors in pleural effusions differed significantly from the corresponding blood samples in pediatric patients after cardiac surgery. The results suggest localized production of specific cytokines within the pleural space, while the distribution of other cytokines in pleural effusions appears to be more reflective of the systemic immune response. We found evidence that on the cellular level, the surface marker CD62L may play a key role in navigating T cells between the blood and pleural effusion. This study confirms that the pleural cavity harbors a unique lymphatic compartment, the analysis of which may be useful for both diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Luca Flögel
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| | - Elisabeth Kaiser
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| | - Muriel Charlotte Hans
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| | - Sybelle Goedicke-Fritz
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| | - Michelle Bous
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| | - Hashim Abdul-Khaliq
- Department of Pediatric Cardiology, Saarland University Medical Center, Homburg, Germany
| | - Martin Poryo
- Department of Pediatric Cardiology, Saarland University Medical Center, Homburg, Germany
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| | - Regine Weber
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| |
Collapse
|
6
|
Kawajiri A, Li J, Koinuma K, Yang Z, Yoon HJ, Yi J, Nagashima H, Ishii M, Gao F, Sato K, Tayama S, Harigae H, Iwakura Y, Ishii N, Sher A, Ishigaki K, Zhu J, Kim KS, Kawabe T. Naturally arising memory-phenotype CD4 + T lymphocytes contain an undifferentiated population that can generate T H1, T H17, and T reg cells. SCIENCE ADVANCES 2024; 10:eadq6618. [PMID: 39630890 PMCID: PMC11619248 DOI: 10.1126/sciadv.adq6618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
Memory-phenotype (MP) CD4+ T lymphocytes develop from naïve cells via self-recognition at homeostasis. While previous studies defined MP cells as a heterogeneous population that comprises T helper 1 (TH1)/17-like subsets, functional significance of the T-bet- Rorγt- subpopulation remains unknown. Here we show that MP lymphocytes as a whole population can differentiate into TH1/17/regulatory T (Treg) cells to mediate mild and persistent inflammation in lymphopenic environments, whereas naïve cells exhibit strong, TH1-dominated responses. Moreover, we demonstrate that MP lymphocytes comprise not only TH1/17-differentiated subsets but a polyclonal, transcriptomically immature "undifferentiated" subpopulation at homeostasis. Furthermore, our data argue that while the T-bet+ Rorγt- MP subset is terminally TH1-differentiated, its undifferentiated counterpart retains the capacity to rapidly proliferate to differentiate into TH1/17/Treg cells, with the latter response tonically constrained by preexisting Treg cells. Together, our results identify undifferentiated MP CD4+ T lymphocytes as a unique precursor that has a diverse differentiation potential to generate TH1/17/Treg cells to contribute to pathogenesis of inflammation.
Collapse
Affiliation(s)
- Akihisa Kawajiri
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Hematology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Jing Li
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Keita Koinuma
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ziying Yang
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hye Jin Yoon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jaeu Yi
- Department of Internal Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biological Science, Ajou University, Suwon, Republic of Korea
| | - Hiroyuki Nagashima
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Minami Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Feng Gao
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kosuke Sato
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Shunichi Tayama
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hideo Harigae
- Department of Hematology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Alan Sher
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kazuyoshi Ishigaki
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kwang Soon Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
7
|
Bazarbachi AH, Mapara MY. Cytokines in hematopoietic cell transplantation and related cellular therapies. Best Pract Res Clin Haematol 2024; 37:101600. [PMID: 40074514 DOI: 10.1016/j.beha.2025.101600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025]
Abstract
Cytokines are pleiotropic molecules involved in hematopoiesis, immune responses, infections, and inflammation. They play critical roles in hematopoietic cell transplantation (HCT) and immune effector cell (IEC) therapies, mediating both therapeutic and adverse effects. Thus, cytokines contribute to the immunopathology of graft-versus-host disease (GVHD), cytokine release syndrome (CRS), and immune effector cell-associated neurotoxicity syndrome (ICANS). This review examines cytokine functions in these contexts, their influence on engraftment and immune recovery post-transplantation, and their role in mediating toxicities. We focus on current and potential uses of cytokines to enhance engraftment and potentiate IEC therapies, as well as strategies to mitigate cytokine-mediated complications using cytokine blockers (e.g., tocilizumab, anakinra) and JAK inhibitors (e.g., ruxolitinib). We discuss new insights into GVHD physiology that have led to novel treatments, such as CSF1R blockade, which is effective in refractory chronic GVHD.
Collapse
Affiliation(s)
- Abdul-Hamid Bazarbachi
- Division of Hematology/Oncology, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, NY, USA
| | - Markus Y Mapara
- Division of Hematology/Oncology, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, NY, USA; Columbia Center for Translational Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, USA.
| |
Collapse
|
8
|
Yosri M, Dokhan M, Aboagye E, Al Moussawy M, Abdelsamed HA. Mechanisms governing bystander activation of T cells. Front Immunol 2024; 15:1465889. [PMID: 39669576 PMCID: PMC11635090 DOI: 10.3389/fimmu.2024.1465889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/31/2024] [Indexed: 12/14/2024] Open
Abstract
The immune system is endowed with the capacity to distinguish between self and non-self, so-called immune tolerance or "consciousness of the immune system." This type of awareness is designed to achieve host protection by eliminating cells expressing a wide range of non-self antigens including microbial-derived peptides. Such a successful immune response is associated with the secretion of a whole spectrum of soluble mediators, e.g., cytokines and chemokines, which not only contribute to the clearance of infected host cells but also activate T cells that are not specific to the original cognate antigen. This kind of non-specific T-cell activation is called "bystander activation." Although it is well-established that this phenomenon is cytokine-dependent, there is evidence in the literature showing the involvement of peptide/MHC recognition depending on the type of T-cell subset (naive vs. memory). Here, we will summarize our current understanding of the mechanism(s) of bystander T-cell activation as well as its biological significance in a wide range of diseases including microbial infections, cancer, auto- and alloimmunity, and chronic inflammatory diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Mohammed Yosri
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo, Egypt
| | - Mohamed Dokhan
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
| | - Elizabeth Aboagye
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
| | - Mouhamad Al Moussawy
- Starzl Transplantation Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hossam A. Abdelsamed
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
- Department of Physiology, Augusta University, Augusta, GA, United States
| |
Collapse
|
9
|
Arango-Franco CA, Ogishi M, Unger S, Delmonte OM, Orrego JC, Yatim A, Velasquez-Lopera MM, Zea-Vera AF, Bohlen J, Chbihi M, Fayand A, Sánchez JP, Rojas J, Seeleuthner Y, Le Voyer T, Philippot Q, Payne KJ, Gervais A, Erazo-Borrás LV, Correa-Londoño LA, Cederholm A, Gallón-Duque A, Goncalves P, Doisne JM, Horev L, Charmeteau-de Muylder B, Álvarez JÁ, Arboleda DM, Pérez-Zapata L, Vásquez-Echeverri E, Moncada-Vélez M, López JA, Caicedo Y, Palterer B, Patiño PJ, Montoya CJ, Chaldebas M, Zhang P, Nguyen T, Ma CS, Jeljeli M, Alzate JF, Cabarcas F, Khan T, Rinchai D, Prétet JL, Boisson B, Marr N, Ibrahim R, Molho-Pessach V, Boisson-Dupuis S, Kiritsi D, Barata JT, Landegren N, Neven B, Abel L, Lisco A, Béziat V, Jouanguy E, Bustamante J, Di Santo JP, Tangye SG, Notarangelo LD, Cheynier R, Natsuga K, Arias AA, Franco JL, Warnatz K, Casanova JL, Puel A. IL-7-dependent and -independent lineages of IL-7R-dependent human T cells. J Clin Invest 2024; 134:e180251. [PMID: 39352394 PMCID: PMC11444196 DOI: 10.1172/jci180251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 08/06/2024] [Indexed: 10/03/2024] Open
Abstract
Infants with biallelic IL7R loss-of-function variants have severe combined immune deficiency (SCID) characterized by the absence of autologous T lymphocytes, but normal counts of circulating B and NK cells (T-B+NK+ SCID). We report 6 adults (aged 22 to 59 years) from 4 kindreds and 3 ancestries (Colombian, Israeli Arab, Japanese) carrying homozygous IL7 loss-of-function variants resulting in combined immunodeficiency (CID). Deep immunophenotyping revealed relatively normal counts and/or proportions of myeloid, B, NK, and innate lymphoid cells. By contrast, the patients had profound T cell lymphopenia, with low proportions of innate-like adaptive mucosal-associated invariant T and invariant NK T cells. They also had low blood counts of T cell receptor (TCR) excision circles, recent thymic emigrant T cells and naive CD4+ T cells, and low overall TCR repertoire diversity, collectively indicating impaired thymic output. The proportions of effector memory CD4+ and CD8+ T cells were high, indicating IL-7-independent homeostatic T cell proliferation in the periphery. Intriguingly, the proportions of other T cell subsets, including TCRγδ+ T cells and some TCRαβ+ T cell subsets (including Th1, Tfh, and Treg) were little affected. Peripheral CD4+ T cells displayed poor proliferation, but normal cytokine production upon stimulation with mitogens in vitro. Thus, inherited IL-7 deficiency impairs T cell development less severely and in a more subset-specific manner than IL-7R deficiency. These findings suggest that another IL-7R-binding cytokine, possibly thymic stromal lymphopoietin, governs an IL-7-independent pathway of human T cell development.
