1
|
Fu Y, Wen Z, Fan J. Interaction of low-density neutrophils with other immune cells in the mechanism of inflammation. Mol Med 2025; 31:133. [PMID: 40205584 PMCID: PMC11983930 DOI: 10.1186/s10020-025-01187-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
Low-density neutrophils (LDNs) are a unique subpopulation of neutrophils, play a significant role in regulating innate and adaptive immunity in various inflammation-related diseases. Emerging evidence suggests that LDNs play a significant role in the pathogenesis and progression of various diseases, including infections, autoimmune disorders, and cancer. In this review, we address the origin, development, and heterogeneity of LDNs, and the roles of LDNs in system homeostasis and diseases. We will focus on the findings of the interaction between LDNs and other immune cells. We will then discuss potential novel therapeutic strategies of intervention in diseases by targeting LDNs.
Collapse
Affiliation(s)
- Yu Fu
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, 15213, USA.
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Jie Fan
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, 15213, USA.
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA.
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, 15213, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
2
|
Cieslak SG, Shahbazi R. Gamma delta T cells and their immunotherapeutic potential in cancer. Biomark Res 2025; 13:51. [PMID: 40148988 PMCID: PMC11951843 DOI: 10.1186/s40364-025-00762-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Gamma-delta (γδ) T cells are a unique subset of T lymphocytes that play diverse roles in immune responses, bridging innate and adaptive immunity. With growing interest in their potential for cancer immunotherapy, a comprehensive and inclusive exploration of γδ T cell families, their development, activation mechanisms, functions, therapeutic implications, and current treatments is essential. This review aims to provide an inclusive and thorough discussion of these topics. Through our discussion, we seek to uncover insights that may harbinger innovative immunotherapeutic strategies. Beginning with an overview of γδ T cell families including Vδ1, Vδ2, and Vδ3, this review highlights their distinct functional properties and contributions to anti-tumor immunity. Despite γδ T cells exhibiting both anti-tumor and pro-tumor activities, our review elucidates strategies to harness the anti-tumor potential of γδ T cells for therapeutic benefit. Moreover, our paper discusses the structural intricacies of the γδ T cell receptor and its significance in tumor recognition. Additionally, this review examines conventional and emerging γδ T cell therapies, encompassing both non-engineered and engineered approaches, with a focus on their efficacy and safety profiles in clinical trials. From multifunctional capabilities to diverse tissue distribution, γδ T cells play a pivotal role in immune regulation and surveillance. By analyzing current research findings, this paper offers insights into the dynamic landscape of γδ T cell-based immunotherapies, underscoring their promise as a potent armamentarium against cancer. Furthermore, by dissecting the complex biology of γδ T cells, we learn valuable information about the anti-cancer contributions of γδ T cells, as well as potential targets for immunotherapeutic interventions.
Collapse
Affiliation(s)
- Stephen G Cieslak
- Division of Hematology/Oncology, Department of Medicine, Indiana University, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA
| | - Reza Shahbazi
- Division of Hematology/Oncology, Department of Medicine, Indiana University, Indianapolis, IN, USA.
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA.
- Tumor Microenvironment & Metastasis, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
- Brown Center for Immunotherapy, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
3
|
Santos AP, Rodrigues LS, Rother N, Mello FCDQ, Magis-Escurra C. The role of neutrophil response in lung damage and post-tuberculosis lung disease: a translational narrative review. Front Immunol 2025; 16:1528074. [PMID: 40124364 PMCID: PMC11925771 DOI: 10.3389/fimmu.2025.1528074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
It is estimated that more than 150 million individuals alive in 2020 had survived tuberculosis (TB). A portion of this large population continues to experience chronic respiratory abnormalities, with or without symptoms, due to previous active pulmonary TB. This condition known as Post-TB Lung Disease (PTLD), involves a complex interaction between pathogen, host and environmental factors. These interactions are believed to drive a hyperinflammatory process in the lungs during active TB, resulting in tissue damage, which may lead to radiological sequelae, impaired pulmonary function, clinical symptoms, such as cough, dyspnea, hemoptysis, and respiratory infections. Such complications impose significant health, financial, and social burdens, which remain poorly understood and inadequately addressed by health care systems. Given the heterogeneity of immune cells and their products infiltrating the airways and the lung parenchyma during acute and chronic inflammation caused by Mycobacterium tuberculosis infection, it is evident that TB immunopathology is multifactorial. Among the various components involved, neutrophils have recently emerged as critical contributors to the deleterious immune response against TB, leading to severe pulmonary damage. In this translational narrative review, we aim to summarize the role of neutrophils and their primary products - proteases (such as elastase), matrix metalloproteinases and neutrophils extracellular traps (NETs) - in pulmonary TB. We highlight new concepts and emerging evidence of neutrophil involvement during the active disease, translating these insights from "bench to bedside" to facilitate dialogue between fundamental researchers and clinical practitioners. Additionally, we present potential targets for future treatment strategies that could mitigate or even prevent PTLD.
Collapse
Affiliation(s)
- Ana Paula Santos
- Pulmonary Diseases Department, Pedro Ernesto University Hospital, State University of Rio de Janeiro, Rio de Janeiro, Brazil
- Thoracic Diseases Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Respiratory Diseases-TB Expert Center, Radboud University Medical Center, Nijmegen, Netherlands
| | - Luciana Silva Rodrigues
- Department of Pathology and Laboratories, Medical Sciences Faculty, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nils Rother
- Department of Nephrology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Cecile Magis-Escurra
- Department of Respiratory Diseases-TB Expert Center, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
4
|
Khan N, Tran KA, Chevre R, Locher V, Richter M, Sun S, Sadeghi M, Pernet E, Herrero-Cervera A, Grant A, Saif A, Downey J, Kaufmann E, Khader SA, Joubert P, Barreiro LB, Yipp BG, Soehnlein O, Divangahi M. β-Glucan reprograms neutrophils to promote disease tolerance against influenza A virus. Nat Immunol 2025; 26:174-187. [PMID: 39779870 PMCID: PMC11785525 DOI: 10.1038/s41590-024-02041-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/21/2024] [Indexed: 01/11/2025]
Abstract
Disease tolerance is an evolutionarily conserved host defense strategy that preserves tissue integrity and physiology without affecting pathogen load. Unlike host resistance, the mechanisms underlying disease tolerance remain poorly understood. In the present study, we investigated whether an adjuvant (β-glucan) can reprogram innate immunity to provide protection against influenza A virus (IAV) infection. β-Glucan treatment reduces the morbidity and mortality against IAV infection, independent of host resistance. The enhanced survival is the result of increased recruitment of neutrophils via RoRγt+ T cells in the lung tissue. β-Glucan treatment promotes granulopoiesis in a type 1 interferon-dependent manner that leads to the generation of a unique subset of immature neutrophils utilizing a mitochondrial oxidative metabolism and producing interleukin-10. Collectively, our data indicate that β-glucan reprograms hematopoietic stem cells to generate neutrophils with a new 'regulatory' function, which is required for promoting disease tolerance and maintaining lung tissue integrity against viral infection.
Collapse
Affiliation(s)
- Nargis Khan
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins Christie Laboratories, McGill University, Montréal, Québec, Canada.
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Kim A Tran
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins Christie Laboratories, McGill University, Montréal, Québec, Canada
| | - Raphael Chevre
- Institute of Experimental Pathology, Centre of Molecular Biology of Inflammation, Münster, Germany
| | - Veronica Locher
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Mathis Richter
- Institute of Experimental Pathology, Centre of Molecular Biology of Inflammation, Münster, Germany
| | - Sarah Sun
- Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Mina Sadeghi
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins Christie Laboratories, McGill University, Montréal, Québec, Canada
| | - Erwan Pernet
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins Christie Laboratories, McGill University, Montréal, Québec, Canada
| | - Andrea Herrero-Cervera
- Institute of Experimental Pathology, Centre of Molecular Biology of Inflammation, Münster, Germany
| | - Alexandre Grant
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins Christie Laboratories, McGill University, Montréal, Québec, Canada
| | - Ahmed Saif
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins Christie Laboratories, McGill University, Montréal, Québec, Canada
| | - Jeffrey Downey
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins Christie Laboratories, McGill University, Montréal, Québec, Canada
| | - Eva Kaufmann
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins Christie Laboratories, McGill University, Montréal, Québec, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - Philippe Joubert
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Québec City, Québec, Canada
| | - Luis B Barreiro
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Bryan G Yipp
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Oliver Soehnlein
- Institute of Experimental Pathology, Centre of Molecular Biology of Inflammation, Münster, Germany
| | - Maziar Divangahi
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins Christie Laboratories, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
5
|
Zhou X, Wu Y, Zhu Z, Lu C, Zhang C, Zeng L, Xie F, Zhang L, Zhou F. Mucosal immune response in biology, disease prevention and treatment. Signal Transduct Target Ther 2025; 10:7. [PMID: 39774607 PMCID: PMC11707400 DOI: 10.1038/s41392-024-02043-4] [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: 06/01/2024] [Revised: 09/05/2024] [Accepted: 10/27/2024] [Indexed: 01/11/2025] Open
Abstract
The mucosal immune system, as the most extensive peripheral immune network, serves as the frontline defense against a myriad of microbial and dietary antigens. It is crucial in preventing pathogen invasion and establishing immune tolerance. A comprehensive understanding of mucosal immunity is essential for developing treatments that can effectively target diseases at their entry points, thereby minimizing the overall impact on the body. Despite its importance, our knowledge of mucosal immunity remains incomplete, necessitating further research. The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has underscored the critical role of mucosal immunity in disease prevention and treatment. This systematic review focuses on the dynamic interactions between mucosa-associated lymphoid structures and related diseases. We delve into the basic structures and functions of these lymphoid tissues during disease processes and explore the intricate regulatory networks and mechanisms involved. Additionally, we summarize novel therapies and clinical research advances in the prevention of mucosal immunity-related diseases. The review also addresses the challenges in developing mucosal vaccines, which aim to induce specific immune responses while maintaining tolerance to non-pathogenic microbes. Innovative therapies, such as nanoparticle vaccines and inhalable antibodies, show promise in enhancing mucosal immunity and offer potential for improved disease prevention and treatment.
Collapse
Affiliation(s)
- Xiaoxue Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yuchen Wu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhipeng Zhu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Chu Lu
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Chunwu Zhang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linghui Zeng
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feng Xie
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
6
|
Ekstedt S, Piersiala K, Kolev A, Farrajota Neves da Silva P, Margolin G, Kumlien Georén S, Cardell LO. Phenotypical differences of neutrophils patrolling tumour-draining lymph nodes in head and neck cancer. Br J Cancer 2024; 131:1893-1900. [PMID: 39543389 PMCID: PMC11628601 DOI: 10.1038/s41416-024-02891-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND The complexity and heterogeneity of neutrophils are recognized, especially their roles in modulating inflammation and cancer immune responses. The detailed functions of neutrophils in human tumour-draining lymph nodes (TDLNs), specifically in the context of head and neck cancer, remain inadequately characterized. AIM This study aims to delineate the phenotypic diversity of neutrophils in TDLNs, non-tumour-draining lymph nodes (nTDLNs) from patients with oral squamous cell carcinoma (OSCC), and to evaluate their correlation with clinical outcomes. METHODS A flow cytometry-based investigation. RESULTS Neutrophils manifest a tissue-specific heterogeneity with significant phenotypic differences between compartments. A substantial fraction of neutrophils displayed an activated CD16highCD62Ldim profile in TDLNs, more prominent in patients with advanced T stages, implicating their involvement in the disease's progression. Notably, the presence of this activated neutrophil phenotype in TDLNs was strongly associated with poorer patient prognosis. CONCLUSIONS The study confirms the heterogeneity of neutrophils in human TDLNs, aligning with findings from animal models but extending them to show clinical relevance in human disease. The correlation of neutrophil phenotypes with cancer progression and prognosis emphasizes the importance of these cells in the tumour-microenvironment. The data suggests a future possibility to develop targeted therapies that modulate the neutrophilic response in OSCC.
Collapse
Affiliation(s)
- Sandra Ekstedt
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Krzysztof Piersiala
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Otorhinolaryngology, Karolinska University Hospital, Stockholm, Sweden
| | - Aeneas Kolev
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Otorhinolaryngology, Karolinska University Hospital, Stockholm, Sweden
- Medical unit Head Neck, Lung and Skin Cancer, Karolinska University Hospital, Stockholm, Sweden
| | | | - Gregori Margolin
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Otorhinolaryngology, Karolinska University Hospital, Stockholm, Sweden
- Medical unit Head Neck, Lung and Skin Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Susanna Kumlien Georén
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Otorhinolaryngology, Karolinska University Hospital, Stockholm, Sweden
| | - Lars-Olaf Cardell
- Division of ENT Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
- Department of Otorhinolaryngology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
7
|
Roy M, Sengupta R, Chakraborty BC, Chatterjee U, von Stebut E, Kaye PM, Chatterjee M. Role of neutrophils in the pathogenesis of Post Kala-azar Dermal Leishmaniasis (PKDL). PLoS Negl Trop Dis 2024; 18:e0012655. [PMID: 39602398 PMCID: PMC11602034 DOI: 10.1371/journal.pntd.0012655] [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: 04/26/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Post Kala-azar Dermal Leishmaniasis (PKDL) is a dermal sequel of visceral leishmaniasis (VL), poses a significant threat to the success of ongoing kala-azar elimination program, due to its potential role in sustaining transmission cycles and complicating disease management strategies. In VL, neutrophils have been identified as the 'first line of defence', having multiple roles in disease pathogenesis, but their role in PKDL, if any, still remains elusive; presenting a critical gap in knowledge, and was the aim of this study. METHODOLOGY/PRINCIPAL FINDINGS In a cohort of PKDL patients, CD66b+ neutrophils were quantified in skin biopsies, followed by immunostaining of FFPE sections to identify activated neutrophils (CD66b+/CD64+) and degranulated (CD66b+/MPO+), along with expression of neutrophil elastase (NE), matrix metalloprotease 9 (MMP9) and collagen I. Plasma levels of neutrophil chemo-attractants CXCL8/1/2/5, CCL2 and 20 and cytokines, (IL-6, IFN-γ, IL-4, IL-10, TNF-α, IL-17 and IL-22, 23) were evaluated by a multiplex assay, while lesional expression of IL-8, IL-10 and IL-17 was evaluated by immunohistochemistry. As compared to healthy individuals (control skin samples), PKDL cases at the lesional sites had an increased number of activated CD66b+ neutrophils (positive for CD64+, MPO+ and NE+). The plasma levels of neutrophil chemo-attractants, pro-inflammatory and regulatory cytokines were raised as was circulating and lesional IL-8, along with an enhanced lesional expression of IL-10 and IL-17A. An increase in circulatory and lesional MMP9 was accompanied by decreased collagen I, suggesting disintegration of matrix integrity. CONCLUSIONS/SIGNIFICANCE Taken together, in PKDL, activated neutrophils possibly contribute towards modulating the lesional landscape. Understanding this involvement of neutrophils in patients with PKDL, particularly in the absence of an animal model, could offer better understanding of the disease pathogenesis and provide insights into novel therapeutic strategies for the ongoing elimination program.