Collapse
Affiliation(s)
- Carlos A Arango-Franco
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Susanne Unger
- Department of Rheumatology and Clinical Immunology and
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University Freiburg, Freiburg, Germany
| | - Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Julio César Orrego
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Ahmad Yatim
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Margarita M Velasquez-Lopera
- Sección de Dermatología, Facultad de Medicina, Universidad de Antioquia, Centro de Investigaciones Dermatológicas (CIDERM), Medellín, Antioquia, Colombia
| | - Andrés F Zea-Vera
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
- Clinical Immunology Clinic, Hospital Universitario del Valle, Cali, Colombia
- Microbiology Department, Universidad del Valle, Cali, Colombia
| | - Jonathan Bohlen
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Marwa Chbihi
- Paris Cité University, Imagine Institute, Paris, France
- Pediatric Immunology, Hematology and Rheumatology Department, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Antoine Fayand
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Juan Pablo Sánchez
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- Microbiology School, University of Antioquia UdeA, Medellín, Colombia
| | - Julian Rojas
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- Microbiology School, University of Antioquia UdeA, Medellín, Colombia
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Kathryn J Payne
- Department of Rheumatology and Clinical Immunology and
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University Freiburg, Freiburg, Germany
| | - Adrian Gervais
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Lucia V Erazo-Borrás
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Luis A Correa-Londoño
- Sección de Dermatología, Facultad de Medicina, Universidad de Antioquia, Centro de Investigaciones Dermatológicas (CIDERM), Medellín, Antioquia, Colombia
| | - Axel Cederholm
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Alejandro Gallón-Duque
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Pedro Goncalves
- Innate Immunity Unit, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | - Jean-Marc Doisne
- Innate Immunity Unit, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | - Liran Horev
- Faculty of Medicine, Hebrew University of Jerusalem, Pediatric Dermatology Service, Department of Dermatology, Hadassah Medical Center, Jerusalem, Israel
- Shamir (Assaf Harofeh) Medical Center, Be'er Ya'akov, Israel
| | | | - Jesús Á Álvarez
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Diana M Arboleda
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Lizet Pérez-Zapata
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Estefanía Vásquez-Echeverri
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Marcela Moncada-Vélez
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Juan A López
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- Microbiology School, University of Antioquia UdeA, Medellín, Colombia
| | | | - Boaz Palterer
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Pablo J Patiño
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Carlos J Montoya
- School of Medicine, University of Antioquia UdeA, Medellin, Colombia
| | - Matthieu Chaldebas
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Peng Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Tina Nguyen
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
| | - Mohamed Jeljeli
- Cochin University Hospital, Biological Immunology Unit, AP-HP, Paris, France
| | - Juan F Alzate
- Centro Nacional de Secuenciación Genómica CNSG, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Felipe Cabarcas
- Centro Nacional de Secuenciación Genómica CNSG, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Taushif Khan
- Research Branch, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Darawan Rinchai
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Jean-Luc Prétet
- Université de Franche-Comté, CNRS, Chrono-environnement & CHU Besançon, Centre National de Référence Papillomavirus, F-25000 Besançon, France
| | - Bertrand Boisson
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Nico Marr
- Research Branch, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Ruba Ibrahim
- Faculty of Medicine, Hebrew University of Jerusalem, Pediatric Dermatology Service, Department of Dermatology, Hadassah Medical Center, Jerusalem, Israel
| | - Vered Molho-Pessach
- Faculty of Medicine, Hebrew University of Jerusalem, Pediatric Dermatology Service, Department of Dermatology, Hadassah Medical Center, Jerusalem, Israel
| | - Stéphanie Boisson-Dupuis
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Dimitra Kiritsi
- Department of Dermatology, University Medical Center of Freiburg, Freiburg, Germany
| | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Nils Landegren
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Centre for Molecular Medicine, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Bénédicte Neven
- Paris Cité University, Imagine Institute, Paris, France
- Pediatric Immunology, Hematology and Rheumatology Department, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Andrea Lisco
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
- Center for the Study of Primary Immunodeficiencies, Necker Hospital for Sick Children, Paris, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Rémi Cheynier
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Ken Natsuga
- Department of Dermatology, Faculty of Medicine and Graduate of Medicine, Hokkaido University, Sapporo, Japan
| | - Andrés A Arias
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
- Microbiology School, University of Antioquia UdeA, Medellín, Colombia
| | - José Luis Franco
- Inborn Errors of Immunity Group, (Primary Immunodeficiencies), School of Medicine, University of Antioquia UdeA, Medellín, Colombia
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology and
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Faculty of Medicine, University Freiburg, Freiburg, Germany
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
- Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, Paris, France
- Howard Hughes Medical Institute, New York, New York, USA
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| |
Collapse
|
10
|
Clark PA, Gogoi M, Rodriguez-Rodriguez N, Ferreira ACF, Murphy JE, Walker JA, Crisp A, Jolin HE, Shields JD, McKenzie ANJ. Recipient tissue microenvironment determines developmental path of intestinal innate lymphoid progenitors. Nat Commun 2024; 15:7809. [PMID: 39242588 PMCID: PMC11379955 DOI: 10.1038/s41467-024-52155-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/27/2024] [Indexed: 09/09/2024] Open
Abstract
Innate lymphoid cells (ILCs) are critical in maintaining tissue homeostasis, and during infection and inflammation. Here we identify, by using combinatorial reporter mice, a rare ILC progenitor (ILCP) population, resident to the small intestinal lamina propria (siLP) in adult mice. Transfer of siLP-ILCP into recipients generates group 1 ILCs (including ILC1 and NK cells), ILC2s and ILC3s within the intestinal microenvironment, but almost exclusively group 1 ILCs in the liver, lung and spleen. Single cell gene expression analysis and high dimensional spectral cytometry analysis of the siLP-ILCPs and ILC progeny indicate that the phenotype of the group 1 ILC progeny is also influenced by the tissue microenvironment. Thus, a local pool of siLP-ILCP can contribute to pan-ILC generation in the intestinal microenvironment but has more restricted potential in other tissues, with a greater propensity than bone marrow-derived ILCPs to favour ILC1 and ILC3 production. Therefore, ILCP potential is influenced by both tissue of origin and the microenvironment during development. This may provide additional flexibility during the tuning of immune reactions.
Collapse
Affiliation(s)
- Paula A Clark
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom.
| | - Mayuri Gogoi
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | | | | - Jane E Murphy
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | | - Alastair Crisp
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Helen E Jolin
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Jacqueline D Shields
- Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, United Kingdom
| | | |
Collapse
|
11
|
López DA, Griffin A, Aguilar LM, Deering-Rice C, Myers EJ, Warren KJ, Welner RS, Beaudin AE. Prenatal inflammation remodels lung immunity and function by programming ILC2 hyperactivation. Cell Rep 2024; 43:114365. [PMID: 38909363 DOI: 10.1016/j.celrep.2024.114365] [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: 01/11/2024] [Revised: 04/30/2024] [Accepted: 05/31/2024] [Indexed: 06/25/2024] Open
Abstract
Here, we examine how prenatal inflammation shapes tissue function and immunity in the lung by reprogramming tissue-resident immune cells from early development. Maternal, but not fetal, type I interferon-mediated inflammation provokes expansion and hyperactivation of group 2 innate lymphoid cells (ILC2s) seeding the developing lung. Hyperactivated ILC2s produce increased IL-5 and IL-13 and are associated with acute Th2 bias, decreased Tregs, and persistent lung eosinophilia into adulthood. ILC2 hyperactivation is recapitulated by adoptive transfer of fetal liver precursors following prenatal inflammation, indicative of developmental programming at the fetal progenitor level. Reprogrammed ILC2 hyperactivation and subsequent lung immune remodeling, including persistent eosinophilia, is concomitant with worsened histopathology and increased airway dysfunction equivalent to papain exposure, indicating increased asthma susceptibility in offspring. Our data elucidate a mechanism by which early-life inflammation results in increased asthma susceptibility in the presence of hyperactivated ILC2s that drive persistent changes to lung immunity during perinatal development.
Collapse
Affiliation(s)
- Diego A López
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Aleah Griffin
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Lorena Moreno Aguilar
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | | | - Elizabeth J Myers
- Department of Neurology, University of Utah, Salt Lake City, UT, USA
| | - Kristi J Warren
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Robert S Welner
- Department of Medicine, University of Alabama, Birmingham, AL, USA
| | - Anna E Beaudin
- Department of Pathology, University of Utah, Salt Lake City, UT, USA; Department of Internal Medicine and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
12
|
Chung DC, Garcia-Batres CR, Millar DG, Wong SWY, Elford AR, Mathews JA, Wang BX, Nguyen LT, Shaw PA, Clarke BA, Bernardini MQ, Sacher AG, Crome SQ, Ohashi PS. Generation of an Inhibitory NK Cell Subset by TGF-β1/IL-15 Polarization. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1904-1912. [PMID: 38668728 PMCID: PMC11149900 DOI: 10.4049/jimmunol.2300834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/02/2024] [Indexed: 06/05/2024]
Abstract
NK cells have been shown to exhibit inflammatory and immunoregulatory functions in a variety of healthy and diseased settings. In the context of chronic viral infection and cancer, distinct NK cell populations that inhibit adaptive immune responses have been observed. To understand how these cells arise and further characterize their immunosuppressive role, we examined in vitro conditions that could polarize human NK cells into an inhibitory subset. TGF-β1 has been shown to induce regulatory T cells in vitro and in vivo; we therefore investigated if TGF-β1 could also induce immunosuppressive NK-like cells. First, we found that TGF-β1/IL-15, but not IL-15 alone, induced CD103+CD49a+ NK-like cells from peripheral blood NK cells, which expressed markers previously associated with inhibitory CD56+ innate lymphoid cells, including high expression of GITR and CD101. Moreover, supernatant from ascites collected from patients with ovarian carcinoma also induced CD103+CD49a+ NK-like cells in vitro in a TGF-β-dependent manner. Interestingly, TGF-β1/IL-15-induced CD103+CD56+ NK-like cells suppressed autologous CD4+ T cells in vitro by reducing absolute number, proliferation, and expression of activation marker CD25. Collectively, these findings provide new insight into how NK cells may acquire an inhibitory phenotype in TGF-β1-rich environments.
Collapse
Affiliation(s)
- Douglas C. Chung
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Carlos R. Garcia-Batres
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Douglas G. Millar
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Stephanie W. Y. Wong
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Alisha R. Elford
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jessica A. Mathews
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Ben X. Wang
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Linh T. Nguyen
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Patricia A. Shaw
- Division of Gynecologic Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Blaise A. Clarke
- Division of Gynecologic Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Marcus Q. Bernardini
- Division of Gynecologic Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Adrian G. Sacher
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Medical Oncology & Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Sarah Q. Crome
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Pamela S. Ohashi
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Tumour Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
13
|
Lahire S, Fichel C, Rubaszewski O, Lerévérend C, Audonnet S, Visneux V, Perotin JM, Deslée G, Le Jan S, Potteaux S, Le Naour R, Pommier A. Elastin-derived peptides favor type 2 innate lymphoid cells in chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2024; 326:L812-L820. [PMID: 38712445 DOI: 10.1152/ajplung.00306.2023] [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: 09/26/2023] [Revised: 03/22/2024] [Accepted: 04/22/2024] [Indexed: 05/08/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a condition characterized by chronic airway inflammation and obstruction, primarily caused by tobacco smoking. Although the involvement of immune cells in COPD pathogenesis is well established, the contribution of innate lymphoid cells (ILCs) remains poorly understood. ILCs are a type of innate immune cells that participate in tissue remodeling processes, but their specific role in COPD has not been fully elucidated. During COPD, the breakdown of pulmonary elastin generates elastin peptides that elicit biological activities on immune cells. This study aimed to investigate the presence of ILC in patients with COPD and examine the impact of elastin peptides on their functionality. Our findings revealed an elevated proportion of ILC2 in the peripheral blood of patients with COPD, and a general activation of ILC as indicated by an increase in their cytokine secretion capacity. Notably, our study demonstrated that serum from patients with COPD promotes ILC2 phenotype, likely due to the elevated concentration of IL-5, a cytokine known to favor ILC2 activation. Furthermore, we uncovered that this increase in IL-5 secretion is partially attributed to its secretion by macrophages upon stimulation by elastin peptides, suggesting an indirect role of elastin peptides on ILC in COPD. These findings shed light on the involvement of ILC in COPD and provide insights into the potential interplay between elastin breakdown, immune cells, and disease progression. Further understanding of the mechanisms underlying ILC activation and their interaction with elastin peptides could contribute to the development of novel therapeutic strategies for COPD management.NEW & NOTEWORTHY Elastin-derived peptides, generated following alveolar degradation during emphysema in patients with COPD, are able to influence the response of type 2 innate lymphoid cells. We show that the orientation of innate lymphoid cells in patients with COPD is shifted toward a type 2 profile and that elastin peptides are indirectly participating in that shift through their influence of macrophages, which in turn impact innate lymphoid cells.
Collapse
Affiliation(s)
- Sarah Lahire
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Caroline Fichel
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Océane Rubaszewski
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Cédric Lerévérend
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
- Institut Godinot, Unicancer, Reims, France
| | - Sandra Audonnet
- Université de Reims Champagne Ardenne, Plateforme de cytométrie en flux, URCACyt, Reims, France
| | - Vincent Visneux
- CHU de Reims, Service des maladies respiratoires, Reims, France
| | - Jeanne-Marie Perotin
- CHU de Reims, Service des maladies respiratoires, Reims, France
- Université de Reims Champagne Ardenne, Inserm UMR-S 1250 Pathologies Pulmonaires et Plasticité Cellulaire (P3Cell), Reims, France
| | - Gaëtan Deslée
- CHU de Reims, Service des maladies respiratoires, Reims, France
- Université de Reims Champagne Ardenne, Inserm UMR-S 1250 Pathologies Pulmonaires et Plasticité Cellulaire (P3Cell), Reims, France
| | - Sébastien Le Jan
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Stéphane Potteaux
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
- Institut Godinot, Unicancer, Reims, France
- Délégation régionale Inserm Paris Ile-de France Centre Nord, Paris, France
| | - Richard Le Naour
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Arnaud Pommier
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| |
Collapse
|
14
|
Park JH, Lee SW, Choi D, Lee C, Sung YC. Harnessing the Power of IL-7 to Boost T Cell Immunity in Experimental and Clinical Immunotherapies. Immune Netw 2024; 24:e9. [PMID: 38455462 PMCID: PMC10917577 DOI: 10.4110/in.2024.24.e9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 03/09/2024] Open
Abstract
The cytokine IL-7 plays critical and nonredundant roles in T cell immunity so that the abundance and availability of IL-7 act as key regulatory mechanisms in T cell immunity. Importantly, IL-7 is not produced by T cells themselves but primarily by non-lymphoid lineage stromal cells and epithelial cells that are limited in their numbers. Thus, T cells depend on cell extrinsic IL-7, and the amount of in vivo IL-7 is considered a major factor in maximizing and maintaining the number of T cells in peripheral tissues. Moreover, IL-7 provides metabolic cues and promotes the survival of both naïve and memory T cells. Thus, IL-7 is also essential for the functional fitness of T cells. In this regard, there has been an extensive effort trying to increase the protein abundance of IL-7 in vivo, with the aim to augment T cell immunity and harness T cell functions in anti-tumor responses. Such approaches started under experimental animal models, but they recently culminated into clinical studies, with striking effects in re-establishing T cell immunity in immunocompromised patients, as well as boosting anti-tumor effects. Depending on the design, glycosylation, and the structure of recombinantly engineered IL-7 proteins and their mimetics, recombinant IL-7 molecules have shown dramatic differences in their stability, efficacy, cellular effects, and overall immune functions. The current review is aimed to summarize the past and present efforts in the field that led to clinical trials, and to highlight the therapeutical significance of IL-7 biology as a master regulator of T cell immunity.
Collapse
Affiliation(s)
- Jung-Hyun Park
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Donghoon Choi
- Research Institute of NeoImmune Tech., Co, Ltd., Bio Open Innovation Center, Pohang 37666, Korea
| | - Changhyung Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Young Chul Sung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| |
Collapse
|
15
|
Russell J, Chen L, Liu A, Wang J, Ghosh S, Zhong X, Shi H, Beutler B, Nair-Gill E. Lrp10 suppresses IL7R limiting CD8 T cell homeostatic expansion and anti-tumor immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.08.570738. [PMID: 38106103 PMCID: PMC10723380 DOI: 10.1101/2023.12.08.570738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Signals emanating from the T cell receptor (TCR), co-stimulatory receptors, and cytokine receptors each influence CD8 T cell fate. Understanding how these signals respond to homeostatic and microenvironmental cues can reveal new ways to therapeutically direct T cell function. Through forward genetic screening in mice, we discovered that loss-of-function mutations in LDL receptor related protein 10 ( Lrp10 ) caused naïve and central memory CD8 T cells to accumulate in peripheral lymphoid organs. Lrp10 encodes a conserved cell surface protein of unknown immunological function. Lrp10 was induced with T cell activation and its expression post-translationally suppressed IL7 receptor (IL7R) levels. Accordingly, Lrp10 deletion enhanced T cell homeostatic expansion through IL7R signaling. Lrp10 -deficient mice were also intrinsically resistant to syngeneic tumors. This phenotype depended on dense tumor infiltration of CD8 T cells that displayed increased memory cell characteristics, reduced terminal exhaustion, and augmented responses to immune checkpoint inhibition. Here, we present Lrp10 as a new negative regulator of CD8 T cell homeostasis and a host factor that controls tumor resistance with implications for immunotherapy.
Collapse
|
16
|
López DA, Griffin A, Aguilar LM, Rice CD, Myers EJ, Warren KJ, Welner R, Beaudin AE. Prenatal inflammation reprograms hyperactive ILC2s that promote allergic lung inflammation and airway dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567899. [PMID: 38045298 PMCID: PMC10690173 DOI: 10.1101/2023.11.20.567899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Allergic asthma is a chronic respiratory disease that initiates in early life, but causal mechanisms are poorly understood. Here we examined how prenatal inflammation shapes allergic asthma susceptibility by reprogramming lung immunity from early development. Induction of Type I interferon-mediated inflammation during development provoked expansion and hyperactivation of group 2 innate lymphoid cells (ILC2s) seeding the developing lung. Hyperactivated ILC2s produced increased IL-5 and IL-13, and were associated with acute Th2 bias, eosinophilia, and decreased Tregs in the lung. The hyperactive ILC2 phenotype was recapitulated by adoptive transfer of a fetal liver precursor following exposure to prenatal inflammation, indicative of developmental programming. Programming of ILC2 function and subsequent lung immune remodeling by prenatal inflammation led to airway dysfunction at baseline and in response to papain, indicating increased asthma susceptibility. Our data provide a link by which developmental programming of progenitors by early-life inflammation drives lung immune remodeling and asthma susceptibility through hyperactivation of lung-resident ILC2s. One Sentence Summary Prenatal inflammation programs asthma susceptibility by inducing the production of hyperactivated ILC2s in the developing lung.
Collapse
|
17
|
Joachim A, Aussel R, Gélard L, Zhang F, Mori D, Grégoire C, Villazala Merino S, Gaya M, Liang Y, Malissen M, Malissen B. Defective LAT signalosome pathology in mice mimics human IgG4-related disease at single-cell level. J Exp Med 2023; 220:e20231028. [PMID: 37624388 PMCID: PMC10457416 DOI: 10.1084/jem.20231028] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/25/2023] [Accepted: 08/08/2023] [Indexed: 08/26/2023] Open
Abstract
Mice with a loss-of-function mutation in the LAT adaptor (LatY136F) develop an autoimmune and type 2 inflammatory disorder called defective LAT signalosome pathology (DLSP). We analyzed via single-cell omics the trajectory leading to LatY136F DLSP and the underlying CD4+ T cell diversification. T follicular helper cells, CD4+ cytotoxic T cells, activated B cells, and plasma cells were found in LatY136F spleen and lung. Such cell constellation entailed all the cell types causative of human IgG4-related disease (IgG4-RD), an autoimmune and inflammatory condition with LatY136F DLSP-like histopathological manifestations. Most previously described T cell-mediated autoimmune manifestations require persistent TCR input. In contrast, following their first engagement by self-antigens, the autoreactive TCR expressed by LatY136F CD4+ T cells hand over their central role in T cell activation to CD28 costimulatory molecules. As a result, all subsequent LatY136F DLSP manifestations, including the production of autoantibodies, solely rely on CD28 engagement. Our findings elucidate the etiology of the LatY136F DLSP and qualify it as a model of IgG4-RD.
Collapse
Affiliation(s)
- Anais Joachim
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Rudy Aussel
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Léna Gélard
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
- Centre d’Immunophénomique, INSERM, CNRS, Aix Marseille Université, Marseille, France
| | - Fanghui Zhang
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
- School of Laboratory Medicine, Henan Key Laboratory for Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang, China
| | - Daiki Mori
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
- Centre d’Immunophénomique, INSERM, CNRS, Aix Marseille Université, Marseille, France
| | - Claude Grégoire
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Sergio Villazala Merino
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Mauro Gaya
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Yinming Liang
- School of Laboratory Medicine, Henan Key Laboratory for Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang, China
| | - Marie Malissen
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
- Centre d’Immunophénomique, INSERM, CNRS, Aix Marseille Université, Marseille, France
- Laboratory of Immunophenomics, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Bernard Malissen
- Aix Marseille Université, INSERM, CNRS, Centre d’Immunologie de Marseille-Luminy, Marseille, France
- Centre d’Immunophénomique, INSERM, CNRS, Aix Marseille Université, Marseille, France
- Laboratory of Immunophenomics, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
18
|
Lancaster JN. Aging of lymphoid stromal architecture impacts immune responses. Semin Immunol 2023; 70:101817. [PMID: 37572552 PMCID: PMC10929705 DOI: 10.1016/j.smim.2023.101817] [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: 03/07/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
The secondary lymphoid organs (SLOs) undergo structural changes with age, which correlates with diminishing immune responses against infectious disease. A growing body of research suggests that the aged tissue microenvironment can contribute to decreased immune function, independent of intrinsic changes to hematopoietic cells with age. Stromal cells impart structural integrity, facilitate fluid transport, and provide chemokine and cytokine signals that are essential for immune homeostasis. Mechanisms that drive SLO development have been described, but their roles in SLO maintenance with advanced age are unknown. Disorganization of the fibroblasts of the T cell and B cell zones may reduce the maintenance of naïve lymphocytes and delay immune activation. Reduced lymphatic transport efficiency with age can also delay the onset of the adaptive immune response. This review focuses on recent studies that describe age-associated changes to the stroma of the lymph nodes and spleen. We also review recent investigations into stromal cell biology, which include high-dimensional analysis of the stromal cell transcriptome and viscoelastic testing of lymph node mechanical properties, as they constitute an important framework for understanding aging of the lymphoid tissues.