Collapse
Affiliation(s)
- Madhurima Roy
- Dept. of Pharmacology, Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Ritika Sengupta
- Dept. of Pharmacology, Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Bidhan Chandra Chakraborty
- Multidisciplinary Research Unit (MRU) Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Uttara Chatterjee
- Pathology, Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| | - Esther von Stebut
- Department of Dermatology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Paul M. Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Mitali Chatterjee
- Dept. of Pharmacology, Institute of Post Graduate Medical Education and Research (IPGME&R), Kolkata, India
| |
Collapse
|
8
|
Lad M, Beniwal AS, Jain S, Shukla P, Kalistratova V, Jung J, Shah SS, Yagnik G, Saha A, Sati A, Babikir H, Nguyen AT, Gill S, Rios J, Young JS, Lui A, Salha D, Diaz A, Aghi MK. Glioblastoma induces the recruitment and differentiation of dendritic-like "hybrid" neutrophils from skull bone marrow. Cancer Cell 2024; 42:1549-1569.e16. [PMID: 39255776 PMCID: PMC11446475 DOI: 10.1016/j.ccell.2024.08.008] [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: 03/08/2023] [Revised: 05/28/2024] [Accepted: 08/08/2024] [Indexed: 09/12/2024]
Abstract
Tumor-associated neutrophil (TAN) effects on glioblastoma (GBM) biology remain under-characterized. We show here that neutrophils with dendritic features-including morphological complexity, expression of antigen presentation genes, and the ability to process exogenous peptide and stimulate major histocompatibility complex (MHC)II-dependent T cell activation-accumulate intratumorally and suppress tumor growth in vivo. Trajectory analysis of patient TAN scRNA-seq identifies this "hybrid" dendritic-neutrophil phenotype as a polarization state that is distinct from canonical cytotoxic TANs, and which differentiates from local precursors. These hybrid-inducible immature neutrophils-which we identified in patient and murine glioblastomas-arise not from circulation, but from local skull marrow. Through labeled skull flap transplantation and targeted ablation, we characterize calvarial marrow as a contributor of antitumoral myeloid antigen-presenting cells (APCs), including TANs, which elicit T cell cytotoxicity and memory. As such, agents augmenting neutrophil egress from skull marrow-such as intracalvarial AMD3100, whose survival-prolonging effect in GBM we report-present therapeutic potential.
Collapse
Affiliation(s)
- Meeki Lad
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Angad S Beniwal
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Saket Jain
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Poojan Shukla
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Venina Kalistratova
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Jangham Jung
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Sumedh S Shah
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Garima Yagnik
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Atul Saha
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Ankita Sati
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Husam Babikir
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Alan T Nguyen
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Sabraj Gill
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Jennifer Rios
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Jacob S Young
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Austin Lui
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Diana Salha
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Aaron Diaz
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA
| | - Manish K Aghi
- University of California, San Francisco (UCSF), Department of Neurosurgery, San Francisco, CA, USA.
| |
Collapse
|
9
|
Avery D, Morandini L, Sheakley L, Grabiec M, Olivares-Navarrete R. CD4 + and CD8 + T cells reduce inflammation and promote bone healing in response to titanium implants. Acta Biomater 2024; 179:385-397. [PMID: 38554889 PMCID: PMC11045310 DOI: 10.1016/j.actbio.2024.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
T cells are adaptive immune cells essential in pathogenic response, cancer, and autoimmune disorders. During the integration of biomaterials with host tissue, T cells modify the local inflammatory environment by releasing cytokines that promote inflammatory resolution following implantation. T cells are vital for the modulation of innate immune cells, recruitment and proliferation of mesenchymal stem cells (MSCs), and formation of functional tissue around the biomaterial implant. We have demonstrated that deficiency of αβ T cells promotes macrophage polarization towards a pro-inflammatory phenotype and attenuates MSC recruitment and proliferation in vitro and in vivo. The goal of this study was to understand how CD4+ and CD8+ T cells, subsets of the αβ T cell family, impact the inflammatory response to titanium (Ti) biomaterials. Deficiency of either CD4+ or CD8+ T cells increased the proportion of pro-inflammatory macrophages, lowered anti-inflammatory macrophages, and diminished MSC recruitment in vitro and in vivo. In addition, new bone formation at the implantation site was significantly reduced in T cell-deficient mice compared to T cell-competent mice. Deficiency of CD4+ T cells exacerbated these effects compared to CD8+ T cell deficiency. Our results show the importance of CD4+ and CD8+ T cells in modulating the inflammatory response and promoting new bone formation in response to modified Ti implants. STATEMENT OF SIGNIFICANCE: CD4+ and CD8+ T cells are essential in modulating the peri-implant microenvironment during the inflammatory response to biomaterial implantation. This study shows that deficiency of either CD4+ or CD8+ T cell subsets altered macrophage polarization and reduced MSC recruitment and proliferation at the implantation site.
Collapse
Affiliation(s)
- Derek Avery
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 70 S. Madison Street, Room 3328, Richmond, VA 23220, United States
| | - Lais Morandini
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 70 S. Madison Street, Room 3328, Richmond, VA 23220, United States
| | - Luke Sheakley
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 70 S. Madison Street, Room 3328, Richmond, VA 23220, United States
| | - Melissa Grabiec
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 70 S. Madison Street, Room 3328, Richmond, VA 23220, United States
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 70 S. Madison Street, Room 3328, Richmond, VA 23220, United States.
| |
Collapse
|
10
|
Di X, Chen J, Li Y, Wang M, Wei J, Li T, Liao B, Luo D. Crosstalk between fibroblasts and immunocytes in fibrosis: From molecular mechanisms to clinical trials. Clin Transl Med 2024; 14:e1545. [PMID: 38264932 PMCID: PMC10807359 DOI: 10.1002/ctm2.1545] [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: 07/17/2023] [Revised: 12/25/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND The impact of fibroblasts on the immune system provides insight into the function of fibroblasts. In various tissue microenvironments, multiple fibroblast subtypes interact with immunocytes by secreting growth factors, cytokines, and chemokines, leading to wound healing, fibrosis, and escape of cancer immune surveillance. However, the specific mechanisms involved in the fibroblast-immunocyte interaction network have not yet been fully elucidated. MAIN BODY AND CONCLUSION Therefore, we systematically reviewed the molecular mechanisms of fibroblast-immunocyte interactions in fibrosis, from the history of cellular evolution and cell subtype divisions to the regulatory networks between fibroblasts and immunocytes. We also discuss how these communications function in different tissue and organ statuses, as well as potential therapies targeting the reciprocal fibroblast-immunocyte interplay in fibrosis. A comprehensive understanding of these functional cells under pathophysiological conditions and the mechanisms by which they communicate may lead to the development of effective and specific therapies targeting fibrosis.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jiawei Chen
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Ya Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Menghua Wang
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jingwen Wei
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Tianyue Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Banghua Liao
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Deyi Luo
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| |
Collapse
|
11
|
Wang M, Dehlinger A, Zapata CF, Golan M, Gallaccio G, Sander LE, Schlickeiser S, Kunkel D, Schmitz-Hübsch T, Sawitzki B, Karni A, Braun J, Loyal L, Thiel A, Bellmann-Strobl J, Paul F, Meyer-Arndt L, Böttcher C. Associations of myeloid cells with cellular and humoral responses following vaccinations in patients with neuroimmunological diseases. Nat Commun 2023; 14:7728. [PMID: 38007484 PMCID: PMC10676398 DOI: 10.1038/s41467-023-43553-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/08/2023] [Indexed: 11/27/2023] Open
Abstract
Disease-modifying therapies (DMTs) are widely used in neuroimmunological diseases such as multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD). Although these treatments are known to predispose patients to infections and affect their responses to vaccination, little is known about the impact of DMTs on the myeloid cell compartment. In this study, we use mass cytometry to examine DMT-associated changes in the innate immune system in untreated and treated patients with MS (n = 39) or NMOSD (n = 23). We also investigated the association between changes in myeloid cell phenotypes and longitudinal responsiveness to homologous primary, secondary, and tertiary SARS-CoV-2 mRNA vaccinations. Multiple DMT-associated myeloid cell clusters, in particular CD64+HLADRlow granulocytes, showed significant correlations with B and T cell responses induced by vaccination. Our findings suggest the potential role of myeloid cells in cellular and humoral responses following vaccination in DMT-treated patients with neuroimmunological diseases.
Collapse
Affiliation(s)
- Meng Wang
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Adeline Dehlinger
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Camila Fernández Zapata
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Maya Golan
- Neuroimmunology and Multiple Sclerosis Unit and Laboratory, Sourasky Medical Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Gerardina Gallaccio
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Leif E Sander
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stephan Schlickeiser
- Institute of Medical Immunology, BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, and Berlin Institute of Health Berlin, Berlin, Germany
| | - Desiree Kunkel
- Flow&MassCytometry Core Facility, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Tanja Schmitz-Hübsch
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Neuroscience Clinical Research Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Birgit Sawitzki
- Translational Immunology, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Arnon Karni
- Neuroimmunology and Multiple Sclerosis Unit and Laboratory, Sourasky Medical Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medicine and Sagol School of Neuroscience Tel Aviv University, Tel Aviv, Israel
| | - Julian Braun
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Immunomics-Regenerative Immunology and Aging, Berlin, Germany
| | - Lucie Loyal
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Immunomics-Regenerative Immunology and Aging, Berlin, Germany
| | - Andreas Thiel
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Immunomics-Regenerative Immunology and Aging, Berlin, Germany
| | - Judith Bellmann-Strobl
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Neuroscience Clinical Research Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Neurology with Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Lil Meyer-Arndt
- Neuroscience Clinical Research Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology with Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Chotima Böttcher
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
| |
Collapse
|
12
|
Sebastian A, Martin KA, Peran I, Hum NR, Leon NF, Amiri B, Wilson SP, Coleman MA, Wheeler EK, Byers SW, Loots GG. Loss of Cadherin-11 in pancreatic ductal adenocarcinoma alters tumor-immune microenvironment. Front Oncol 2023; 13:1286861. [PMID: 37954069 PMCID: PMC10639148 DOI: 10.3389/fonc.2023.1286861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/12/2023] [Indexed: 11/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the top five deadliest forms of cancer with very few treatment options. The 5-year survival rate for PDAC is 10% following diagnosis. Cadherin 11 (Cdh11), a cell-to-cell adhesion molecule, has been suggested to promote tumor growth and immunosuppression in PDAC, and Cdh11 inhibition significantly extended survival in mice with PDAC. However, the mechanisms by which Cdh11 deficiency influences PDAC progression and anti-tumor immune responses have yet to be fully elucidated. To investigate Cdh11-deficiency induced changes in PDAC tumor microenvironment (TME), we crossed p48-Cre; LSL-KrasG12D/+; LSL-Trp53R172H/+ (KPC) mice with Cdh11+/- mice and performed single-cell RNA sequencing (scRNA-seq) of the non-immune (CD45-) and immune (CD45+) compartment of KPC tumor-bearing Cdh11 proficient (KPC-Cdh11+/+) and Cdh11 deficient (KPC-Cdh11+/-) mice. Our analysis showed that Cdh11 is expressed primarily in cancer-associated fibroblasts (CAFs) and at low levels in epithelial cells undergoing epithelial-to-mesenchymal transition (EMT). Cdh11 deficiency altered the molecular profile of CAFs, leading to a decrease in the expression of myofibroblast markers such as Acta2 and Tagln and cytokines such as Il6, Il33 and Midkine (Mdk). We also observed a significant decrease in the presence of monocytes/macrophages and neutrophils in KPC-Cdh11+/- tumors while the proportion of T cells was increased. Additionally, myeloid lineage cells from Cdh11-deficient tumors had reduced expression of immunosuppressive cytokines that have previously been shown to play a role in immune suppression. In summary, our data suggests that Cdh11 deficiency significantly alters the fibroblast and immune microenvironments and contributes to the reduction of immunosuppressive cytokines, leading to an increase in anti-tumor immunity and enhanced survival.