Collapse
Affiliation(s)
- Jessica N Lancaster
- Department of Immunology, Mayo Clinic, 13400 E. Shea Blvd., Scottsdale, AZ, USA; Department of Cancer Biology, Mayo Clinic, 13400 E. Shea Blvd., Scottsdale, AZ, USA.
| |
Collapse
|
19
|
Hou W, Zhang L, Chen J, Gu Y, Lv X, Zhang X, Li J, Liu H, Gao R. Expression Improvement of Recombinant Plasmids of the Interleukin-7 Gene in Chitosan-Derived Nanoparticles and Their Elevation of Mice Immunity. BIOLOGY 2023; 12:biology12050667. [PMID: 37237481 DOI: 10.3390/biology12050667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/27/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023]
Abstract
To investigate a safe and effective approach for enhancing the in vivo expression of recombinant genes and improving the systemic immunity of animals against infectious diseases, we employed the interleukin-7 (IL-7) gene from Tibetan pigs to construct a recombinant eukaryotic plasmid (VRTPIL-7). We first examined VRTPIL-7's bioactivity on porcine lymphocytes in vitro and then encapsulated it with polyethylenimine (PEI), chitosan copolymer (CS), PEG-modified galactosylated chitosan (CS-PEG-GAL) and methoxy poly (ethylene glycol) (PEG) and PEI-modified CS (CS-PEG-PEI) nanoparticles using the ionotropic gelation technique. Next, we intramuscularly or intraperitoneally injected mice with various nanoparticles containing VRTPIL-7 to evaluate their immunoregulatory effects in vivo. We observed a significant increase in neutralizing antibodies and specific IgG levels in response to the rabies vaccine in the treated mice compared to the controls. Treated mice also exhibited increased leukocytes, CD8+ and CD4+ T lymphocytes, and elevated mRNA levels of toll-like receptors (TLR1/4/6/9), IL-1, IL-2, IL-4, IL-6, IL-7, IL-23, and transforming growth factor-beta (TGF-β). Notably, the recombinant IL-7 gene encapsulated in CS-PEG-PEI induced the highest levels of immunoglobulins, CD4+ and CD8+ T cells, TLRs, and cytokines in the mice's blood, suggesting that chitosan-PEG-PEI may be a promising carrier for in vivo IL-7 gene expression and enhanced innate and adaptive immunity for the prevention of animal diseases.
Collapse
Affiliation(s)
- Wenli Hou
- Key Laboratory for Bioresource and Eco-Environment of the Education Ministry, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Linhan Zhang
- Key Laboratory for Bioresource and Eco-Environment of the Education Ministry, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Jianlin Chen
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, China
| | - Yiren Gu
- Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Xuebin Lv
- Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Xiuyue Zhang
- Key Laboratory for Bioresource and Eco-Environment of the Education Ministry, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Jiangling Li
- Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Hui Liu
- R&D Center, Chengdu Kanghua Biological Products Co., Ltd., Chengdu 610100, China
| | - Rong Gao
- Key Laboratory for Bioresource and Eco-Environment of the Education Ministry, College of Life Sciences, Sichuan University, Chengdu 610064, China
| |
Collapse
|
20
|
Schroeder JH, Beattie G, Lo JW, Zabinski T, Powell N, Neves JF, Jenner RG, Lord GM. CD90 is not constitutively expressed in functional innate lymphoid cells. Front Immunol 2023; 14:1113735. [PMID: 37114052 PMCID: PMC10126679 DOI: 10.3389/fimmu.2023.1113735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/28/2023] [Indexed: 04/29/2023] Open
Abstract
Huge progress has been made in understanding the biology of innate lymphoid cells (ILC) by adopting several well-known concepts in T cell biology. As such, flow cytometry gating strategies and markers, such as CD90, have been applied to indentify ILC. Here, we report that most non-NK intestinal ILC have a high expression of CD90 as expected, but surprisingly a sub-population of cells exhibit low or even no expression of this marker. CD90-negative and CD90-low CD127+ ILC were present amongst all ILC subsets in the gut. The frequency of CD90-negative and CD90-low CD127+ ILC was dependent on stimulatory cues in vitro and enhanced by dysbiosis in vivo. CD90-negative and CD90-low CD127+ ILC were a potential source of IL-13, IFNγ and IL-17A at steady state and upon dysbiosis- and dextran sulphate sodium-elicited colitis. Hence, this study reveals that, contrary to expectations, CD90 is not constitutively expressed by functional ILC in the gut.
Collapse
Affiliation(s)
- Jan-Hendrik Schroeder
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Gordon Beattie
- Cancer Research UK (CRUK) City of London Centre Single Cell Genomics Facility, University College London Cancer Institute, University College London (UCL), London, United Kingdom
- Genomics Translational Technology Platform, University College London (UCL) Cancer Institute, University College London, London, United Kingdom
| | - Jonathan W. Lo
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Tomasz Zabinski
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Nick Powell
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Joana F. Neves
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Richard G. Jenner
- University College London (UCL) Cancer Institute, University College London, London, United Kingdom
| | - Graham M. Lord
- School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
21
|
Guo N, Li N, Jia L, Jiang Q, Schreurs M, van Unen V, de Sousa Lopes SMC, Vloemans AA, Eggermont J, Lelieveldt B, Staal FJT, de Miranda NFCC, Pascutti MF, Koning F. Immune subset-committed proliferating cells populate the human foetal intestine throughout the second trimester of gestation. Nat Commun 2023; 14:1318. [PMID: 36899020 PMCID: PMC10006174 DOI: 10.1038/s41467-023-37052-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 03/01/2023] [Indexed: 03/12/2023] Open
Abstract
The intestine represents the largest immune compartment in the human body, yet its development and organisation during human foetal development is largely unknown. Here we show the immune subset composition of this organ during development, by longitudinal spectral flow cytometry analysis of human foetal intestinal samples between 14 and 22 weeks of gestation. At 14 weeks, the foetal intestine is mainly populated by myeloid cells and three distinct CD3-CD7+ ILC, followed by rapid appearance of adaptive CD4+, CD8+ T and B cell subsets. Imaging mass cytometry identifies lymphoid follicles from week 16 onwards in a villus-like structure covered by epithelium and confirms the presence of Ki-67+ cells in situ within all CD3-CD7+ ILC, T, B and myeloid cell subsets. Foetal intestinal lymphoid subsets are capable of spontaneous proliferation in vitro. IL-7 mRNA is detected within both the lamina propria and the epithelium and IL-7 enhances proliferation of several subsets in vitro. Overall, these observations demonstrate the presence of immune subset-committed cells capable of local proliferation in the developing human foetal intestine, likely contributing to the development and growth of organized immune structures throughout most of the 2nd trimester, which might influence microbial colonization upon birth.
Collapse
Affiliation(s)
- Nannan Guo
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Na Li
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- State Key Laboratory of Zoonotic Diseases, Institute of Zoonoses, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Li Jia
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Qinyue Jiang
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Mette Schreurs
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Vincent van Unen
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | | | | | - Jeroen Eggermont
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Frank J T Staal
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | - M Fernanda Pascutti
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands.
| | - Frits Koning
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
22
|
Kawabe T. Homeostasis and immunological function of self-driven memory-phenotype CD4 + T lymphocytes. Immunol Med 2023; 46:1-8. [PMID: 36218322 DOI: 10.1080/25785826.2022.2129370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
CD4+ T lymphocytes play an essential role in adaptive immune responses. In pathogen infection, naïve CD4+ T cells that strongly respond to foreign antigens robustly proliferate to differentiate into effector/memory cells, contributing to elimination of the pathogen concerned. In addition to this conventional T cell activation pathway, naïve T cells can also weakly respond to self antigens in the periphery to spontaneously acquire a memory phenotype through homeostatic proliferation in steady state. Such 'memory-phenotype' (MP) CD4+ T lymphocytes are distinguishable from foreign antigen-specific memory cells in terms of marker expression. Once generated, MP cells are maintained by rapid proliferation while differentiating into the T-bet+ 'MP1' subset, with the latter response promoted by IL-12 homeostatically produced by type 1 dendritic cells. Importantly, MP1 cells possess innate immune function; they can produce IFN-γ in response to IL-12 and IL-18 to contribute to host defense against pathogens. Similarly, the presence of RORγt+ 'MP17' and Gata3hi 'MP2' cells as well as their potential immune functions have been proposed. In this review, I will discuss our current understanding on the unique mechanisms of generation, maintenance, and differentiation of MP CD4+ T lymphocytes as well as their functional significance in various disease conditions.
Collapse
Affiliation(s)
- Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
23
|
Abstract
Mucosal associated invariant T (MAIT) cells are innate-like T lymphocytes, strikingly enriched at mucosal surfaces and characterized by a semi-invariant αβ T cell receptor (TCR) recognizing microbial derived intermediates of riboflavin synthesis presented by the MHC-Ib molecule MR1. At barrier sites MAIT cells occupy a prime position for interaction with commensal microorganisms, comprising the microbiota. The microbiota is a rich source of riboflavin derived antigens required in early life to promote intra-thymic MAIT cell development and sustain a life-long population of tissue resident cells. A symbiotic relationship is thought to be maintained in health whereby microbes promote maturation and homeostasis, and in turn MAIT cells can engage a TCR-dependent "tissue repair" program in the presence of commensal organisms conducive to sustaining barrier function and integrity of the microbial community. MAIT cell activation can be induced in a MR1-TCR dependent manner or through MR1-TCR independent mechanisms via pro-inflammatory cytokines interleukin (IL)-12/-15/-18 and type I interferon. MAIT cells provide immunity against bacterial, fungal and viral pathogens. However, MAIT cells may have deleterious effects through insufficient or exacerbated effector activity and have been implicated in autoimmune, inflammatory and allergic conditions in which microbial dysbiosis is a shared feature. In this review we summarize the current knowledge on the role of the microbiota in the development and maintenance of circulating and tissue resident MAIT cells. We also explore how microbial dysbiosis, alongside changes in intestinal permeability and imbalance between pro- and anti-inflammatory components of the immune response are together involved in the potential pathogenicity of MAIT cells. Whilst there have been significant improvements in our understanding of how the microbiota shapes MAIT cell function, human data are relatively lacking, and it remains unknown if MAIT cells can conversely influence the composition of the microbiota. We speculate whether, in a human population, differences in microbiomes might account for the heterogeneity observed in MAIT cell frequency across mucosal sites or between individuals, and response to therapies targeting T cells. Moreover, we speculate whether manipulation of the microbiota, or harnessing MAIT cell ligands within the gut or disease-specific sites could offer novel therapeutic strategies.
Collapse
Affiliation(s)
- Maisha F. Jabeen
- Respiratory Medicine Unit, Experimental Medicine Division, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Timothy S. C. Hinks
- Respiratory Medicine Unit, Experimental Medicine Division, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
24
|
Mincham KT, Snelgrove RJ. OMIP-086: Full spectrum flow cytometry for high-dimensional immunophenotyping of mouse innate lymphoid cells. Cytometry A 2023; 103:110-116. [PMID: 36331092 PMCID: PMC10953369 DOI: 10.1002/cyto.a.24702] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 10/17/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
This 25-parameter, 22-color full spectrum flow cytometry panel was designed and optimized for the comprehensive enumeration and functional characterization of innate lymphoid cell (ILC) subsets in mouse tissues. The panel presented here allows the discrimination of ILC progenitors (ILCP), ILC1, ILC2, NCR+ ILC3, NCR- ILC3, CCR6+ lymphoid tissue-inducer (LTi)-like ILC3 and mature natural killer (NK) cell populations. Further characterization of ILC and NK cell functional profiles in response to stimulation is provided by the inclusion of subset-specific cytokine markers, and proliferation markers. Development and optimization of this panel was performed on freshly isolated cells from adult BALB/c lungs and small intestine lamina propria, and ex vivo stimulation with phorbol 12-myrisate 13-acetate, ionomycin, and pro-ILC activating cytokines.