Collapse
Affiliation(s)
- Aimy Sebastian
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Kelly A. Martin
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Ivana Peran
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University Medical Center, Washington, DC, United States
| | - Nicholas R. Hum
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Nicole F. Leon
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Beheshta Amiri
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Stephen P. Wilson
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Matthew A. Coleman
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Elizabeth K. Wheeler
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
| | - Stephen W. Byers
- Georgetown-Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University Medical Center, Washington, DC, United States
| | - Gabriela G. Loots
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States
- University of California Davis Health, Department of Orthopaedic Surgery, Sacramento, CA, United States
| |
Collapse
|
13
|
Song J, Zhang Y, Bai Y, Sun X, Lu Y, Guo Y, He Y, Gao M, Chi X, Heng BC, Zhang X, Li W, Xu M, Wei Y, You F, Zhang X, Lu D, Deng X. The Deubiquitinase OTUD1 Suppresses Secretory Neutrophil Polarization And Ameliorates Immunopathology of Periodontitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303207. [PMID: 37639212 PMCID: PMC10602526 DOI: 10.1002/advs.202303207] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/12/2023] [Indexed: 08/29/2023]
Abstract
Tissue-infiltrating neutrophils (TINs) secrete various signaling molecules to establish paracrine communication within the inflammatory milieu. It is imperative to identify molecular mediators that control this secretory phenotype of TINs. The present study uncovers a secretory neutrophil subset that exhibits increased pro-inflammatory cytokine production and enhanced migratory capacity which is highly related with periodontal pathogenesis. Further analysis identifies the OTU domain-containing protein 1 (OTUD1) plays a regulatory role in this secretory neutrophil polarization. In human and mouse periodontitis, the waning of inflammation is correlated with OTUD1 upregulation, whereas severe periodontitis is induced when neutrophil-intrinsic OTUD1 is depleted. Mechanistically, OTUD1 interacts with SEC23B, a component of the coat protein II complex (COPII). By removing the K63-linked polyubiquitin chains on SEC23B Lysine 81, the deubiquitinase OTUD1 negatively regulates the COPII secretory machinery and limits protein ER-to-Golgi trafficking, thus restricting the surface expression of integrin-regulated proteins, CD9 and CD47. Accordingly, blockade of protein transport by Brefeldin A (BFA) curbs recruitment of Otud1-deficient TINs and attenuates inflammation-induced alveolar bone destruction. The results thus identify OTUD1 signaling as a negative feedback loop that limits the polarization of neutrophils with secretory phenotype and highlight the potential application of BFA in the treatment of periodontal inflammation.
Collapse
Affiliation(s)
- Jia Song
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology National Clinical Research Center for Oral DiseasesNational Engineering Research Center of Oral Biomaterials and Digital Medical Devices NMPAKey Laboratory for Dental Materials Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yuning Zhang
- Department of OrthodonticsPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yunyang Bai
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology National Clinical Research Center for Oral DiseasesNational Engineering Research Center of Oral Biomaterials and Digital Medical Devices NMPAKey Laboratory for Dental Materials Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xiaowen Sun
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yanhui Lu
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yusi Guo
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology National Clinical Research Center for Oral DiseasesNational Engineering Research Center of Oral Biomaterials and Digital Medical Devices NMPAKey Laboratory for Dental Materials Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Ying He
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology National Clinical Research Center for Oral DiseasesNational Engineering Research Center of Oral Biomaterials and Digital Medical Devices NMPAKey Laboratory for Dental Materials Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Min Gao
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology National Clinical Research Center for Oral DiseasesNational Engineering Research Center of Oral Biomaterials and Digital Medical Devices NMPAKey Laboratory for Dental Materials Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xiaopei Chi
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology National Clinical Research Center for Oral DiseasesNational Engineering Research Center of Oral Biomaterials and Digital Medical Devices NMPAKey Laboratory for Dental Materials Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Boon Chin Heng
- National Center for Stomatology National Clinical Research Center for Oral DiseasesNational Engineering Research Center of Oral Biomaterials and Digital Medical Devices NMPAKey Laboratory for Dental Materials Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Central LaboratoryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xin Zhang
- Institute of Systems BiomedicineSchool of Basic Medical SciencesNHC Key Laboratory of Medical ImmunologyBeijing Key Laboratory of Tumor Systems BiologyPeking University Health Science CenterBeijing100191P. R. China
| | - Wenjing Li
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology National Clinical Research Center for Oral DiseasesNational Engineering Research Center of Oral Biomaterials and Digital Medical Devices NMPAKey Laboratory for Dental Materials Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Mingming Xu
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology National Clinical Research Center for Oral DiseasesNational Engineering Research Center of Oral Biomaterials and Digital Medical Devices NMPAKey Laboratory for Dental Materials Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yan Wei
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology National Clinical Research Center for Oral DiseasesNational Engineering Research Center of Oral Biomaterials and Digital Medical Devices NMPAKey Laboratory for Dental Materials Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Fuping You
- Institute of Systems BiomedicineSchool of Basic Medical SciencesNHC Key Laboratory of Medical ImmunologyBeijing Key Laboratory of Tumor Systems BiologyPeking University Health Science CenterBeijing100191P. R. China
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology National Clinical Research Center for Oral DiseasesNational Engineering Research Center of Oral Biomaterials and Digital Medical Devices NMPAKey Laboratory for Dental Materials Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Peking University‐Yunnan Baiyao International Medical Research CenterBeijing100191P. R. China
| | - Dan Lu
- Institute of Systems BiomedicineSchool of Basic Medical SciencesNHC Key Laboratory of Medical ImmunologyBeijing Key Laboratory of Tumor Systems BiologyPeking University Health Science CenterBeijing100191P. R. China
| | - Xuliang Deng
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology National Clinical Research Center for Oral DiseasesNational Engineering Research Center of Oral Biomaterials and Digital Medical Devices NMPAKey Laboratory for Dental Materials Beijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Peking University‐Yunnan Baiyao International Medical Research CenterBeijing100191P. R. China
| |
Collapse
|
14
|
Di X, Gao X, Peng L, Ai J, Jin X, Qi S, Li H, Wang K, Luo D. Cellular mechanotransduction in health and diseases: from molecular mechanism to therapeutic targets. Signal Transduct Target Ther 2023; 8:282. [PMID: 37518181 PMCID: PMC10387486 DOI: 10.1038/s41392-023-01501-9] [Citation(s) in RCA: 122] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 08/01/2023] Open
Abstract
Cellular mechanotransduction, a critical regulator of numerous biological processes, is the conversion from mechanical signals to biochemical signals regarding cell activities and metabolism. Typical mechanical cues in organisms include hydrostatic pressure, fluid shear stress, tensile force, extracellular matrix stiffness or tissue elasticity, and extracellular fluid viscosity. Mechanotransduction has been expected to trigger multiple biological processes, such as embryonic development, tissue repair and regeneration. However, prolonged excessive mechanical stimulation can result in pathological processes, such as multi-organ fibrosis, tumorigenesis, and cancer immunotherapy resistance. Although the associations between mechanical cues and normal tissue homeostasis or diseases have been identified, the regulatory mechanisms among different mechanical cues are not yet comprehensively illustrated, and no effective therapies are currently available targeting mechanical cue-related signaling. This review systematically summarizes the characteristics and regulatory mechanisms of typical mechanical cues in normal conditions and diseases with the updated evidence. The key effectors responding to mechanical stimulations are listed, such as Piezo channels, integrins, Yes-associated protein (YAP) /transcriptional coactivator with PDZ-binding motif (TAZ), and transient receptor potential vanilloid 4 (TRPV4). We also reviewed the key signaling pathways, therapeutic targets and cutting-edge clinical applications of diseases related to mechanical cues.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xiaoshuai Gao
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Liao Peng
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Jianzhong Ai
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xi Jin
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Shiqian Qi
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Hong Li
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Kunjie Wang
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| | - Deyi Luo
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| |
Collapse
|
15
|
Arora A, Singh A. Exploring the role of neutrophils in infectious and noninfectious pulmonary disorders. Int Rev Immunol 2023; 43:41-61. [PMID: 37353973 DOI: 10.1080/08830185.2023.2222769] [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: 03/25/2022] [Accepted: 05/31/2023] [Indexed: 06/25/2023]
Abstract
With the change in global environment, respiratory disorders are becoming more threatening to the health of people all over the world. These diseases are closely linked to performance of immune system. Within the innate arm of immune system, Neutrophils are an important moiety to serve as an immune defense barrier. They are one of the first cells recruited to the site of infection and plays a critical role in pathogenesis of various pulmonary diseases. It is established that the migration and activation of neutrophils can lead to inflammation either directly or indirectly and this inflammation caused is very crucial for the clearance of pathogens and resolution of infection. However, the immunopathological mechanisms involved to carry out the same is very complex and not well understood. Despite there being studies concentrating on the role of neutrophils in multiple respiratory diseases, there is still a long way to go in order to completely understand the complexity of the participation of neutrophils and mechanisms involved in the development of these respiratory diseases. In the present article, we have reviewed the literature to comprehensively provide an insight in the current development and advancements about the role of neutrophils in infectious respiratory disorders including viral respiratory disorders such as Coronavirus disease (COVID-19) and bacterial pulmonary disorders with a focused review on pulmonary tuberculosis as well as in noninfectious disorders like Chronic obstructive pulmonary disease (COPD) and asthma. Also, future directions into research and therapeutic targets have been discussed for further exploration.
Collapse
Affiliation(s)
- Alisha Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Archana Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
16
|
Li GQ, Xia J, Zeng W, Luo W, Liu L, Zeng X, Cao D. The intestinal γδ T cells: functions in the gut and in the distant organs. Front Immunol 2023; 14:1206299. [PMID: 37398661 PMCID: PMC10311558 DOI: 10.3389/fimmu.2023.1206299] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Located in the frontline against the largest population of microbiota, the intestinal mucosa of mammals has evolved to become an effective immune system. γδ T cells, a unique T cell subpopulation, are rare in circulation blood and lymphoid tissues, but rich in the intestinal mucosa, particularly in the epithelium. Via rapid production of cytokines and growth factors, intestinal γδ T cells are key contributors to epithelial homeostasis and immune surveillance of infection. Intriguingly, recent studies have revealed that the intestinal γδ T cells may play novel exciting functions ranging from epithelial plasticity and remodeling in response to carbohydrate diets to the recovery of ischemic stroke. In this review article, we update regulatory molecules newly defined in lymphopoiesis of the intestinal γδ T cells and their novel functions locally in the intestinal mucosa, such as epithelial remodeling, and distantly in pathological setting, e.g., ischemic brain injury repair, psychosocial stress responses, and fracture repair. The challenges and potential revenues in intestinal γδ T cell studies are discussed.
Collapse
Affiliation(s)
- Guo-Qing Li
- Department of Gastroenterology, Clinical Research Center, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research on Gastrointestinal Tumors, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jiliang Xia
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weihong Zeng
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weijia Luo
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Logen Liu
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research on Gastrointestinal Tumors, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xi Zeng
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Deliang Cao
- Department of Gastroenterology, Clinical Research Center, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
17
|
Lad BM, Beniwal AS, Jain S, Shukla P, Jung J, Shah SS, Yagnik G, Babikir H, Nguyen AT, Gill S, Young JS, Lui A, Salha D, Diaz A, Aghi MK. Glioblastoma induces the recruitment and differentiation of hybrid neutrophils from skull bone marrow. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.24.534105. [PMID: 36993266 PMCID: PMC10055347 DOI: 10.1101/2023.03.24.534105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Tumor-associated neutrophil (TAN) effects on glioblastoma biology remain under-characterized. We show here that 'hybrid' neutrophils with dendritic features - including morphological complexity, expression of antigen presentation genes, and the ability to process exogenous peptide and stimulate MHCII-dependent T cell activation - accumulate intratumorally and suppress tumor growth in vivo . Trajectory analysis of patient TAN scRNA-seq identifies this phenotype as a polarization state which is distinct from canonical cytotoxic TANs and differentiates intratumorally from immature precursors absent in circulation. Rather, these hybrid-inducible immature neutrophils - which we identified in patient and murine glioblastomas - arise from local skull marrow. Through labeled skull flap transplantation and targeted ablation, we characterize calvarial marrow as a potent contributor of antitumoral myeloid APCs, including hybrid TANs and dendritic cells, which elicit T cell cytotoxicity and memory. As such, agents augmenting neutrophil egress from skull marrow - such as intracalvarial AMD3100 whose survival prolonging-effect in GBM we demonstrate - present therapeutic potential.
Collapse
|
18
|
Abstract
Current cancer immunotherapies are primarily predicated on αβ T cells, with a stringent dependence on MHC-mediated presentation of tumour-enriched peptides or unique neoantigens that can limit their efficacy and applicability in various contexts. After two decades of preclinical research and preliminary clinical studies involving very small numbers of patients, γδ T cells are now being explored as a viable and promising approach for cancer immunotherapy. The unique features of γδ T cells, including their tissue tropisms, antitumour activity that is independent of neoantigen burden and conventional MHC-dependent antigen presentation, and combination of typical properties of T cells and natural killer cells, make them very appealing effectors in multiple cancer settings. Herein, we review the main functions of γδ T cells in antitumour immunity, focusing on human γδ T cell subsets, with a particular emphasis on the differences between Vδ1+ and Vδ2+ γδ T cells, to discuss their prognostic value in patients with cancer and the key therapeutic strategies that are being developed in an attempt to improve the outcomes of these patients.