Collapse
Affiliation(s)
- Kyle T. Mincham
- National Heart and Lung InstituteImperial College LondonLondonUK
| | | |
Collapse
|
25
|
van de Pavert SA. Layered origins of lymphoid tissue inducer cells. Immunol Rev 2023; 315:71-78. [PMID: 36705244 DOI: 10.1111/imr.13189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The Innate Lymphoid Cell (ILC) family is a relatively recently described immune cell family involved in innate immune responses and tissue homeostasis. Lymphoid Tissue Inducer (LTi) cells are part of the type 3 (ILC3) family. The ILC3 family is the main ILC population within the embryo, in which the LTi cells are critically associated with embryonic lymph node formation. Recent studies have shown more insights in ILC origin and residency from local embryonic and tissue resident precursors. Embryonic LTi cells originating from a different hemogenic endothelial source were shown to be replaced by HSC derived progenitors in adult. This review will discuss the layered origin of the ILC3 family with an emphasis on the LTi cell lineage.
Collapse
Affiliation(s)
- Serge A van de Pavert
- Aix-Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| |
Collapse
|
26
|
Fernandes MB, Barata JT. IL-7 and IL-7R in health and disease: An update through COVID times. Adv Biol Regul 2023; 87:100940. [PMID: 36503870 DOI: 10.1016/j.jbior.2022.100940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
The role of IL-7 and IL-7R for normal lymphoid development and an adequately functioning immune system has been recognized for long, with severe immune deficiency and lymphoid leukemia as extreme examples of the consequences of deregulation of the IL-7-IL-7R axis. In this review, we provide an update (focusing on the past couple of years) on IL-7 and IL-7R in health and disease. We highlight the findings on IL-7/IL-7R signaling mechanisms and the, sometimes controversial, impact of IL-7 and its receptor on leukocyte biology, COVID-19, acute lymphoblastic leukemia, and different solid tumors, as well as their relevance as therapeutic tools or targets.
Collapse
Affiliation(s)
- Marta B Fernandes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisbon, Portugal.
| |
Collapse
|
27
|
Donor natural killer cells trigger production of β-2-microglobulin to enhance post-bone marrow transplant immunity. Blood 2022; 140:2323-2334. [PMID: 35984965 DOI: 10.1182/blood.2021015297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 07/14/2022] [Indexed: 11/20/2022] Open
Abstract
Allogeneic hematopoietic transplantation is a powerful treatment for hematologic malignancies. Posttransplant immune incompetence exposes patients to disease relapse and infections. We previously demonstrated that donor alloreactive natural killer (NK) cells ablate recipient hematopoietic targets, including leukemia. Here, in murine models, we show that infusion of donor alloreactive NK cells triggers recipient dendritic cells (DCs) to synthesize β-2-microglobulin (B2M) that elicits the release of c-KIT ligand and interleukin-7 that greatly accelerate posttransplant immune reconstitution. An identical chain of events was reproduced by infusing supernatants of alloreactive NK/DC cocultures. Similarly, human alloreactive NK cells triggered human DCs to synthesize B2M that induced interleukin-7 production by thymic epithelial cells and thereby supported thymocyte cellularity in vitro. Chromatography fractionation of murine and human alloreactive NK/DC coculture supernatants identified a protein with molecular weight and isoelectric point of B2M, and mass spectrometry identified amino acid sequences specific of B2M. Anti-B2M antibody depletion of NK/DC coculture supernatants abrogated their immune-rebuilding effect. B2M knock-out mice were unable to undergo accelerated immune reconstitution, but infusion of (wild-type) NK/DC coculture supernatants restored their ability to undergo accelerated immune reconstitution. Similarly, silencing the B2M gene in human DCs, before coculture with alloreactive NK cells, prevented the increase in thymocyte cellularity in vitro. Finally, human recombinant B2M increased thymocyte cellularity in a thymic epithelial cells/thymocyte culture system. Our studies uncover a novel therapeutic principle for treating posttransplant immune incompetence and suggest that, upon its translation to the clinic, patients may benefit from adoptive transfer of large numbers of cytokine-activated, ex vivo-expanded donor alloreactive NK cells.
Collapse
|
28
|
Taylor CA, Watson RA, Tong O, Ye W, Nassiri I, Gilchrist JJ, de Los Aires AV, Sharma PK, Koturan S, Cooper RA, Woodcock VK, Jungkurth E, Shine B, Coupe N, Payne MJ, Church DN, Naranbhai V, Groha S, Emery P, Mankia K, Freedman ML, Choueiri TK, Middleton MR, Gusev A, Fairfax BP. IL7 genetic variation and toxicity to immune checkpoint blockade in patients with melanoma. Nat Med 2022; 28:2592-2600. [PMID: 36526722 PMCID: PMC9800275 DOI: 10.1038/s41591-022-02095-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/18/2022] [Indexed: 12/23/2022]
Abstract
Treatment with immune checkpoint blockade (ICB) frequently triggers immune-related adverse events (irAEs), causing considerable morbidity. In 214 patients receiving ICB for melanoma, we observed increased severe irAE risk in minor allele carriers of rs16906115, intronic to IL7. We found that rs16906115 forms a B cell-specific expression quantitative trait locus (eQTL) to IL7 in patients. Patients carrying the risk allele demonstrate increased pre-treatment B cell IL7 expression, which independently associates with irAE risk, divergent immunoglobulin expression and more B cell receptor mutations. Consistent with the role of IL-7 in T cell development, risk allele carriers have distinct ICB-induced CD8+ T cell subset responses, skewing of T cell clonality and greater proportional repertoire occupancy by large clones. Finally, analysis of TCGA data suggests that risk allele carriers independently have improved melanoma survival. These observations highlight key roles for B cells and IL-7 in both ICB response and toxicity and clinical outcomes in melanoma.
Collapse
Affiliation(s)
- Chelsea A Taylor
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Robert A Watson
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| | - Orion Tong
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Weiyu Ye
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Isar Nassiri
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - James J Gilchrist
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Alba Verge de Los Aires
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Piyush Kumar Sharma
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Surya Koturan
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Rosalin A Cooper
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Victoria K Woodcock
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| | - Elsita Jungkurth
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Brian Shine
- Department of Clinical Biochemistry, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Nicholas Coupe
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| | - Miranda J Payne
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| | - David N Church
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Vivek Naranbhai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Center for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Stefan Groha
- Department of Medical Oncology, Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard & MIT, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- National Institute for Health Research Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Kulveer Mankia
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- National Institute for Health Research Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Matthew L Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard & MIT, Cambridge, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard & MIT, Cambridge, MA, USA
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Mark R Middleton
- Department of Oncology, University of Oxford, Oxford, UK
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Alexander Gusev
- Department of Medical Oncology, Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard & MIT, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
| | - Benjamin P Fairfax
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
- Department of Oncology, University of Oxford, Oxford, UK.
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
29
|
Kim KS. Regulation of T cell repertoires by commensal microbiota. Front Cell Infect Microbiol 2022; 12:1004339. [PMID: 36310871 PMCID: PMC9606468 DOI: 10.3389/fcimb.2022.1004339] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
The gut microbiota plays an important role in regulating the host immune systems. It is well established that various commensal microbial species can induce the differentiation of CD4+ T helper subsets such as Foxp3+ regulatory T (Treg) cells and Th17 cells in antigen-dependent manner. The ability of certain microbial species to induce either Treg cells or Th17 cells is often linked to the altered susceptibility to certain immune disorders that are provoked by aberrant T cell response against self-antigens. These findings raise an important question as to how gut microbiota can regulate T cell repertoire and the activation of autoreactive T cells. This review will highlight microbiota-dependent regulation of thymic T cell development, maintenance of T cell repertoire in the secondary lymphoid tissues and the intestine, and microbiota-mediated modulation of autoreactive and tumor neoantigen-specific T cells in autoimmune diseases and tumors, respectively.
Collapse
|
30
|
Cuéllar C, Rodero M, Pérez-Griera J, Galindo-Regal L, Lopez-Chulia F, García-Ballesteros C, Carlos Andreu-Ballester J. Association between anti-Anisakis simplex antibodies and interleukin-7 levels. Int Immunopharmacol 2022; 111:109134. [PMID: 35964412 DOI: 10.1016/j.intimp.2022.109134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/27/2022] [Accepted: 08/03/2022] [Indexed: 11/18/2022]
Abstract
IL-7 is a crucial factor for the development of lymphocytes, and it is absolutely necessary for γδ T cells. Mice deficient in L-7 have a deficit of B and αβ T lymphocytes, and an absence of mature γδ TCR cells. IL-7 is essential for the survival, development and maturation of Schistosoma sp., although its production is associated with protection against intestinal helminths. The presence of anti-Anisakis simplex antibodies, especially IgA, is related to a lower frequency in CD3 + CD56 + αβ + lymphocytes and all subpopulations of γδ T cells. In this work, the relationship of IL-7 with humoral and cellular responses against A. simplex in 100 healthy subjects was studied. We have found significantly higher IL-7 levels in anti-A. simplex IgA-positive subjects (p < 0.001). The positivity of anti-A. simplex IgA was associated with a significant reduction in the frequency of CD3 + αβ+ (p < 0.01), CD3 + CD4 + αβ+, CD3 + CD8 + αβ+, CD3 + CD56 + αβ+, CD3 + γδ+, CD3 + CD4-CD8-γδ+ and CD3 + CD56 + γδ+ (p < 0.05) cells. In the case of NKT cells, this same phenomenon was also associated with IgE positivity. There was a weak inverse correlation (Spearman) of IL-7 levels with the frequencies of CD3 + CD4 + αβ+ (-0.125, p = 0.047), CD3 + CD8 + αβ+ (-0.204, p = 0.032), CD3 + CD56 + αβ+ (-0.247, p = 0.007), CD3 + γδ+ (-0.267, p = 0.007), CD3 + CD4-CD8-γδ+ (-0.266, p = 0.003), and CD3 + CD8 + γδ + (-0.302, p = 0.002) cells. The role of NKT cells in the anti-A. simplex response was confirmed and an association between IL and 7 levels and specific antibodies, especially IgA, was demonstrated. The higher production of IL-7 would represent a compensatory mechanism in response to the reduction in lymphocyte populations associated with the response against this parasite.