Collapse
|
19
|
Martins RP, Souza FN. Editorial: Cell-mediated immunity in ruminants: Novel approaches and insights. Front Vet Sci 2023; 10:1177315. [PMID: 37035800 PMCID: PMC10080147 DOI: 10.3389/fvets.2023.1177315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Affiliation(s)
- Rodrigo Prado Martins
- ISP, INRAE, Université de Tours, UMR1282, Nouzilly, France
- *Correspondence: Rodrigo Prado Martins
| | - Fernando Nogueira Souza
- Veterinary Clinical Immunology Research Group, Departamento de Clínica Médica, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil
- Fernando Nogueira Souza
| |
Collapse
|
20
|
Zahid KR, Raza U, Tumbath S, Jiang L, Xu W, Huang X. Neutrophils: Musketeers against immunotherapy. Front Oncol 2022; 12:975981. [PMID: 36091114 PMCID: PMC9453237 DOI: 10.3389/fonc.2022.975981] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/01/2022] [Indexed: 11/24/2022] Open
Abstract
Neutrophils, the most copious leukocytes in human blood, play a critical role in tumorigenesis, cancer progression, and immune suppression. Recently, neutrophils have attracted the attention of researchers, immunologists, and oncologists because of their potential role in orchestrating immune evasion in human diseases including cancer, which has led to a hot debate redefining the contribution of neutrophils in tumor progression and immunity. To make this debate fruitful, this review seeks to provide a recent update about the contribution of neutrophils in immune suppression and tumor progression. Here, we first described the molecular pathways through which neutrophils aid in cancer progression and orchestrate immune suppression/evasion. Later, we summarized the underlying molecular mechanisms of neutrophil-mediated therapy resistance and highlighted various approaches through which neutrophil antagonism may heighten the efficacy of the immune checkpoint blockade therapy. Finally, we have highlighted several unsolved questions and hope that answering these questions will provide a new avenue toward immunotherapy revolution.
Collapse
Affiliation(s)
- Kashif Rafiq Zahid
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Umar Raza
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Pakistan
| | - Soumya Tumbath
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lingxiang Jiang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Wenjuan Xu
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiumei Huang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
- *Correspondence: Xiumei Huang,
| |
Collapse
|
21
|
Cao Z, Zhao M, Sun H, Hu L, Chen Y, Fan Z. Roles of mitochondria in neutrophils. Front Immunol 2022; 13:934444. [PMID: 36081497 PMCID: PMC9447286 DOI: 10.3389/fimmu.2022.934444] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/19/2022] [Indexed: 01/25/2023] Open
Abstract
Neutrophils are the most abundant leukocyte in human blood. They are critical for fighting infections and are involved in inflammatory diseases. Mitochondria are indispensable for eukaryotic cells, as they control the biochemical processes of respiration and energy production. Mitochondria in neutrophils have been underestimated since glycolysis is a major metabolic pathway for fuel production in neutrophils. However, several studies have shown that mitochondria are greatly involved in multiple neutrophil functions as well as neutrophil-related diseases. In this review, we focus on how mitochondrial components, metabolism, and related genes regulate neutrophil functions and relevant diseases.
Collapse
Affiliation(s)
- Ziming Cao
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT, United States
| | - Meng Zhao
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States,Department of Microbiology and Immunology, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Hao Sun
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Liang Hu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yunfeng Chen
- Department of Biochemistry and Molecular Biology and Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT, United States,*Correspondence: Zhichao Fan,
| |
Collapse
|
22
|
Identification of the function of γδ1 T cells in the lung cancer microenvironments. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2022; 24:1365-1371. [PMID: 35091999 DOI: 10.1007/s12094-022-02780-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 01/11/2022] [Indexed: 12/27/2022]
Abstract
PURPOSE To investigate whether γδ1 T cells derived from lung cancer tissues have immunosuppressive function and to verify the mechanism of immunosuppressive effect. METHODS Fresh lung cancer tissue samples were collected, some of them were prepared tissue sections, the others were isolated and amplified into TILs cells, γδ1 T cells were isolated from TILs cells by immunomagnetic beads kits, and then cloned and amplified. The immunomodulatory effects of γδ1 T cells on naive and effector CD4+ T cells were detected by immunohistochemistry, flow cytometry, CCK8, ELISA and transwell culture. RESULTS A high proportion of γδ1 T cells was found in lung cancer tissues. The cultural supernatants of γδ1 T cells could inhibit the proliferation of naive CD4+ T cells and decrease the secretion level of IL-2 by effector CD4+ T cells. Further studies showed that the expression levels of IL-8, MIP-1α, MIP-1β and RANTES were higher than that of IFN-γ, GM-CSF and TNF-α, TNF-β, however, their neutralizing antibodies could not block the immunosuppressive activity of the supernatant. CONCLUSION γδ1 T cells play an negative immunoregulation function in lung cancer microenvironments, and have obvious immunosuppressive effects on proliferation and cytokine release of naive CD4+ T cells and effector CD4+ T cells. Preliminary evidence from this study suggests that the mechanism of immunosuppressive effects is mediated by the soluble factors in γδ1 T cell culture supernatants, but its exact molecular mechanism needs to be further explored.
Collapse
|
23
|
Bhattacharya P, Ismail N, Saxena A, Gannavaram S, Dey R, Oljuskin T, Akue A, Takeda K, Yu J, Karmakar S, Dagur PK, McCoy JP, Nakhasi HL. Neutrophil-dendritic cell interaction plays an important role in live attenuated Leishmania vaccine induced immunity. PLoS Negl Trop Dis 2022; 16:e0010224. [PMID: 35192633 PMCID: PMC8896671 DOI: 10.1371/journal.pntd.0010224] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 03/04/2022] [Accepted: 02/02/2022] [Indexed: 11/19/2022] Open
Abstract
Background Neutrophils are involved in the initial host responses to pathogens. Neutrophils can activate T cell responses either independently or through indirect involvement of Dendritic cells (DCs). Recently we have demonstrated direct neutrophil-T cell interactions that initiate adaptive immune responses following immunization with live attenuated Leishmania donovani centrin deleted parasite vaccine (LdCen-/-). However, neutrophil-DC interactions in T cell priming in vaccine immunity in general are not known. In this study we evaluated the interaction between neutrophils and DCs during LdCen-/- infection and compared with wild type parasite (LdWT) both in vitro and in vivo. Methodology/findings LdCen-/- parasite induced increased expression of CCL3 in neutrophils caused higher recruitment of DCs capable of inducing a strong proinflammatory response and elevated co-stimulatory molecule expression compared to LdWT infection. To further illustrate neutrophil-DCs interactions in vivo, we infected LYS-eGFP mice with red fluorescent LdWT/LdCen-/- parasites and sort selected DCs that engulfed the neutrophil containing parasites or DCs that acquired the parasites directly in the ear draining lymph nodes (dLN) 5d post infection. The DCs predominantly acquired the parasites by phagocytosing infected neutrophils. Specifically, DCs containing LdCen-/- parasitized neutrophils exhibited a proinflammatory phenotype, increased expression of costimulatory molecules and initiated higher CD4+T cell priming ex-vivo. Notably, potent DC activation occurred when LdCen-/- parasites were acquired indirectly via engulfment of parasitized neutrophils compared to direct engulfment of LdCen-/- parasites by DCs. Neutrophil depletion in LdCen-/- infected mice significantly abrogated expression of CCL3 resulting in decreased DC recruitment in ear dLN. This event led to poor CD4+Th1 cell priming ex vivo that correlated with attenuated Tbet expression in ear dLN derived CD4+ T cells in vivo. Conclusions Collectively, LdCen-/- containing neutrophils phagocytized by DC markedly influence the phenotype and antigen presenting capacity of DCs early on and thus play an immune-regulatory role in shaping vaccine induced host protective response. Visceral Leishmaniasis (VL), caused by the protozoan parasites of the genus Leishmania is a neglected tropical disease. Leishmania donovani is the principal causative agent of VL in East Africa and the Indian subcontinent whereas in Europe, North Africa, and Latin America VL is mainly caused by Leishmania infantum. No licensed vaccine exists against VL. We have reported previously that live attenuated centrin gene-deleted L. donovani (LdCen-/-) parasite vaccine induced strong innate immunity which leads to a protective Th1 response in animal models. We recently demonstrated that neutrophils play an indispensable role following immunization with LdCen-/- parasites in inducing protective Th1 immune response. However, neutrophils also secrete chemokines that attract other innate cells such as dendritic cells and regulate their activities. In the current study we analyzed the interplay between neutrophils and DCs, and its effects on T cell activation during LdCen-/- infection and compared with wild type parasite (LdWT) infection. We observed that higher recruitment of DCs occurred in LdCen-/- infected mice ear draining lymph nodes compared to LdWT. This recruitment is facilitated by increased secretion of the chemokine CCL3 by neutrophils. A markedly decreased DC recruitment was observed in LdCen-/- infected mice following CCL3 neutralization indicating the key role of neutrophils in DC recruitment. Further, we demonstrated that DCs that ingest LdCen-/- infected neutrophils are better activated than those that acquire the parasites independent of neutrophils. Notably neutrophil depletion in LdCen-/- infected mice also attenuated activation of DCs in the ear dLN that resulted in poor CD4+T cell priming. Our results reveal that interaction between neutrophils and DCs play an important role in shaping proinflammatory immune response induced by a live attenuated Leishmania vaccine.
Collapse
Affiliation(s)
- Parna Bhattacharya
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, United States of America
- * E-mail: (PB); (HLN)
| | - Nevien Ismail
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Ankit Saxena
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Timur Oljuskin
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Adovi Akue
- Division of Bacterial, Parasitic, and Allergenic Products, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Kazuyo Takeda
- Division of Blood Components and Devices, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - James Yu
- Division of Blood Components and Devices, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Subir Karmakar
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Pradeep K. Dagur
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John Philip McCoy
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hira L. Nakhasi
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, United States of America
- * E-mail: (PB); (HLN)
| |
Collapse
|
24
|
Dysconnectivity of a brain functional network was associated with blood inflammatory markers in depression. Brain Behav Immun 2021; 98:299-309. [PMID: 34450247 DOI: 10.1016/j.bbi.2021.08.226] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/09/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE There is increasing evidence for a subgroup of major depressive disorder (MDD) associated with heightened peripheral blood inflammatory markers. In this study, we aimed to understand the mechanistic brain-immune axis in inflammation-linked depression by investigating associations between functional connectivity (FC) of brain networks and peripheral blood immune markers in depression. METHODS Resting-state functional magnetic resonance imaging (fMRI) and peripheral blood inflammatory markers (C-reactive protein; CRP, interleukin-6; IL-6 and immune cells) were collected on N = 46 healthy controls (HC; CRP ≤ 3 mg/L) and N = 83 cases of depression, stratified further into low CRP cases (loCRP cases; ≤ 3 mg/L; N = 50) and high CRP cases (hiCRP cases; > 3 mg/L; N = 33). In a two-part analysis, network-based statistics (NBS) was firstly used to ascertain whole-brain FC differences in HC vs hiCRP cases. Secondly, we investigated the association between this network of interconnected brain regions and continuous measures of peripheral CRP (N = 83), IL-6 (N = 72), neutrophils and CD4+ T-cells (N = 36) in depression cases only. RESULTS Case-control NBS testing revealed a single network of abnormally attenuated FC in the high CRP depression cases compared to healthy controls. Connections within this network were mainly between brain regions located in the left insula/frontal operculum and posterior cingulate cortex, which were assigned to ventral attention and default mode canonical fMRI networks respectively. Within-group analysis across all depression cases, secondarily demonstrated that FC within the identified network significantly negatively scaled with CRP, IL-6 and neutrophils. CONCLUSIONS The findings suggest that inflammation is associated with disruption of functional connectivity within a brain network deemed critical for interoceptive signalling, e.g. accurate communication of peripheral bodily signals such as immune states to the brain, with implications for the pathogenesis of inflammation-linked depression.
Collapse
|
25
|
Volpedo G, Pacheco-Fernandez T, Bhattacharya P, Oljuskin T, Dey R, Gannavaram S, Satoskar AR, Nakhasi HL. Determinants of Innate Immunity in Visceral Leishmaniasis and Their Implication in Vaccine Development. Front Immunol 2021; 12:748325. [PMID: 34712235 PMCID: PMC8546207 DOI: 10.3389/fimmu.2021.748325] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/24/2021] [Indexed: 12/22/2022] Open
Abstract
Leishmaniasis is endemic to the tropical and subtropical regions of the world and is transmitted by the bite of an infected sand fly. The multifaceted interactions between Leishmania, the host innate immune cells, and the adaptive immunity determine the severity of pathogenesis and disease development. Leishmania parasites establish a chronic infection by subversion and attenuation of the microbicidal functions of phagocytic innate immune cells such as neutrophils, macrophages and dendritic cells (DCs). Other innate cells such as inflammatory monocytes, mast cells and NK cells, also contribute to resistance and/or susceptibility to Leishmania infection. In addition to the cytokine/chemokine signals from the innate immune cells, recent studies identified the subtle shifts in the metabolic pathways of the innate cells that activate distinct immune signal cascades. The nexus between metabolic pathways, epigenetic reprogramming and the immune signaling cascades that drive the divergent innate immune responses, remains to be fully understood in Leishmania pathogenesis. Further, development of safe and efficacious vaccines against Leishmaniasis requires a broader understanding of the early interactions between the parasites and innate immune cells. In this review we focus on the current understanding of the specific role of innate immune cells, the metabolomic and epigenetic reprogramming and immune regulation that occurs during visceral leishmaniasis, and the strategies used by the parasite to evade and modulate host immunity. We highlight how such pathways could be exploited in the development of safe and efficacious Leishmania vaccines.