Collapse
Affiliation(s)
- Carmen Cuéllar
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain.
| | - Marta Rodero
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | - Jaime Pérez-Griera
- Departamento de Biopatología, Hospital Clínico Universitario, 46010 Valencia, Spain
| | - Lorena Galindo-Regal
- Laboratorio de Biología Molecular, Hospital Arnau de Vilanova, 46015 Valencia, Spain
| | | | | | | |
Collapse
|
31
|
Tufa A, Gebremariam TH, Manyazewal T, Getinet T, Webb DL, Hellström PM, Genet S. Inflammatory mediators profile in patients hospitalized with COVID-19: A comparative study. Front Immunol 2022; 13:964179. [PMID: 35958594 PMCID: PMC9359079 DOI: 10.3389/fimmu.2022.964179] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/01/2022] [Indexed: 01/08/2023] Open
Abstract
Abnormal inflammatory mediator concentrations during SARS-CoV-2 infection may represent disease severity. We aimed to assess plasma inflammatory mediator concentrations in patients with SARS-CoV-2 in Addis Ababa, Ethiopia. In this study, 260 adults: 126 hospitalized patients with confirmed COVID-19 sorted into severity groups: severe (n=68) and mild or moderate (n=58), and 134 healthy controls were enrolled. We quantified 39 plasma inflammatory mediators using multiplex ELISA. Spearman rank correlation and Mann-Whitney U test were used to identify mechanistically coupled inflammatory mediators and compare disease severity. Compared to healthy controls, patients with COVID-19 had significantly higher levels of interleukins 1α, 2, 6, 7, 8, 10 and 15, C-reactive protein (CRP), serum amyloid A (SAA), intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion protein 1 (VCAM-1), IFN-γ-inducible protein-10 (IP-10, CXCL10), macrophage inflammatory protein-1 alpha (MIP-1α, CCL3), eotaxin-3 (CCL26), interferon-gamma (IFN-γ), tumor necrosis factor-α (TNF-α), basic fibroblast growth factor (bFGF), placental growth factor (PlGF), and fms-like tyrosine kinase 1 (Flt-1). Patients with severe COVID-19 had higher IL-10 and lower macrophage-derived chemokine (MDC, CCL22) compared to the mild or moderate group (P<0.05). In the receiver operating characteristic curve, SAA, IL-6 and CRP showed strong sensitivity and specificity in predicting the severity and prognosis of COVID-19. Greater age and higher CRP had a significant association with disease severity (P<0.05). Our findings reveal that CRP, SAA, VCAM-1, CXCL10, CCL22 and IL-10 levels are promising biomarkers for COVID-19 disease severity, suggesting that plasma inflammatory mediators could be used as warning indicators of COVID-19 severity, aid in COVID-19 prognosis and treatment.
Collapse
Affiliation(s)
- Abdisa Tufa
- Department of Medical Biochemistry, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Tewodros Haile Gebremariam
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Tsegahun Manyazewal
- Centre for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Tewodros Getinet
- School of Public Health, Saint Paul’s Hospital Millennium Medical College, Addis Ababa, Ethiopia
| | - Dominic-Luc Webb
- Gastroenterology and Hepatology Unit, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Per M. Hellström
- Gastroenterology and Hepatology Unit, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Solomon Genet
- Department of Medical Biochemistry, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
32
|
Kwok T, Medovich SC, Silva-Junior IA, Brown EM, Haug JC, Barrios MR, Morris KA, Lancaster JN. Age-Associated Changes to Lymph Node Fibroblastic Reticular Cells. FRONTIERS IN AGING 2022; 3:838943. [PMID: 35821826 PMCID: PMC9261404 DOI: 10.3389/fragi.2022.838943] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/07/2022] [Indexed: 12/26/2022]
Abstract
The decreased proportion of antigen-inexperienced, naïve T cells is a hallmark of aging in both humans and mice, and contributes to reduced immune responses, particularly against novel and re-emerging pathogens. Naïve T cells depend on survival signals received during their circulation among the lymph nodes by direct contacts with stroma, in particular fibroblastic reticular cells. Macroscopic changes to the architecture of the lymph nodes have been described, but it is unclear how lymph node stroma are altered with age, and whether these changes contribute to reduced naïve T cell maintenance. Here, using 2-photon microscopy, we determined that the aged lymph node displayed increased fibrosis and correspondingly, that naïve T-cell motility was impaired in the aged lymph node, especially in proximity to fibrotic deposition. Functionally, adoptively transferred young naïve T-cells exhibited reduced homeostatic turnover in aged hosts, supporting the role of T cell-extrinsic mechanisms that regulate their survival. Further, we determined that early development of resident fibroblastic reticular cells was impaired, which may correlate to the declining levels of naïve T-cell homeostatic factors observed in aged lymph nodes. Thus, our study addresses the controversy as to whether aging impacts the composition lymph node stroma and supports a model in which impaired differentiation of lymph node fibroblasts and increased fibrosis inhibits the interactions necessary for naïve T cell homeostasis.
Collapse
Affiliation(s)
- Tina Kwok
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | | | | | - Elise M Brown
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | - Joel C Haug
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | | | - Karina A Morris
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, United States
| | | |
Collapse
|
33
|
Kawabe T, Ciucci T, Kim KS, Tayama S, Kawajiri A, Suzuki T, Tanaka R, Ishii N, Jankovic D, Zhu J, Sprent J, Bosselut R, Sher A. Redefining the Foreign Antigen and Self-Driven Memory CD4 + T-Cell Compartments via Transcriptomic, Phenotypic, and Functional Analyses. Front Immunol 2022; 13:870542. [PMID: 35707543 PMCID: PMC9190281 DOI: 10.3389/fimmu.2022.870542] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/29/2022] [Indexed: 01/03/2023] Open
Abstract
Under steady-state conditions, conventional CD4+ T lymphocytes are classically divided into naïve (CD44lo CD62Lhi) and memory (CD44hi CD62Llo) cell compartments. While the latter population is presumed to comprise a mixture of distinct subpopulations of explicit foreign antigen (Ag)-specific “authentic” memory and foreign Ag-independent memory-phenotype (MP) cells, phenotypic markers differentially expressed in these two cell types have yet to be identified. Moreover, while MP cells themselves have been previously described as heterogeneous, it is unknown whether they consist of distinct subsets defined by marker expression. In this study, we demonstrate using combined single-cell RNA sequencing and flow cytometric approaches that self-driven MP CD4+ T lymphocytes are divided into CD127hi Sca1lo, CD127hi Sca1hi, CD127lo Sca1hi, and CD127lo Sca1lo subpopulations that are Bcl2lo, while foreign Ag-specific memory cells are CD127hi Sca1hi Bcl2hi. We further show that among the four MP subsets, CD127hi Sca1hi lymphocytes represent the most mature and cell division-experienced subpopulation derived from peripheral naïve precursors. Finally, we provide evidence arguing that this MP subpopulation exerts the highest responsiveness to Th1-differentiating cytokines and can induce colitis. Together, our findings define MP CD4+ T lymphocytes as a unique, self-driven population consisting of distinct subsets that differ from conventional foreign Ag-specific memory cells in marker expression and establish functional relevance for the mature subset of CD127hi Sca1hi MP cells.
Collapse
Affiliation(s)
- Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Thomas Ciucci
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.,David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, NY, United States
| | - Kwang Soon Kim
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, South Korea
| | - Shunichi Tayama
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akihisa Kawajiri
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takumi Suzuki
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Riou Tanaka
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Dragana Jankovic
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jonathan Sprent
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Rémy Bosselut
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
34
|
Hasanvand A. COVID-19 and the role of cytokines in this disease. Inflammopharmacology 2022; 30:789-798. [PMID: 35505267 PMCID: PMC9064717 DOI: 10.1007/s10787-022-00992-2] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 04/05/2022] [Indexed: 12/13/2022]
Abstract
Studies have shown that SARS-CoV-2 has the ability to activate and mature proinflammatory cytokines in the body. Cytokine markers are a group of polypeptide signalling molecules that can induce and regulate many cellular biological processes by stimulating cell receptors at the surface. SARS-CoV-2 has been shown to be associated with activation of innate immunity, and an increase in neutrophils, mononuclear phagocytes, and natural killer cells has been observed, as well as a decrease in T cells including CD4+ and CD8. It is noteworthy that during the SARS-CoV-2 infection, an increase in the secretion or production of IL-6 and IL-8 is seen in COVID-19 patients along with a decrease in CD4+ and CD8+ and T cells in general. SARS-CoV-2 has been shown to significantly increase Th2, Th1/Th17 cells and antibody production in the body of patients with COVID-19. Specific immune profiles of SARS-CoV-2 infection can lead to secondary infections and dysfunction of various organs in the body. It has been shown that Interleukins (such as IL-1, IL-4, IL-6, IL-7, IL-10, IL-12, IL-17, and IL-18), IFN-γ, TNF-α,TGF-β and NF-κB play major roles in the body's inflammatory response to SARS-CoV-2 infection. The most important goal of this review is to study the role of inflammatory cytokines in COVID-19.
Collapse
Affiliation(s)
- Amin Hasanvand
- Department of Physiology and Pharmacology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
35
|
Crosstalk between ILC2s and Th2 CD4+ T Cells in Lung Disease. J Immunol Res 2022; 2022:8871037. [PMID: 35592688 PMCID: PMC9113865 DOI: 10.1155/2022/8871037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/30/2022] [Accepted: 04/18/2022] [Indexed: 12/03/2022] Open
Abstract
Cytokine secretion, such as interleukin-4 (IL-4), IL-5, IL-9, IL-13, and amphiregulin (Areg), by type 2 innate lymphoid cells (ILC2s) is indispensable for homeostasis, remodeling/repairing tissue structure, inflammation, and tumor immunity. Often viewed as the innate cell surrogate of T helper type 2 (Th2) cells, ILC2s not only secrete the same type 2 cytokines, but are also inextricably related to CD4+T cells in terms of cell origin and regulatory factors, bridging between innate and adaptive immunity. ILC2s interact with CD4+T cells to play a leading role in a variety of diseases through secretory factors. Here, we review the latest progress on ILC2s and CD4+T cells in the lung, the close relationship between the two, and their relevance in the lung disease and immunity. This literature review aids future research in pulmonary type 2 immune diseases and guides innovative treatment approaches for these diseases.
Collapse
|
36
|
Schroeder JH, Howard JK, Lord GM. Transcription factor-driven regulation of ILC1 and ILC3. Trends Immunol 2022; 43:564-579. [PMID: 35618586 PMCID: PMC10166716 DOI: 10.1016/j.it.2022.04.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 10/18/2022]
Abstract
Mammalian innate lymphoid cells (ILCs) have functional relevance under both homeostatic and disease settings, such as inflammatory bowel disease (IBD), particularly in the context of maintaining the integrity of mucosal surfaces. Early reports highlighted group 1 and 3 ILC regulatory transcription factors (TFs), T-box expressed in T cells (T-bet; Tbx21) and RAR-related orphan nuclear receptor γt (RORγt; Rorc), as key regulators of ILC biology. Since then, other canonical TFs have been shown to have a role in the development and function of ILC subsets. In this review, we focus on recent insights into the balance between mature ILC1 and ILC3 based on these TFs and how they interact with other key cell-intrinsic molecular pathways. We outline how this TF interplay might be explored to identify novel candidate therapeutic avenues for human diseases.
Collapse
|
37
|
Sugimura R, Wang CY. The Role of Innate Lymphoid Cells in Cancer Development and Immunotherapy. Front Cell Dev Biol 2022; 10:803563. [PMID: 35557940 PMCID: PMC9086356 DOI: 10.3389/fcell.2022.803563] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/11/2022] [Indexed: 11/29/2022] Open
Abstract
Innate Lymphoid Cells (ILCs) are an elusive type of innate immune cell that was only discovered recently. Their tissue residency and dependency makes them a niche group of cells that bridge the adaptive and innate immune system. The nomenclature and classification of ILCs have been challenging due to their heterogeneity. The currently agreed ILC classification splits the cells into two categories including cytotoxic and helper ILCs. The tumour microenvironment is often hostile for immune cells. Remodeling the microenvironment and regulating other immune cells—achieved by ILCs-can enhance anti-tumor effects. How ILCs regulate other immune cells in the tumor microenvironment remains to be understood. Here we review current understanding of the role of ILCs in the tumor microenvironment. ILCs recruit CD8 positive T and memory T cells in PDAC, ILCs are also able to help CD108 positive B cells migrate toward tumour locations. In NSCLC, ILC3s are seen helping resident macrophages enhancing the mucus immunity to cancer cells. We then highlight the roles of cytokines and immune checkpoint pathways in ILCs and its implication in immunotherapy.
Collapse
|
38
|
Worthington AK, Cool T, Poscablo DM, Hussaini A, Beaudin AE, Forsberg EC. IL7Rα, but not Flk2, is required for hematopoietic stem cell reconstitution of tissue-resident lymphoid cells. Development 2022; 149:274067. [PMID: 35072209 PMCID: PMC8917444 DOI: 10.1242/dev.200139] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022]
Abstract
Tissue-resident lymphoid cells (TLCs) span the spectrum of innate-to-adaptive immune function. Unlike traditional, circulating lymphocytes that are continuously generated from hematopoietic stem cells (HSCs), many TLCs are of fetal origin and poorly generated from adult HSCs. Here, we sought to further understand murine TLC development and the roles of Flk2 and IL7Rα, two cytokine receptors with known function in traditional lymphopoiesis. Using Flk2- and Il7r-Cre lineage tracing, we found that peritoneal B1a cells, splenic marginal zone B (MZB) cells, lung ILC2s and regulatory T cells (Tregs) were highly labeled. Despite high labeling, loss of Flk2 minimally affected the generation of these cells. In contrast, loss of IL7Rα, or combined deletion of Flk2 and IL7Rα, dramatically reduced the number of B1a cells, MZBs, ILC2s and Tregs, both in situ and upon transplantation, indicating an intrinsic and essential role for IL7Rα. Surprisingly, reciprocal transplants of wild-type HSCs showed that an IL7Rα−/− environment selectively impaired reconstitution of TLCs when compared with TLC numbers in situ. Taken together, our data defined Flk2- and IL7Rα-positive TLC differentiation paths, and revealed functional roles of Flk2 and IL7Rα in TLC establishment. Summary: Tissue-resident lymphoid cells develop via IL7Rα-positive progenitors and are repopulated by transplanted adult hematopoietic stem cells; however, such TLC lymphopoiesis cannot be fully rescued in IL7Rα−/− recipient mice.
Collapse
Affiliation(s)
- Atesh K Worthington
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.,Program in Biomedical Science and Engineering: Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Taylor Cool
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.,Program in Biomedical Science and Engineering: Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Donna M Poscablo
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.,Program in Biomedical Science and Engineering: Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Adeel Hussaini
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Anna E Beaudin
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.,Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.,Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
39
|
Mkhikian H, Hayama KL, Khachikyan K, Li C, Zhou RW, Pawling J, Klaus S, Tran PQN, Ly KM, Gong AD, Saryan H, Hai JL, Grigoryan D, Lee PL, Newton BL, Raffatellu M, Dennis JW, Demetriou M. Age-associated impairment of T cell immunity is linked to sex-dimorphic elevation of N-glycan branching. NATURE AGING 2022; 2:231-242. [PMID: 35528547 PMCID: PMC9075523 DOI: 10.1038/s43587-022-00187-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 02/02/2022] [Indexed: 11/08/2022]
Abstract
Impaired T cell immunity with aging increases mortality from infectious disease. The branching of Asparagine-linked glycans is a critical negative regulator of T cell immunity. Here we show that branching increases with age in females more than males, in naïve more than memory T cells, and in CD4+ more than CD8+ T cells. Female sex hormones and thymic output of naïve T cells (TN) decrease with age, however neither thymectomy nor ovariectomy altered branching. Interleukin-7 (IL-7) signaling was increased in old female more than male mouse TN cells, and triggered increased branching. N-acetylglucosamine, a rate-limiting metabolite for branching, increased with age in humans and synergized with IL-7 to raise branching. Reversing elevated branching rejuvenated T cell function and reduced severity of Salmonella infection in old female mice. These data suggest sex-dimorphic antagonistic pleiotropy, where IL-7 initially benefits immunity through TN maintenance but inhibits TN function by raising branching synergistically with age-dependent increases in N-acetylglucosamine.
Collapse
Affiliation(s)
- Haik Mkhikian
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Ken L Hayama
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
| | - Khachik Khachikyan
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Carey Li
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Raymond W Zhou
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Judy Pawling
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Suzi Klaus
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
| | - Phuong Q N Tran
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Kim M Ly
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Andrew D Gong
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Hayk Saryan
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Jasper L Hai
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - David Grigoryan
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Philip L Lee
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Barbara L Newton
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Manuela Raffatellu
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Center for Mucosal Immunology, Allergy, and Vaccines, Chiba University-UC San Diego, La Jolla, CA, USA
| | - James W Dennis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Michael Demetriou
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA, USA.
- Department of Neurology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
40
|
Sheikh A, Jackson J, Shim HB, Yau C, Seo JH, Abraham N. Selective dependence on IL-7 for antigen-specific CD8 T cell responses during airway influenza infection. Sci Rep 2022; 12:135. [PMID: 34997007 PMCID: PMC8741933 DOI: 10.1038/s41598-021-03936-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/06/2021] [Indexed: 11/08/2022] Open
Abstract
Interleukin-7 (IL-7) is a cytokine known for its importance in T cell development and survival. How IL-7 shapes CD8 T cell responses during an acute viral infection is less understood. We had previously shown that IL-7 signaling deficient mice have reduced accumulation of influenza-specific CD8 T cells following influenza infection. We sought to determine whether IL-7 affects early CD8 T cell expansion in the mediastinal lymph node and effector function in the lungs. Using IL-7Rα signaling deficient mice, we show that IL-7 is required for a normal sized mediastinal lymph node and the early clonal expansion of influenza-specific CD8 T cells therein. We show that IL-7 plays a cell-intrinsic role in the accumulation of NP366-374 and PA224-233-specific CD8 T cells in the lymph node. We also found that IL-7 shapes terminal differentiation, degranulation and cytokine production to a greater extent in PA224-233-specific than NP366-374-specific CD8 T cells. We further demonstrate that IL-7 is induced in the lung tissue by viral infection and we characterize multiple cellular sources that contribute to IL-7 production. Our findings on IL-7 and its effects on lower respiratory diseases will be important for expanding the utility of therapeutics that are currently available.
Collapse
Affiliation(s)
- Abdalla Sheikh
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Jennie Jackson
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Hanjoo Brian Shim
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Clement Yau
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
- Duke-NUS Medical School, 8 College Road, Singapore, Singapore
| | - Jung Hee Seo
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Ninan Abraham
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada.
- Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
41
|
ILC Differentiation in the Thymus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1365:25-39. [DOI: 10.1007/978-981-16-8387-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
42
|
A Validation Study on Immunophenotypic Differences in T-lymphocyte Chromosomal Radiosensitivity between Newborns and Adults in South Africa. RADIATION 2021. [DOI: 10.3390/radiation2010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Children have an increased risk of developing radiation-induced secondary malignancies compared to adults, due to their high radiosensitivity and longer life expectancy. In contrast to the epidemiological evidence, there is only a handful of radiobiology studies which investigate the difference in radiosensitivity between children and adults at a cellular level. In this study, the previous results on the potential age dependency in chromosomal radiosensitivity were validated again by means of the cytokinesis-block micronucleus (CBMN) assay in T-lymphocytes isolated from the umbilical cord and adult peripheral blood of a South African population. The isolated cells were irradiated with 60Co γ-rays at doses ranging from 0.5 Gy to 4 Gy. Increased radiosensitivities of 34%, 42%, 29%, 26% and 16% were observed for newborns compared to adults at 0.5, 1, 2, 3 and 4 Gy, respectively. An immunophenotypic evaluation with flow cytometry revealed a significant change in the fraction of naïve (CD45RA+) T-lymphocytes in CD4+ and CD8+ T-lymphocytes with age. Newborns co-expressed an average of 91.05% CD45RA+ (range: 80.80–98.40%) of their CD4+ cells, while this fraction decreased to an average of 39.08% (range: 12.70–58.90%) for adults. Similar observations were made for CD8+ cells. This agrees with previous published results that the observed differences in chromosomal radiosensitivity between newborn and adult T-lymphocytes could potentially be linked to their immunophenotypic profiles.
Collapse
|
43
|
Kawabe T, Sher A. Memory-phenotype CD4+ T cells: a naturally arising T lymphocyte population possessing innate immune function. Int Immunol 2021; 34:189-196. [PMID: 34897483 PMCID: PMC8962445 DOI: 10.1093/intimm/dxab108] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022] Open
Abstract
In conventional adaptive immune responses, upon recognition of foreign antigens, naive CD4+ T lymphocytes are activated to differentiate into effector/memory cells. In addition, emerging evidence suggests that in the steady state, naive CD4+ T cells spontaneously proliferate in response to self-antigens to acquire a memory phenotype (MP) through homeostatic proliferation. This expansion is particularly profound in lymphopenic environments but also occurs in lymphoreplete, normal conditions. The 'MP T lymphocytes' generated in this manner are maintained by rapid proliferation in the periphery and they tonically differentiate into T-bet-expressing 'MP1' cells. Such MP1 CD4+ T lymphocytes can exert innate effector function, producing IFN-γ in response to IL-12 in the absence of antigen recognition, thereby contributing to host defense. In this review, we will discuss our current understanding of how MP T lymphocytes are generated and persist in steady-state conditions, their populational heterogeneity as well as the evidence for their effector function. We will also compare these properties with those of a similar population of innate memory cells previously identified in the CD8+ T lymphocyte lineage.
Collapse
Affiliation(s)
- Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan,Correspondence to: T. Kawabe; E-mail: or A. Sher; E-mail:
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA,Correspondence to: T. Kawabe; E-mail: or A. Sher; E-mail:
| |
Collapse
|
44
|
Olguín-Martínez E, Ruiz-Medina BE, Licona-Limón P. Tissue-Specific Molecular Markers and Heterogeneity in Type 2 Innate Lymphoid Cells. Front Immunol 2021; 12:757967. [PMID: 34759931 PMCID: PMC8573327 DOI: 10.3389/fimmu.2021.757967] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/21/2021] [Indexed: 12/25/2022] Open
Abstract
Innate lymphoid cells (ILCs) are the most recently described group of lymphoid subpopulations. These tissue-resident cells display a heterogeneity resembling that observed on different groups of T cells, hence their categorization as cytotoxic NK cells and helper ILCs type 1, 2 and 3. Each one of these groups is highly diverse and expresses different markers in a context-dependent manner. Type 2 innate lymphoid cells (ILC2s) are activated in response to helminth parasites and regulate the immune response. They are involved in the etiology of diseases associated with allergic responses as well as in the maintenance of tissue homeostasis. Markers associated with their identification differ depending on the tissue and model used, making the study and understanding of these cells a cumbersome task. This review compiles evidence for the heterogeneity of ILC2s as well as discussion and analyses of molecular markers associated with their identity, function, tissue-dependent expression, and how these markers contribute to the interaction of ILC2s with specific microenvironments to maintain homeostasis or respond to pathogenic challenges.