Collapse
Affiliation(s)
- Greta Volpedo
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Thalia Pacheco-Fernandez
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Parna Bhattacharya
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Timur Oljuskin
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Ranadhir Dey
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Sreenivas Gannavaram
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Abhay R Satoskar
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Hira L Nakhasi
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
26
|
Pierre N, Salée C, Vieujean S, Bequet E, Merli AM, Siegmund B, Meuwis MA, Louis E. Review article: distinctions between ileal and colonic Crohn's disease: from physiology to pathology. Aliment Pharmacol Ther 2021; 54:779-791. [PMID: 34297423 DOI: 10.1111/apt.16536] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/15/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ileal and colonic Crohn's disease seem to be two separate entities. AIMS To describe the main physiological distinctions between the small and the large intestine and to analyse the differences between ileal and colonic Crohn's disease. METHODS The relevant literature was critically examined and synthesised. RESULTS The small and large intestine have fundamental distinctions (anatomy, cellular populations, immune defence, microbiota). The differences between ileal and colonic Crohn's disease are highlighted by a heterogeneous body of evidence including clinical features (natural history of the disease, efficacy of treatments, and monitoring), epidemiological data (smoking status, age, gender) and biological data (genetics, microbiota, immunity, mesenteric fat). However, the contribution of these factors to disease location remains poorly understood. CONCLUSION The classification of ileal and colonic Crohn's disease as distinct subphenotypes is well supported by the literature. Understanding of these differences could be exploited to develop more individualised patient care.
Collapse
Affiliation(s)
- Nicolas Pierre
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium
| | - Catherine Salée
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium
| | - Sophie Vieujean
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium.,Hepato-Gastroenterology and Digestive Oncology Department, Liège University Hospital, Liège, Belgium
| | - Emeline Bequet
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium.,Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Liège University Hospital, Liège, Belgium
| | - Angela-Maria Merli
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium
| | - Britta Siegmund
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Marie-Alice Meuwis
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium.,Hepato-Gastroenterology and Digestive Oncology Department, Liège University Hospital, Liège, Belgium
| | - Edouard Louis
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium.,Hepato-Gastroenterology and Digestive Oncology Department, Liège University Hospital, Liège, Belgium
| |
Collapse
|
27
|
Frasheri I, Heym R, Ern C, Summer B, Hennessen TG, Högg C, Reichl FX, Folwaczny M. Salivary and gingival CXCL8 correlation with periodontal status, periodontal pathogens and smoking. Oral Dis 2021; 28:2267-2276. [PMID: 34388304 DOI: 10.1111/odi.13994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Neutrophil granulocytes have been proposed to play a major role in the mediation of periodontitis-associated tissue destruction. Their recruitment and activation are regulated by the chemokine CXCL8. This study aimed to delineate the dependency of CXCL8-expression in gingival crevicular fluid (GCF) and saliva on periodontal status, bacterial infection and smoking, in patients with periodontitis. METHODS The study cohort comprised 279 subjects with untreated periodontitis. Probing pocket depth (PPD), gingival recession, bleeding on probing (BOP), plaque index, and bone loss were evaluated. CXCL8 was determined in saliva and GCF using flow cytometry. RESULTS Considering the entire study sample, CXCL8 levels were correlated with the mean PPD (ρ=0.131; p=0.029), severity of periodontitis (ρ=0.121; p=0.043), BOP (ρ=0.204; p=0.001) and smoking (ρ=-0.219; p<0.0001) in GCF; and, in whole saliva, with mean PPD (ρ=0.154; p=0.010) severity of periodontitis (ρ=0.140; p=0.020), gender (ρ=0.178; p=0.003) and smoking (ρ=-0.156; p=0.010). Subgroup analysis among non-smokers revealed significantly higher amounts of CXCL8 in GCF (p=0.012) and saliva (p=0.026) comparing subjects with mean PPD ≤3mm and >3mm. CONCLUSION The current study revealed a strong dependency of CXCL8-expression in GCF on the severity and activity of periodontal disease. Smoking causes a significant reduction of CXCL8-expression in saliva and GCF.
Collapse
Affiliation(s)
- Iris Frasheri
- Department of Conservative Dentistry and Periodontology, University Hospital, LMU Munich, Germany
| | - Richard Heym
- Department of Conservative Dentistry and Periodontology, University Hospital, LMU Munich, Germany
| | - Christina Ern
- Department of Conservative Dentistry and Periodontology, University Hospital, LMU Munich, Germany
| | - Burkhard Summer
- Department of Dermatology and Allergology, University Hospital, LMU Munich, Germany
| | - Till G Hennessen
- Department of Conservative Dentistry and Periodontology, University Hospital, LMU Munich, Germany
| | - Christof Högg
- Walther Straub Institute of Pharmacology and Toxicology, University Hospital, LMU Munich, Germany
| | - Franz-Xaver Reichl
- Department of Conservative Dentistry and Periodontology, University Hospital, LMU Munich, Germany.,Walther Straub Institute of Pharmacology and Toxicology, University Hospital, LMU Munich, Germany
| | - Matthias Folwaczny
- Department of Conservative Dentistry and Periodontology, University Hospital, LMU Munich, Germany
| |
Collapse
|
28
|
Older but Not Wiser: the Age-Driven Changes in Neutrophil Responses during Pulmonary Infections. Infect Immun 2021; 89:IAI.00653-20. [PMID: 33495271 DOI: 10.1128/iai.00653-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Elderly individuals are at increased risk of life-threatening pulmonary infections. Neutrophils are a key determinant of the disease course of pathogen-induced pneumonia. Optimal host defense balances initial robust pulmonary neutrophil responses to control pathogen numbers, ultimately followed by the resolution of inflammation to prevent pulmonary damage. Recent evidence suggests that phenotypic and functional heterogeneity in neutrophils impacts host resistance to pulmonary pathogens. Apart from their apparent role in innate immunity, neutrophils also orchestrate subsequent adaptive immune responses during infection. Thus, the outcome of pulmonary infections can be shaped by neutrophils. This review summarizes the age-driven impairment of neutrophil responses and the contribution of these cells to the susceptibility of the elderly to pneumonia. We describe how aging is accompanied by changes in neutrophil recruitment, resolution, and function. We discuss how systemic and local changes alter the neutrophil phenotype in aged hosts. We highlight the gap in knowledge of whether these changes in neutrophils also contribute to the decline in adaptive immunity seen with age. We further detail the factors that drive dysregulated neutrophil responses in the elderly and the pathways that may be targeted to rebalance neutrophil activity and boost host resistance to pulmonary infections.
Collapse
|
29
|
Tchalla EYI, Bhalla M, Wohlfert EA, Bou Ghanem EN. Neutrophils Are Required During Immunization With the Pneumococcal Conjugate Vaccine for Protective Antibody Responses and Host Defense Against Infection. J Infect Dis 2021; 222:1363-1370. [PMID: 32391562 DOI: 10.1093/infdis/jiaa242] [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: 02/04/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022] Open
Abstract
Neutrophils can shape adaptive immunity; however, their role in vaccine-induced protection against infections in vivo remains unclear. Here, we tested their role in the clinically relevant polysaccharide conjugate vaccine against Streptococcus pneumoniae (pneumococcus). We antibody depleted neutrophils during vaccination, allowed them to recover, and 4 weeks later challenged mice with pneumococci. We found that while isotype-treated vaccinated controls were protected against an otherwise lethal infection in naive mice, full protection was lost upon neutrophil depletion. Compared to vaccinated controls, neutrophil-depleted mice had higher lung bacterial burdens, increased incidence of bacteremia, and lower survival rates. Sera from neutrophil-depleted mice had less antipneumococcal IgG2c and IgG3, were less efficient at inducing opsonophagocytic killing of bacteria by neutrophils in vitro, and were worse at protecting naive mice against pneumococcal pneumonia. In summary, neutrophils are required during vaccination for optimal host protection, which has important implications for future vaccine design against pneumococci and other pathogens.
Collapse
Affiliation(s)
- Essi Y I Tchalla
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, USA
| | - Manmeet Bhalla
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, USA
| | - Elizabeth A Wohlfert
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, USA
| | - Elsa N Bou Ghanem
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, New York, USA
| |
Collapse
|
30
|
Vonwirth V, Bülbül Y, Werner A, Echchannaoui H, Windschmitt J, Habermeier A, Ioannidis S, Shin N, Conradi R, Bros M, Tenzer S, Theobald M, Closs EI, Munder M. Inhibition of Arginase 1 Liberates Potent T Cell Immunostimulatory Activity of Human Neutrophil Granulocytes. Front Immunol 2021; 11:617699. [PMID: 33717053 PMCID: PMC7952869 DOI: 10.3389/fimmu.2020.617699] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Myeloid cell arginase-mediated arginine depletion with consecutive inhibition of T cell functions is a key component of tumor immune escape. Both, granulocytic myeloid-derived suppressor cells (G-MDSC) and conventional mature human polymorphonuclear neutrophil granulocytes (PMN) express high levels of arginase 1 and can act as suppressor cells of adaptive anti-cancer immunity. Here we demonstrate that pharmacological inhibition of PMN-derived arginase 1 not only prevents the suppression of T cell functions but rather leads to a strong hyperactivation of T cells. Human PMN were incubated in cell culture medium in the absence or presence of an arginase inhibitor. T cells from healthy donors were then activated either polyclonally or in an antigen-specific manner in the supernatants of the PMN cultures at different PMN-T cell ratios. T cell proliferation was completely suppressed in these supernatants in the absence of an arginase inhibitor. Arginase inhibition led to a strong hyperinduction of T cell proliferation, which exceeded control activation conditions up to 25-fold. The hyperinduction was correlated with higher PMN-T cell ratios and was only apparent when PMN arginase activity was blocked sufficiently. The T cell stimulatory factor was liberated very early by PMN and was present in the < 3 kDa fraction of the PMN supernatants. Increased T cell production of specific proinflammatory cytokines by PMN supernatant in the presence of arginase inhibitor was apparent. Upon arginase inhibition, downregulation of important T cell membrane activation and costimulation proteins was completely prevented or de novo induction accelerated. Antigen-specific T cell cytotoxicity against tumor cells was enhanced by PMN supernatant itself and could be further increased by PMN arginase blockade. Finally, we analyzed anergic T cells from multiple myeloma patients and noticed a complete reversal of anergy and the induction of strong proliferation upon T cell activation in PMN supernatants by arginase inhibition. In summary, we discovered a potent PMN-mediated hyperactivation of human T cells, which is apparent only when PMN arginase-mediated arginine depletion is concurrently inhibited. Our findings are clearly relevant for the analysis and prevention of human tumor immune escape in conjunction with the application of arginase inhibitors already being developed clinically.
Collapse
Affiliation(s)
- Verena Vonwirth
- Third Department of Medicine (Hematology, Oncology and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Yagmur Bülbül
- Third Department of Medicine (Hematology, Oncology and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Anke Werner
- Third Department of Medicine (Hematology, Oncology and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Hakim Echchannaoui
- Third Department of Medicine (Hematology, Oncology and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Mainz, Germany
| | - Johannes Windschmitt
- Third Department of Medicine (Hematology, Oncology and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alice Habermeier
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | - Niu Shin
- Incyte Research Institute, Incyte Corporation, Wilmington, DE, United States
| | - Roland Conradi
- Transfusion Center, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stefan Tenzer
- Institute of Immunology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias Theobald
- Third Department of Medicine (Hematology, Oncology and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Mainz, Germany.,Research Center of Immune Therapy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ellen Ildicho Closs
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Markus Munder
- Third Department of Medicine (Hematology, Oncology and Pneumology), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,Research Center of Immune Therapy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
31
|
Bhattacharya P, Dey R, Saxena A, Karmakar S, Ismail N, Gannavaram S, Dagur PK, Satoskar M, Satoskar S, De Paoli S, Takeda K, McCoy JP, Nakhasi HL. Essential Role of Neutrophils in the Protective Immune Response Induced by a Live Attenuated Leishmania Vaccine. THE JOURNAL OF IMMUNOLOGY 2020; 205:3333-3347. [PMID: 33177159 DOI: 10.4049/jimmunol.2000829] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022]
Abstract
No licensed vaccine exists against visceral leishmaniasis (VL), a disease caused by the Leishmania donovani parasite. We have previously reported both macrophages and dendritic cells play important role in the protection induced by a live attenuated centrin gene-deleted L. donovani (LdCen-/- ) parasite vaccine. The role of neutrophils in orchestrating the initial innate response to pathogens is widely recognized. To investigate the early interaction of LdCen-/- with neutrophils, we immunized mice intradermally in the ear pinna with LdCen-/- Compared with LdWT infection, LdCen-/- parasites induced higher recruitment of neutrophils to the ear dermis and ear draining lymph nodes (dLN) as early as 6-18 h after immunization, which were predominantly proinflammatory in nature. Neutrophils from ear dLN of LdCen-/- -immunized mice exhibited heightened expression of costimulatory molecules and attenuated expression of coinhibitory molecules necessary for higher T cell activation. Further phenotypic characterization revealed heterogeneous neutrophil populations containing Nα and Nβ subtypes in the ear dLN. Of the two, the parasitized Nα subset from LdCen-/- -immunized mice exhibited much stronger Ag-specific CD4+ T cell proliferation ex vivo. Adoptive transfer of neutrophils bearing LdCen-/- parasites induced an increased Th1 response in naive mice. Importantly, neutrophil depletion significantly abrogated Ag-specific CD4+ T cell proliferation in LdCen-/- -immunized mice and impaired protection against virulent challenge. Conversely, replenishing of neutrophils significantly restored the LdCen-/- -induced host-protective response. These results suggest that neutrophils are indispensable for protective immunity induced by LdCen-/- parasite vaccine.
Collapse
Affiliation(s)
- Parna Bhattacharya
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993;
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - Ankit Saxena
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Subir Karmakar
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - Nevien Ismail
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - Pradeep K Dagur
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | | | | | - Silvia De Paoli
- Office of Blood Research and Review, U.S. Food and Drug Administration, Silver Spring, MD 20993; and
| | - Kazuyo Takeda
- Microscopy and Imaging Core Facility, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993
| | - John Philip McCoy
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Disease, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993;
| |
Collapse
|
32
|
Abstract
Brucellosis is a bacterial disease of domestic animals and humans. The pathogenic ability of Brucella organisms relies on their stealthy strategy and their capacity to replicate within host cells and to induce long-lasting infections. Brucella organisms barely induce neutrophil activation and survive within these leukocytes by resisting microbicidal mechanisms. Very few Brucella-infected neutrophils are found in the target organs, except for the bone marrow, early in infection. Still, Brucella induces a mild reactive oxygen species formation and, through its lipopolysaccharide, promotes the premature death of neutrophils, which release chemokines and express "eat me" signals. This effect drives the phagocytosis of infected neutrophils by mononuclear cells that become thoroughly susceptible to Brucella replication and vehicles for bacterial dispersion. The premature death of the infected neutrophils proceeds without NETosis, necrosis/oncosis, or classical apoptosis morphology. In the absence of neutrophils, the Th1 response exacerbates and promotes bacterial removal, indicating that Brucella-infected neutrophils dampen adaptive immunity. This modulatory effect opens a window for bacterial dispersion in host tissues before adaptive immunity becomes fully activated. However, the hyperactivation of immunity is not without a price, since neutropenic Brucella-infected animals develop cachexia in the early phases of the disease. The delay in the immunological response seems a sine qua non requirement for the development of long-lasting brucellosis. This property may be shared with other pathogenic alphaproteobacteria closely related to Brucella We propose a model in which Brucella-infected polymorphonuclear neutrophils (PMNs) function as "Trojan horse" vehicles for bacterial dispersal and as modulators of the Th1 adaptive immunity in infection.
Collapse
|
33
|
Kabelitz D, Serrano R, Kouakanou L, Peters C, Kalyan S. Cancer immunotherapy with γδ T cells: many paths ahead of us. Cell Mol Immunol 2020; 17:925-939. [PMID: 32699351 PMCID: PMC7609273 DOI: 10.1038/s41423-020-0504-x] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/27/2020] [Indexed: 12/12/2022] Open
Abstract
γδ T cells play uniquely important roles in stress surveillance and immunity for infections and carcinogenesis. Human γδ T cells recognize and kill transformed cells independently of human leukocyte antigen (HLA) restriction, which is an essential feature of conventional αβ T cells. Vγ9Vδ2 γδ T cells, which prevail in the peripheral blood of healthy adults, are activated by microbial or endogenous tumor-derived pyrophosphates by a mechanism dependent on butyrophilin molecules. γδ T cells expressing other T cell receptor variable genes, notably Vδ1, are more abundant in mucosal tissue. In addition to the T cell receptor, γδ T cells usually express activating natural killer (NK) receptors, such as NKp30, NKp44, or NKG2D which binds to stress-inducible surface molecules that are absent on healthy cells but are frequently expressed on malignant cells. Therefore, γδ T cells are endowed with at least two independent recognition systems to sense tumor cells and to initiate anticancer effector mechanisms, including cytokine production and cytotoxicity. In view of their HLA-independent potent antitumor activity, there has been increasing interest in translating the unique potential of γδ T cells into innovative cellular cancer immunotherapies. Here, we discuss recent developments to enhance the efficacy of γδ T cell-based immunotherapy. This includes strategies for in vivo activation and tumor-targeting of γδ T cells, the optimization of in vitro expansion protocols, and the development of gene-modified γδ T cells. It is equally important to consider potential synergisms with other therapeutic strategies, notably checkpoint inhibitors, chemotherapy, or the (local) activation of innate immunity.
Collapse
Affiliation(s)
- Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, D-24105, Kiel, Germany.
| | - Ruben Serrano
- Institute of Immunology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, D-24105, Kiel, Germany
| | - Léonce Kouakanou
- Institute of Immunology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, D-24105, Kiel, Germany
| | - Christian Peters
- Institute of Immunology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, D-24105, Kiel, Germany
| | - Shirin Kalyan
- Faculty of Medicine, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| |
Collapse
|
34
|
Li K, Ran R, Jiang Z, Fan C, Li T, Yin Z. Changes in T-lymphocyte subsets and risk factors in human immunodeficiency virus-negative patients with active tuberculosis. Infection 2020; 48:585-595. [PMID: 32472529 PMCID: PMC7395032 DOI: 10.1007/s15010-020-01451-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 05/21/2020] [Indexed: 11/08/2022]
Abstract
Purpose Immune function imbalance is closely associated with the occurrence and development of infectious diseases. We studied the characteristics of changes in T-lymphocyte subsets and their risk factors in HIV-negative patients with active tuberculosis (ATB). Methods T-lymphocyte subsets in 275 HIV-negative ATB patients were quantitatively analyzed and compared with an Mycobacteriumtuberculosis-free control group. Single-factor and multifactor analyses of clinical and laboratory characteristics of patients were also conducted. Results In ATB patients, CD4 and CD8 T-cell counts decreased, and the levels were positively interrelated (r = 0.655, P < 0.0001). After 4 weeks of antituberculosis treatment, CD4 and CD8 T-cell counts increased significantly but remained lower than in the control group. CD4 and CD8 cell counts were negatively associated with the extent of lesions detected in the chest by computed tomography (all P < 0.05). Although not reflected in the CD4/CD8 ratio, CD4 and CD8 cell counts differed between drug-resistant TB patients and drug-susceptible TB patients (P = 0.030). The multivariate analysis showed prealbumin, alpha-1 globulin, body mass index, and platelet count were independent risk factors for decreased CD4 cell count (all P < 0.05), while age and platelet count were independent risk factors for decreased CD8 cell count (all P < 0.05). Conclusion CD4 and CD8 T-cell counts showed the evident value in predicting ATB severity. An increase in the CD4/CD8 ratio may be a critical clue of drug resistance in ATB. Although the factors influencing CD4 and CD8 are not identical, our results indicated the importance of serum protein and platelets to ATB patients’ immune function. Electronic supplementary material The online version of this article (10.1007/s15010-020-01451-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kui Li
- Department of Infectious Diseases, Ankang Central Hospital, Ankang, Shaanxi, China.,Department of Infectious Diseases, Ankang Central Hospital, Hubei University of Medicine, Hubei, China
| | - Renyu Ran
- Department of Infectious Diseases, Ankang Central Hospital, Ankang, Shaanxi, China
| | - Zicheng Jiang
- Department of Infectious Diseases, Ankang Central Hospital, Ankang, Shaanxi, China.,Department of Infectious Diseases, Ankang Central Hospital, Hubei University of Medicine, Hubei, China
| | - Chuanqi Fan
- Department of Infectious Diseases, Ankang Central Hospital, Ankang, Shaanxi, China
| | - Tao Li
- Department of Infectious Diseases, Ankang Central Hospital, Ankang, Shaanxi, China
| | - Zhiguo Yin
- Department of Pharmacy, Ankang Central Hospital, No. 85, South Jinzhou Road, Hanbin District, Ankang, 725000, Shaanxi, China.
| |
Collapse
|
35
|
Olusola BA, Kabelitz D, Olaleye DO, Odaibo GN. Early HIV infection is associated with reduced proportions of gamma delta T subsets as well as high creatinine and urea levels. Scand J Immunol 2020; 91:e12868. [PMID: 32052490 PMCID: PMC7335456 DOI: 10.1111/sji.12868] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 12/19/2019] [Accepted: 02/01/2020] [Indexed: 01/02/2023]
Abstract
Renal dysfunctions are major predictors of co-morbidities and mortality in HIV-infected individuals. Unconventional T cells have been shown to regulate kidney functions. However, there is dearth of information on the effect of HIV-associated nephropathies on γδ and DN T cells. It is also not clear whether γδ T cell perturbations observed during the early stages of HIV infection occur before immune activation. In this study, we investigated the relationship between creatinine and urea on the number of unconventional T cells in HIV-infected individuals at the early and chronic stages of infection. Persons in the chronic stage of infection were divided into treatment naïve and exposed groups. Treatment exposed individuals were further subdivided into groups with undetectable and detectable HIV-1RNA in their blood. Creatinine and urea levels were significantly higher among persons in the early HIV infection compared with the other groups. Proportions of γδ T, γδ + CD8, γδ + CD16 cells were also significantly reduced in the early stage of HIV infection (P < .01). Markers of immune activation, CD4 + HLA-DR and CD8 + HLA-DR, were also significantly reduced during early HIV infection (P < .01). Taken together, our findings suggest that high levels of renal markers as well as reduced proportions of gamma delta T cells are associated with the early stages of HIV infection. This event likely occurs before systemic immune activation reaches peak levels. This study provides evidence for the need for early HIV infection diagnosis and treatment.
Collapse
Affiliation(s)
- Babatunde A. Olusola
- Department of Virology, College of Medicine, University of
Ibadan, Ibadan, Oyo State, Nigeria
| | - Dieter Kabelitz
- Institute of Immunology, UKSH Campus Kiel,
Christian-Albrechts-University, Kiel, Germany
| | - David O. Olaleye
- Department of Virology, College of Medicine, University of
Ibadan, Ibadan, Oyo State, Nigeria
| | - Georgina N. Odaibo
- Department of Virology, College of Medicine, University of
Ibadan, Ibadan, Oyo State, Nigeria
| |
Collapse
|
36
|
Ocaranza MP, Moya J, Jalil JE, Lavandero S, Kalergis AM, Molina C, Gabrielli L, Godoy I, Córdova S, Castro P, Mac Nab P, Rossel V, García L, González J, Mancilla C, Fierro C, Farías L. Rho-kinase pathway activation and apoptosis in circulating leucocytes in patients with heart failure with reduced ejection fraction. J Cell Mol Med 2019; 24:1413-1427. [PMID: 31778027 PMCID: PMC6991691 DOI: 10.1111/jcmm.14819] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/11/2019] [Accepted: 09/01/2019] [Indexed: 01/01/2023] Open
Abstract
Background Increased Rho‐kinase activity in circulating leucocytes is observed in heart failure with reduced ejection fraction (HFrEF). However, there is little information in HFrEF regarding other Rho‐kinase pathway components an on the relationship between Rho‐kinase and apoptosis. Here, Rho‐kinase activation levels and phosphorylation of major downstream molecules and apoptosis levels were measured for the first time both in HFrEF patients and healthy individuals. Methods Cross‐sectional study comparing HFrEF patients (n = 20) and healthy controls (n = 19). Rho‐kinase activity in circulating leucocytes (peripheral blood mononuclear cells, PBMCs) was determined by myosin light chain phosphatase 1 (MYPT1) and ezrin‐radixin‐moesin (ERM) phosphorylation. Rho‐kinase cascade proteins phosphorylation p38‐MAPK, myosin light chain‐2, JAK and JNK were also analysed along with apoptosis. Results MYPT1 and ERM phosphorylation were significantly elevated in HFrEF patients, (3.9‐ and 4.8‐fold higher than in controls, respectively). JAK phosphorylation was significantly increased by 300% over controls. Phosphorylation of downstream molecules p38‐MAPK and myosin light chain‐2 was significantly higher by 360% and 490%, respectively, while JNK phosphorylation was reduced by 60%. Catecholamine and angiotensin II levels were significantly higher in HFrEF patients, while angiotensin‐(1‐9) levels were lower. Apoptosis in circulating leucocytes was significantly increased in HFrEF patients by 2.8‐fold compared with controls and significantly correlated with Rho‐kinase activation. Conclusion Rho‐kinase pathway is activated in PMBCs from HFrEF patients despite optimal treatment, and it is closely associated with neurohormonal activation and with apoptosis. ROCK cascade inhibition might induce clinical benefits in HFrEF patients, and its assessment in PMBCs could be useful to evaluate reverse remodelling and disease regression.
Collapse
Affiliation(s)
- Maria Paz Ocaranza
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jackeline Moya
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge E Jalil
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sergio Lavandero
- Faculty of Chemical and Pharmaceutical Sciences, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile.,Cardiology Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alexis M Kalergis
- Departament of Molecular Genetics and Microbiology, Faculty of Biological Sciences, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristián Molina
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luigi Gabrielli
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Iván Godoy
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Samuel Córdova
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Castro
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paul Mac Nab
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Victor Rossel
- Department of Medicine, Hospital del Salvador, Medical School, Universidad de Chile, Santiago, Chile
| | - Lorena García
- Faculty of Chemical and Pharmaceutical Sciences, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - Javier González
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristián Mancilla
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camila Fierro
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis Farías
- Department of Cardiovascular Diseases, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
37
|
La Manna MP, Orlando V, Paraboschi EM, Tamburini B, Di Carlo P, Cascio A, Asselta R, Dieli F, Caccamo N. Mycobacterium tuberculosis Drives Expansion of Low-Density Neutrophils Equipped With Regulatory Activities. Front Immunol 2019; 10:2761. [PMID: 31849955 PMCID: PMC6892966 DOI: 10.3389/fimmu.2019.02761] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/12/2019] [Indexed: 11/19/2022] Open
Abstract
In human tuberculosis (TB) neutrophils represent the most commonly infected phagocyte but their role in protection and pathology is highly contradictory. Moreover, a subset of low-density neutrophils (LDNs) has been identified in TB, but their functions remain unclear. Here, we have analyzed total neutrophils and their low-density and normal-density (NDNs) subsets in patients with active TB disease, in terms of frequency, phenotype, functional features, and gene expression signature. Full-blood counts from Healthy Donors (H.D.), Latent TB infected, active TB, and cured TB patients were performed. Frequency, phenotype, burst activity, and suppressor T cell activity of the two different subsets were assessed by flow cytometry while NETosis and phagocytosis were evaluated by confocal microscopy. Expression analysis was performed by using the semi-quantitative RT-PCR array technology. Elevated numbers of total neutrophils and a high neutrophil/lymphocyte ratio distinguished patients with active TB from all the other groups. PBMCs of patients with active TB disease contained elevated percentages of LDNs compared with those of H.D., with an increased expression of CD66b, CD33, CD15, and CD16 compared to NDNs. Transcriptomic analysis of LDNs and NDNs purified from the peripheral blood of TB patients identified 12 genes differentially expressed: CCL5, CCR5, CD4, IL10, LYZ, and STAT4 were upregulated, while CXCL8, IFNAR1, NFKB1A, STAT1, TICAM1, and TNF were downregulated in LDNs, as compared to NDNs. Differently than NDNs, LDNs failed to phagocyte live Mycobacterium tuberculosis (M. tuberculosis) bacilli, to make oxidative burst and NETosis, but caused significant suppression of antigen-specific and polyclonal T cell proliferation which was partially mediated by IL-10. These insights add a little dowel of knowledge in understanding the pathogenesis of human TB.