Collapse
Affiliation(s)
- Enrique Olguín-Martínez
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, Mexico
| | - Blanca E Ruiz-Medina
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, Mexico
| | - Paula Licona-Limón
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, Mexico
| |
Collapse
|
45
|
Bekele Y, Sui Y, Berzofsky JA. IL-7 in SARS-CoV-2 Infection and as a Potential Vaccine Adjuvant. Front Immunol 2021; 12:737406. [PMID: 34603318 PMCID: PMC8484798 DOI: 10.3389/fimmu.2021.737406] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/31/2021] [Indexed: 01/10/2023] Open
Abstract
IL-7/IL-7R signaling is critical for development, maturation, maintenance and survival of many lymphocytes in the thymus and periphery. IL-7 has been used as immunotherapy in pre-clinical and clinical studies to treat cancer, HIV infection and sepsis. Here, we discuss the critical function of IL-7 in diagnosis, prognosis and treatment of COVID-19 patients. We also summarize a promising role of IL-7 as a vaccine adjuvant. It could potentially enhance the immune responses to vaccines especially against SARS-CoV-2 or other new vaccines.
Collapse
Affiliation(s)
- Yonas Bekele
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
46
|
Abstract
Conventional CD4+ and CD8+ T lymphocytes comprise a mixture of naive and memory cells. Generation and survival of these T-cell subsets is under strict homeostatic control and reflects contact with self-major histocompatibility complex (MHC) and certain cytokines. Naive T cells arise in the thymus via T-cell receptor (TCR)-dependent positive selection to self-peptide/MHC complexes and are then maintained in the periphery through self-MHC interaction plus stimulation via interleukin-7 (IL-7). By contrast, memory T cells are largely MHC-independent for their survival but depend strongly on stimulation via cytokines. Whereas typical memory T cells are generated in response to foreign antigens, some arise spontaneously through contact of naive precursors with self-MHC ligands; we refer to these cells as memory-phenotype (MP) T cells. In this review, we discuss the generation and homeostasis of naive T cells and these two types of memory T cells, focusing on their relative interaction with MHC ligands and cytokines.
Collapse
Affiliation(s)
- Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Jaeu Yi
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Jonathan Sprent
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| |
Collapse
|
47
|
Jujube Powder Enhances Cyclophosphamide Efficiency against Murine Colon Cancer by Enriching CD8 + T Cells While Inhibiting Eosinophilia. Nutrients 2021; 13:nu13082700. [PMID: 34444860 PMCID: PMC8401958 DOI: 10.3390/nu13082700] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
Cyclophosphamide (CTX) is widely applied in cancer treatment. However, the outcome is often compromised by lymphopenia, myelosuppression, and gut dysbiosis. Here, we used jujube powder to enhance CTX efficiency through nurturing gut microbiota in order to facilitate favorable metabolisms. It was observed that the oral administration of jujube powder enriched CD8+ T cells in mouse MC38 colon tumor microenvironment and increased the diversity of gut microbiota and the abundance of Bifidobacteriales, which is helpful to the production of butyrate in the cecum content. The application of jujube powder also stimulated the production of white blood cells, especially CD8+ T cells in peripheral and bone marrow, while inhibiting the growth of eosinophils in peripheral blood and the production of IL-7 and GM-CSF in serum. All these are conductive to the significant inhibition of the tumor growth, suggesting the high potential of nurturing gut microbiota with natural products for improving the efficiency of chemotherapy.
Collapse
|
48
|
Vandooren J, Pereira RVS, Ugarte-Berzal E, Rybakin V, Noppen S, Stas MR, Bernaerts E, Ganseman E, Metzemaekers M, Schols D, Proost P, Opdenakker G. Internal Disulfide Bonding and Glycosylation of Interleukin-7 Protect Against Proteolytic Inactivation by Neutrophil Metalloproteinases and Serine Proteases. Front Immunol 2021; 12:701739. [PMID: 34276694 PMCID: PMC8278288 DOI: 10.3389/fimmu.2021.701739] [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: 04/28/2021] [Accepted: 06/14/2021] [Indexed: 11/13/2022] Open
Abstract
Interleukin 7 (IL-7) is a cell growth factor with a central role in normal T cell development, survival and differentiation. The lack of IL-7–IL-7 receptor(R)-mediated signaling compromises lymphoid development, whereas increased signaling activity contributes to the development of chronic inflammation, cancer and autoimmunity. Gain-of-function alterations of the IL-7R and the signaling through Janus kinases (JAKs) and signal transducers and activators of transcription (STATs) are enriched in T cell acute lymphoblastic leukemia (T-ALL) and autocrine production of IL-7 by T-ALL cells is involved in the phenotypes of leukemic initiation and oncogenic spreading. Several IL-7-associated pathologies are also characterized by increased presence of matrix metalloproteinase-9 (MMP-9), due to neutrophil degranulation and its regulated production by other cell types. Since proteases secreted by neutrophils are known to modulate the activity of many cytokines, we investigated the interactions between IL-7, MMP-9 and several other neutrophil-derived proteases. We demonstrated that MMP-9 efficiently cleaved human IL-7 in the exposed loop between the α-helices C and D and that this process is delayed by IL-7 N-linked glycosylation. Functionally, the proteolytic cleavage of IL-7 did not influence IL-7Rα binding and internalization nor the direct pro-proliferative effects of IL-7 on a T-ALL cell line (HPB-ALL) or in primary CD8+ human peripheral blood mononuclear cells. A comparable effect was observed for the neutrophil serine proteases neutrophil elastase, proteinase 3 and combinations of neutrophil proteases. Hence, glycosylation and disulfide bonding as two posttranslational modifications influence IL-7 bioavailability in the human species: glycosylation protects against proteolysis, whereas internal cysteine bridging under physiological redox state keeps the IL-7 conformations as active proteoforms. Finally, we showed that mouse IL-7 does not contain the protease-sensitive loop and, consequently, was not cleaved by MMP-9. With the latter finding we discovered differences in IL-7 biology between the human and mouse species.
Collapse
Affiliation(s)
- Jennifer Vandooren
- Laboratory of Immunobiology, Rega Institute for Medical Research/KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Rafaela Vaz Sousa Pereira
- Laboratory of Immunobiology, Rega Institute for Medical Research/KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Estefania Ugarte-Berzal
- Laboratory of Immunobiology, Rega Institute for Medical Research/KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Vasily Rybakin
- Laboratory of Immunobiology, Rega Institute for Medical Research/KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Sam Noppen
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research/KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Melissa R Stas
- Laboratory of Immunobiology, Rega Institute for Medical Research/KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Eline Bernaerts
- Laboratory of Immunobiology, Rega Institute for Medical Research/KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Eva Ganseman
- Laboratory of Molecular Immunology, Rega Institute for Medical Research/KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Mieke Metzemaekers
- Laboratory of Molecular Immunology, Rega Institute for Medical Research/KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research/KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Rega Institute for Medical Research/KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Rega Institute for Medical Research/KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| |
Collapse
|
49
|
Xu H, Cai L, Li Z, Zhang L, Wang G, Xie R, Jiang Y, Yuan Y, Nie H. Dual effect of IL-7/IL-7R signalling on the osteoimmunological system: a potential therapeutic target for rheumatoid arthritis. Immunology 2021; 164:161-172. [PMID: 33934341 DOI: 10.1111/imm.13351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 03/08/2021] [Accepted: 03/30/2021] [Indexed: 12/18/2022] Open
Abstract
The IL-7/IL-7R pathway plays a vital role in the immune system, especially in the inflammatory response. Monocytes/macrophages (osteoclast precursors) have been recently recognized as important participants in the osteoclastogenesis of rheumatoid arthritis (RA) patients. Here, we aimed to investigate the therapeutic potential of IL-7/IL-7R pathway in RA and to determine whether it could restrain osteoclastogenic functions and therefore ameliorate RA. Firstly, collagen-induced arthritis (CIA) mice were administered with IL-7Rα-target antibodies to assess their therapeutic effect on arthritis. We found that blockade of the IL-7/IL-7R pathway protected CIA mice from bone destruction in addition to inducing inflammatory remission, by altering the RANKL/RANK/OPG ratio and consequently decreasing osteoclast formation. To explore the effect and mechanism of this pathway, bone marrow cells were induced to osteoclasts and treated with IL-7, a STAT5 inhibitor or supernatants from T cells. The results showed that the IL-7/IL-7R pathway played a direct inhibitory role in osteoclast differentiation via STAT5 signalling pathway in a RANKL-induced manner. We applied flow cytometry to analyse the effect of IL-7 on T-cell RANKL expression and found that IL-7/IL-7R pathway had an indirect role in the osteoclast differentiation process by enhancing the RANKL expression on T cells. In conclusion, the IL-7/IL-7R pathway exhibited a dual effect on osteoclastogenesis of CIA mice by interacting with osteoimmunology processes and could be a novel therapeutic target for autoimmune diseases such as RA.
Collapse
Affiliation(s)
- Haiyan Xu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Cai
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zijian Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lili Zhang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guojue Wang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rongli Xie
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of General Surgery, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongshuai Jiang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyang Yuan
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Nie
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
50
|
Cruz-Garcia L, Badie C, Anbalagan S, Moquet J, Gothard L, O'Brien G, Somaiah N, Ainsbury EA. An ionising radiation-induced specific transcriptional signature of inflammation-associated genes in whole blood from radiotherapy patients: a pilot study. Radiat Oncol 2021; 16:83. [PMID: 33941218 PMCID: PMC8094544 DOI: 10.1186/s13014-021-01807-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/13/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND This communication reports the identification of a new panel of transcriptional changes in inflammation-associated genes observed in response to ionising radiation received by radiotherapy patients. METHODS Peripheral blood samples were taken with ethical approval and informed consent from a total of 20 patients undergoing external beam radiotherapy for breast, lung, gastrointestinal or genitourinary tumours. Nanostring nCounter analysis of transcriptional changes was carried out in samples prior and 24 h post-delivery of the 1st radiotherapy fraction, just prior to the 5th or 6th fraction, and just before the last fraction. RESULTS Statistical analysis with BRB-ArrayTools, GLM MANOVA and nSolver, revealed a radiation responsive panel of genes which varied by patient group (type of cancer) and with time since exposure (as an analogue for dose received), which may be useful as a biomarker of radiation response. CONCLUSION Further validation in a wider group of patients is ongoing, together with work towards a full understanding of patient specific responses in support of personalised approaches to radiation medicine.
Collapse
Affiliation(s)
| | - Christophe Badie
- PHE CRCE, Chilton, Didcot, Oxford, OX11 0RQ, UK
- Environmental Research Group within the School of Public Health, Faculty of Medicine at Imperial College of Science, Technology and Medicine, London, UK
| | - Selvakumar Anbalagan
- Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, London, SM2 5NG, UK
| | | | - Lone Gothard
- Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, London, SM2 5NG, UK
| | | | - Navita Somaiah
- Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, London, SM2 5NG, UK
| | - Elizabeth A Ainsbury
- PHE CRCE, Chilton, Didcot, Oxford, OX11 0RQ, UK.
- Environmental Research Group within the School of Public Health, Faculty of Medicine at Imperial College of Science, Technology and Medicine, London, UK.
| |
Collapse
|