Collapse
Affiliation(s)
- Marco Pio La Manna
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University of Palermo, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostic, University of Palermo, Palermo, Italy
| | - Valentina Orlando
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University of Palermo, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostic, University of Palermo, Palermo, Italy
| | | | - Bartolo Tamburini
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University of Palermo, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostic, University of Palermo, Palermo, Italy
| | - Paola Di Carlo
- Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Antonio Cascio
- Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Humanitas Clinical and Research Center-IRCCS, Milan, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University of Palermo, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostic, University of Palermo, Palermo, Italy
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University of Palermo, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostic, University of Palermo, Palermo, Italy
| |
Collapse
|
38
|
Li Y, Wang W, Yang F, Xu Y, Feng C, Zhao Y. The regulatory roles of neutrophils in adaptive immunity. Cell Commun Signal 2019; 17:147. [PMID: 31727175 PMCID: PMC6854633 DOI: 10.1186/s12964-019-0471-y] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/25/2019] [Indexed: 12/16/2022] Open
Abstract
Neutrophils have long been considered as cells playing a crucial role in the immune defence against invading pathogens. Accumulating evidence strongly supported the direct and indirect regulatory effects of neutrophils on adaptive immunity. Exogenous cytokines or cytokines produced in an autocrine manner as well as a cell-to-cell contact between neutrophils and T cells could induce the expression of MHC-II and costimulatory molecules on neutrophils, supporting that neutrophils may function as antigen-presenting cells (APCs) in respects of presenting antigens and activating T cells. In addition to the inflammatory roles, neutrophils also have the propensity and ability to suppress the immune response through different mechanisms. In this review, we will mainly highlight the heterogeneity and functional plasticity of neutrophils and the antigen-presenting capacity of different neutrophil subsets. We also discuss mechanisms relevant to the regulatory effects of neutrophils on adaptive immunity. Understanding how neutrophils modulate adaptive immunity may provide novel strategies and new therapeutic approaches for diseases associated with neutrophils.
Collapse
Affiliation(s)
- Yang Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China
| | - Wei Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Fan Yang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China
| | - Yanan Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China
| | - Chang Feng
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
39
|
Gelatinase B/matrix metalloproteinase-9 and other neutrophil proteases switch off interleukin-2 activity. Biochem J 2019; 476:2191-2208. [PMID: 31262730 DOI: 10.1042/bcj20180382] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/11/2019] [Accepted: 07/01/2019] [Indexed: 12/27/2022]
Abstract
Interleukin 2 (IL-2) is critical for T cell development and homeostasis, being a key regulator of adaptive immune responses in autoimmunity, hypersensitivity reactions and cancer. Therefore, its abundance in serum and peripheral tissues needs tight control. Here, we described a new mechanism contributing to the immunobiology of IL-2. We demonstrated, both in biochemical and cell-based assays, that IL-2 is subject to proteolytic processing by neutrophil matrix metalloproteinase-9 (MMP-9). IL-2 fragments produced after cleavage by MMP-9 remained linked by a disulfide bond and displayed a reduced affinity for all IL-2 receptor subunits and a distinct pattern and timing of signal transduction. Stimulation of IL-2-dependent cells, including murine CTLL-2 and primary human regulatory T cells, with cleaved IL-2 resulted in significantly decreased proliferation. The concerted action of neutrophil proteases destroyed IL-2. Our data suggest that in neutrophil-rich inflammatory conditions in vivo, neutrophil MMP-9 may reduce the abundance of signaling-competent IL-2 and generate a fragment that competes with IL-2 for receptor binding, whereas the combined activity of granulocyte proteases has the potential to degrade and thus eliminate bioavailable IL-2.
Collapse
|
40
|
Advanced Glycated End Products Alter Neutrophil Effect on Regulation of CD 4+ T Cell Differentiation Through Induction of Myeloperoxidase and Neutrophil Elastase Activities. Inflammation 2019; 42:559-571. [PMID: 30343390 DOI: 10.1007/s10753-018-0913-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
CD4+ T cell subset imbalance plays an important role in the development of diabetic complications. Neutrophils have recently been known as the regulator of CD4+ T cell differentiation. However, whether neutrophils affect CD4+ T cell population in diabetes is still elusive. In this study, we investigated the effect of neutrophils stimulated with advanced glycated end products (AGEs), the marker of diabetes, on CD4+ T cell differentiation and its underlying mechanism. Our data showed that the cultural medium of healthy adult neutrophils treated with AGEs increased expressions of both Th1 (IFN-γ) and Th17 (IL-17) phenotypes and the transcription factors of Th1 (Tbet) and Th17 (RORγt) in naive CD4+T cells and CD4+CD25+FoxP3+ (Treg) T cells in vitro. Next, we found that AGEs induced the generations of myeloperoxidase (MPO) and neutrophil elastase (NE) in neutrophils; inhibition of MPO or NE attenuated the effect of AGE-stimulated neutrophils on CD4+ T cell bias. Furthermore, receptor for AGEs (RAGE) inhibitor interrupted AGE-induced MPO and NE expressions, but MPO and NE inhibitions did not change AGE-increased RAGE gene expression. These results suggested that AGEs drive the effect of neutrophils on CD4+ T cell differentiation into pro-inflammatory program through inducing MPO and NE productions in neutrophils, which is mediated by AGE-RAGE interaction.
Collapse
|
41
|
Ilatovskaya DV, Pitts C, Clayton J, Domondon M, Troncoso M, Pippin S, DeLeon-Pennell KY. CD8 + T-cells negatively regulate inflammation post-myocardial infarction. Am J Physiol Heart Circ Physiol 2019; 317:H581-H596. [PMID: 31322426 DOI: 10.1152/ajpheart.00112.2019] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The adaptive immune response is key for cardiac wound healing post-myocardial infarction (MI) despite low T-cell numbers. We hypothesized that CD8+ T-cells regulate the inflammatory response, leading to decreased survival and cardiac function post-MI. We performed permanent occlusion of the left anterior descending coronary artery on C57BL/6J and CD8atm1mak mice (deficient in functional CD8+ T-cells). CD8atm1mak mice had increased survival at 7 days post-MI compared with that of the wild-type (WT) and improved cardiac physiology at day 7 post-MI. Despite having less mortality, 100% of the CD8atm1mak group died because of cardiac rupture compared with only 33% of the WT. Picrosirius red staining and collagen immunoblotting indicated an acceleration of fibrosis in the infarct area as well as remote area in the CD8atm1mak mice; however, this increase was due to elevated soluble collagen implicating poor scar formation. Plasma and tissue inflammation were exacerbated as indicated by higher levels of Cxcl1, Ccl11, matrix metalloproteinase (MMP)-2, and MMP-9. Immunohistochemistry and flow cytometry indicated that the CD8atm1mak group had augmented numbers of neutrophils and macrophages at post-MI day 3 and increased mast cell markers at post-MI day 7. Cleavage of tyrosine-protein kinase MER was increased in the CD8atm1mak mice, resulting in delayed removal of necrotic tissue. In conclusion, despite having improved cardiac physiology and overall survival, CD8atm1mak mice had increased innate inflammation and poor scar formation, leading to higher incidence of cardiac rupture. Our data suggest that the role of CD8+ T-cells in post-MI recovery may be both beneficial and detrimental to cardiac remodeling and is mediated via a cell-specific mechanism.NEW & NOTEWORTHY We identified new mechanisms implicating CD8+ T-cells as regulators of the post-myocardial infarction (MI) wound healing process. Mice without functional CD8+ T-cells had improved cardiac physiology and less mortality 7 days post MI compared with wild-type animals. Despite having better overall survival, animals lacking functional CD8+ T-cells had delayed removal of necrotic tissue, leading to poor scar formation and increased cardiac rupture, suggesting that CD8+ T-cells play a dual role in the cardiac remodeling process.
Collapse
Affiliation(s)
- Daria V Ilatovskaya
- Division of Nephrology, Departments of Medicine and Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Cooper Pitts
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Joshua Clayton
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Mark Domondon
- Division of Nephrology, Departments of Medicine and Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Miguel Troncoso
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Sarah Pippin
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina.,Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
42
|
Oberg HH, Wesch D, Kalyan S, Kabelitz D. Regulatory Interactions Between Neutrophils, Tumor Cells and T Cells. Front Immunol 2019; 10:1690. [PMID: 31379875 PMCID: PMC6657370 DOI: 10.3389/fimmu.2019.01690] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 07/04/2019] [Indexed: 12/16/2022] Open
Abstract
Apart from their activity in combating infections, neutrophils play an important role in regulating the tumor microenvironment. Neutrophils can directly kill (antibody-coated) cancer cells, and support other immune anti-tumoral strategies. On the other hand, neutrophils can also exert pro-tumorigenic activities via the production of factors which promote cancer growth, angiogenesis and metastasis formation. The balance of anti- and pro-cancer activity is influenced by the particularly delicate interplay that exists between neutrophils and T lymphocytes. In murine models, it has been reported that γδ T cells are a major source of IL-17 that drives the recruitment and pro-tumorigenic differentiation of neutrophils. This, however, contrasts with the well-studied anti-tumor activity of γδ T cells in experimental models and the anti-tumor activity of human γδ T cells. In this article, we first review the reciprocal interactions between neutrophils, tumor cells and T lymphocytes with a special focus on their interplay with γδ T cells, followed by the presentation of our own recent results. We have previously shown that zoledronic acid (ZOL)-activated neutrophils inhibit γδ T-cell proliferation due to the production of reactive oxygen species, arginase-1 and serine proteases. We now demonstrate that killing of ductal pancreatic adenocarcinoma (PDAC) cells by freshly isolated resting human γδ T cells was reduced in the presence of neutrophils and even more pronounced so after activation of neutrophils with ZOL. In contrast, direct T-cell receptor-dependent activation by γδ T cell-specific pyrophosphate antigens or by bispecific antibodies enhanced the cytotoxic activity and cytokine/granzyme B production of resting human γδ T cells, thereby overriding the suppression by ZOL-activated neutrophils. Additionally, the coculture of purified neutrophils with autologous short-term expanded γδ T cells enhanced rather than inhibited γδ T-cell cytotoxicity against PDAC cells. Purified neutrophils alone also exerted a small but reproducible lysis of PDAC cells which was further enhanced in the presence of γδ T cells. The latter set-up was associated with improved granzyme B and IFN-γ release which was further increased in the presence of ZOL. Our present results demonstrate that the presence of neutrophils can enhance the killing capacity of activated γδ T cells. We discuss these results in the broader context of regulatory interactions between neutrophils and T lymphocytes.
Collapse
Affiliation(s)
- Hans-Heinrich Oberg
- Institute of Immunology, Christian-Albrechts-University of Kiel, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Daniela Wesch
- Institute of Immunology, Christian-Albrechts-University of Kiel, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Shirin Kalyan
- Clinical Research Development Laboratory, Department of Medicine, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts-University of Kiel, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
43
|
Abstract
Three decades of research in hematopoietic stem cell transplantation and HIV/AIDS fields have shaped a picture of immune restoration disorders. This manuscript overviews the molecular biology of interferon networks, the molecular pathogenesis of immune reconstitution inflammatory syndrome, and post-hematopoietic stem cell transplantation immune restoration disorders (IRD). It also summarizes the effects of thymic involution on T cell diversity, and the results of the assessment of diagnostic biomarkers of IRD, and tested targeted immunomodulatory treatments.
Collapse
Affiliation(s)
- Hesham Mohei
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | - Usha Kellampalli
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | | |
Collapse
|
44
|
Neutrophils Dampen Adaptive Immunity in Brucellosis. Infect Immun 2019; 87:IAI.00118-19. [PMID: 30804100 PMCID: PMC6479033 DOI: 10.1128/iai.00118-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/18/2019] [Indexed: 01/03/2023] Open
Abstract
Brucella organisms are intracellular stealth pathogens of animals and humans. The bacteria overcome the assault of innate immunity at early stages of an infection. Brucella organisms are intracellular stealth pathogens of animals and humans. The bacteria overcome the assault of innate immunity at early stages of an infection. Removal of polymorphonuclear neutrophils (PMNs) at the onset of adaptive immunity against Brucella abortus favored bacterial elimination in mice. This was associated with higher levels of interferon gamma (IFN-γ) and a higher proportion of cells expressing interleukin 6 (IL-6) and inducible nitric oxide synthase (iNOS), compatible with M1 macrophages, in PMN-depleted B. abortus-infected (PMNd-Br) mice. At later times in the acute infection phase, the amounts of IFN-γ fell while IL-6, IL-10, and IL-12 became the predominant cytokines in PMNd-Br mice. IL-4, IL-1β, and tumor necrosis factor alpha (TNF-α) remained at background levels at all times of the infection. Depletion of PMNs at the acute stages of infection promoted the premature resolution of spleen inflammation. The efficient removal of bacteria in the PMNd-Br mice was not due to an increase of antibodies, since the immunoglobulin isotype responses to Brucella antigens were dampened. Anti-Brucella antibodies abrogated the production of IL-6, IL-10, and IL-12 but did not affect the levels of IFN-γ at later stages of infection in PMNd-Br mice. These results demonstrate that PMNs have an active role in modulating the course of B. abortus infection after the adaptive immune response has already developed.
Collapse
|
45
|
Castell SD, Harman MF, Morón G, Maletto BA, Pistoresi-Palencia MC. Neutrophils Which Migrate to Lymph Nodes Modulate CD4 + T Cell Response by a PD-L1 Dependent Mechanism. Front Immunol 2019; 10:105. [PMID: 30761151 PMCID: PMC6362305 DOI: 10.3389/fimmu.2019.00105] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/14/2019] [Indexed: 01/20/2023] Open
Abstract
It is well known that neutrophils are rapidly recruited to a site of injury or infection and perform a critical role in pathogen clearance and inflammation. However, they are also able to interact with and regulate innate and adaptive immune cells and some stimuli induce the migration of neutrophils to lymph nodes (LNs). Previously, we demonstrated that the immune complex (IC) generated by injecting OVA into the footpad of OVA/CFA immunized mice induced the migration of OVA+ neutrophils to draining LNs (dLNs). Here we investigate the effects of these neutrophils which reach dLNs on CD4+ T cell response. Our findings here strongly support a dual role for neutrophils in dLNs regarding CD4+ T cell response modulation. On the one hand, the CD4+ T cell population expands after the influx of OVA+ neutrophils to dLNs. These CD4+ T cells enlarge their proliferative response, activation markers and IL-17 and IFN-γ cytokine production. On the other hand, these neutrophils also restrict CD4+ T cell expansion. The neutrophils in the dLNs upregulate PD-L1 molecules and are capable of suppressing CD4+ T cell proliferation. These results indicate that neutrophils migration to dLNs have an important role in the homeostasis of adaptive immunity. This report describes for the first time that the influx of neutrophils to dLNs dependent on IC presence improves CD4+ T cell response, at the same time controlling CD4+ T cell proliferation through a PD-L1 dependent mechanism.
Collapse
Affiliation(s)
- Sofía D Castell
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - María F Harman
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - Gabriel Morón
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - Belkys A Maletto
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - María C Pistoresi-Palencia
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| |
Collapse
|
46
|
Lambert S, Hambro CA, Johnston A, Stuart PE, Tsoi LC, Nair RP, Elder JT. Neutrophil Extracellular Traps Induce Human Th17 Cells: Effect of Psoriasis-Associated TRAF3IP2 Genotype. J Invest Dermatol 2018; 139:1245-1253. [PMID: 30528823 DOI: 10.1016/j.jid.2018.11.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/08/2018] [Accepted: 11/23/2018] [Indexed: 01/14/2023]
Abstract
Psoriasis lesions are rich in IL-17-producing T cells as well as neutrophils, which release webs of DNA-protein complexes known as neutrophil extracellular traps (NETs). Because we and others have observed increased NETosis in psoriatic lesions, we hypothesized that NETs contribute to increased T helper type 17 (Th17) cells in psoriasis. After stimulating peripheral blood mononuclear cells with anti-CD3/CD28 beads for 7 days, we found significantly higher percentages of CD3+CD4+IL-17+ (Th17) cells in the presence versus absence of NETs, as assessed by flow cytometry, IL-17 ELISA, and IL17A/F and RORC mRNAs. Memory, but not naïve, T cells were competent and monocytes were required for CD3/CD28-mediated Th17 induction, with or without NETs. Th17 induction was enhanced by the T allele of rs33980500 (T/C), a psoriasis risk-associated variant in the TRAF3IP2 gene encoding the D10N variant of Act1, a key mediator of IL-17 signal transduction. Global transcriptome analysis of CD3/CD28-stimulated peripheral blood mononuclear cells by RNA sequencing confirmed the stimulatory effects of NETs, demonstrated NET-induced enhancement of cytokine gene expression, and verified that the effect of Act1 D10N was greater in the presence of NETs. Collectively, these results implicate NETs and the Act1 D10N variant in human Th17 induction from peripheral blood mononuclear cells, with ramifications for immunogenetic studies of psoriasis and other autoimmune diseases.
Collapse
Affiliation(s)
- Sylviane Lambert
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Caely A Hambro
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrew Johnston
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Philip E Stuart
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA; Department of Computational Medicine and Bioinformatics, Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Rajan P Nair
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - James T Elder
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA; Ann Arbor Veterans Affairs Hospital, Ann Arbor, Michigan, USA.
| |
Collapse
|
47
|
Baban B, Marchetti C, Khodadadi H, Malik A, Emami G, Lin PC, Arbab AS, Riccardi C, Mozaffari MS. Glucocorticoid-Induced Leucine Zipper Promotes Neutrophil and T-Cell Polarization with Protective Effects in Acute Kidney Injury. J Pharmacol Exp Ther 2018; 367:483-493. [PMID: 30301736 DOI: 10.1124/jpet.118.251371] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/01/2018] [Indexed: 12/11/2022] Open
Abstract
The glucocorticoid-induced leucine zipper (GILZ) mediates anti-inflammatory effects of glucocorticoids. Acute kidney injury (AKI) mobilizes immune/inflammatory mechanisms, causing tissue injury, but the impact of GILZ in AKI is not known. Neutrophils play context-specific proinflammatory [type 1 neutrophil (N1)] and anti-inflammatory [type 2 neutrophil (N2)] functional roles. Also, regulatory T lymphocytes (Tregs) and regulatory T-17 (Treg17) cells exert counterinflammatory effects, including the suppression of effector T lymphocytes [e.g., T-helper (Th) 17 cells]. Thus, utilizing cell preparations of mice kidneys subjected to AKI or sham operation, we determined the effects of GILZ on T cells and neutrophil subtypes in the context of its renoprotective effect; these studies used the transactivator of transcription (TAT)-GILZ or the TAT peptide. AKI increased N1 and Th-17 cells but reduced N2, Tregs, and Treg17 cells in association with increased interleukin (IL)-17+ but reduced IL-10+ cells accompanied with the disruption of mitochondrial membrane potential (ψ m) and increased apoptosis/necrosis compared with sham kidneys. TAT-GILZ, compared with TAT, treatment reduced N1 and Th-17 cells but increased N2 and Tregs, without affecting Treg17 cells, in association with a reduction in IL-17+ cells but an increase in IL-10+ cells; TAT-GILZ caused less disruption of ψ m and reduced cell death in AKI. Importantly, TAT-GILZ increased perfusion of the ischemic-reperfused kidney but reduced tissue edema compared with TAT. Utilizing splenic T cells and bone marrow-derived neutrophils, we further showed marked reduction in the proliferation of Th cells in response to TAT-GILZ compared with response to TAT. Collectively, the results indicate that GILZ exerts renoprotection accompanied by the upregulation of the regulatory/suppressive arm of immunity in AKI, likely via regulating cross talk between T cells and neutrophils.
Collapse
Affiliation(s)
- Babak Baban
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Cristina Marchetti
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Hesam Khodadadi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Aneeq Malik
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Golnaz Emami
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Ping-Chang Lin
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Ali S Arbab
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Carlo Riccardi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| | - Mahmood S Mozaffari
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia (B.B., H.K., A.M., G.E., M.S.M.) and Georgia Cancer Center (P.-C.L., A.S.A.), Augusta University, Augusta, Georgia; and Department of Medicine, University of Perugia, Perugia, Italy (C.M., C.R.)
| |
Collapse
|
48
|
Tumour-elicited neutrophils engage mitochondrial metabolism to circumvent nutrient limitations and maintain immune suppression. Nat Commun 2018; 9:5099. [PMID: 30504842 PMCID: PMC6269473 DOI: 10.1038/s41467-018-07505-2] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 10/28/2018] [Indexed: 02/08/2023] Open
Abstract
Neutrophils are a vital component of immune protection, yet in cancer they may promote tumour progression, partly by generating reactive oxygen species (ROS) that disrupts lymphocyte functions. Metabolically, neutrophils are often discounted as purely glycolytic. Here we show that immature, c-Kit+ neutrophils subsets can engage in oxidative mitochondrial metabolism. With limited glucose supply, oxidative neutrophils use mitochondrial fatty acid oxidation to support NADPH oxidase-dependent ROS production. In 4T1 tumour-bearing mice, mitochondrial fitness is enhanced in splenic neutrophils and is driven by c-Kit signalling. Concordantly, tumour-elicited oxidative neutrophils are able to maintain ROS production and T cell suppression when glucose utilisation is restricted. Consistent with these findings, peripheral blood neutrophils from patients with cancer also display increased immaturity, mitochondrial content and oxidative phosphorylation. Together, our data suggest that the glucose-restricted tumour microenvironment induces metabolically adapted, oxidative neutrophils to maintain local immune suppression. Neutrophils normally fulfil their metabolic demands by glycolysis and have limited mitochondrial activity. Here the authors show that tumours promote neutrophils adapted to oxidative mitochondria metabolism that function in the glucose-restrained tumour microenvironment to promote tumour growth by maintaining local immune suppression.
Collapse
|
49
|
de Koning C, Langenhorst J, van Kesteren C, Lindemans CA, Huitema ADR, Nierkens S, Boelens JJ. Innate Immune Recovery Predicts CD4 + T Cell Reconstitution after Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2018; 25:819-826. [PMID: 30359735 DOI: 10.1016/j.bbmt.2018.10.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/15/2018] [Indexed: 02/06/2023]
Abstract
Innate immune cells are the first to recover after allogeneic hematopoietic cell transplantation (HCT). Nevertheless, reports of innate immune cell recovery and their relation to adaptive recovery after HCT are largely lacking. Especially predicting CD4+ T cell reconstitution is of clinical interest, because this parameter directly associates with survival chances after HCT. We evaluated whether innate recovery relates to CD4+ T cell reconstitution probability and investigated differences between innate recovery after cord blood transplantation (CBT) and bone marrow transplantation (BMT). We developed a multivariate, combined nonlinear mixed-effects model for monocytes, neutrophils, and natural killer (NK) cell recovery after transplantation. A total of 205 patients undergoing a first HCT (76 BMT, 129 CBT) between 2007 and 2016 were included. The median age was 7.3years (range, .16 to 23). Innate recovery was highly associated with CD4+ T cell reconstitution probability (P < .001) in multivariate analysis correcting for covariates. Monocyte (P < .001), neutrophil (P < .001), and NK cell (P < .001) recovery reached higher levels during the first 200days after CBT compared with BMT. The higher innate recovery after CBT may be explained by increased proliferation capacity (measured by Ki-67 expression) of innate cells in CB grafts compared with BM grafts (P = .041) and of innate cells in vivo after CBT compared with BMT (P = .048). At an individual level, patients with increased innate recovery after either CBT or BMT had received grafts with higher proliferating innate cells (CB; P = .004, BM; P = .01, respectively). Our findings implicate the use of early innate immune monitoring to predict the chance of CD4+ T cell reconstitution after HCT, with respect to higher innate recovery after CBT compared with BMT.
Collapse
Affiliation(s)
- Coco de Koning
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jurgen Langenhorst
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Charlotte van Kesteren
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Caroline A Lindemans
- Pediatric Blood and Marrow Transplantation Program, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Alwin D R Huitema
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Stefan Nierkens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands; Pediatric Blood and Marrow Transplantation Program, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jaap Jan Boelens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands; Pediatric Blood and Marrow Transplantation Program, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands; Pediatric Stem Cell Transplant and Cellular Therapies, Memorial Sloan Kettering Cancer Center, New York, NY, USA..
| |
Collapse
|
50
|
Costa S, Bevilacqua D, Cassatella MA, Scapini P. Recent advances on the crosstalk between neutrophils and B or T lymphocytes. Immunology 2018; 156:23-32. [PMID: 30259972 DOI: 10.1111/imm.13005] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/12/2018] [Accepted: 09/21/2018] [Indexed: 12/21/2022] Open
Abstract
An increasing body of literature supports a role for neutrophils as players in the orchestration of adaptive immunity. During acute and chronic inflammatory conditions, neutrophils rapidly migrate not only to sites of inflammation, but also to draining lymph nodes and spleen, where they engage bidirectional interactions with B- and T-lymphocyte subsets. Accordingly, a relevant role of neutrophils in modulating B-cell responses under homeostatic conditions has recently emerged. Moreover, specialized immunoregulatory properties towards B or T cells acquired by distinct neutrophil populations, originating under pathological conditions, have been consistently described. In this article, we summarize the most recent data from human studies and murine models on the ability of neutrophils to modulate adaptive immune responses under physiological and pathological conditions and the mechanisms behind these processes.
Collapse
Affiliation(s)
- Sara Costa
- Department of Medicine, Section of General Pathology, School of Medicine, University of Verona, Verona, Italy
| | - Dalila Bevilacqua
- Department of Medicine, Section of General Pathology, School of Medicine, University of Verona, Verona, Italy
| | - Marco A Cassatella
- Department of Medicine, Section of General Pathology, School of Medicine, University of Verona, Verona, Italy
| | - Patrizia Scapini
- Department of Medicine, Section of General Pathology, School of Medicine, University of Verona, Verona, Italy
| |
Collapse
|