1
|
Moon CY, Belabed M, Park MD, Mattiuz R, Puleston D, Merad M. Dendritic cell maturation in cancer. Nat Rev Cancer 2025; 25:225-248. [PMID: 39920276 PMCID: PMC11954679 DOI: 10.1038/s41568-024-00787-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 02/09/2025]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that are present at low abundance in the circulation and tissues; they serve as crucial immune sentinels by continually sampling their environment, migrating to secondary lymphoid organs and shaping adaptive immune responses through antigen presentation. Owing to their ability to orchestrate tolerogenic or immunogenic responses to a specific antigen, DCs have a pivotal role in antitumour immunity and the response to immune checkpoint blockade and other immunotherapeutic approaches. The multifaceted functions of DCs are acquired through a complex, multistage process called maturation. Although the role of inflammatory triggers in driving DC maturation was established decades ago, less is known about DC maturation in non-inflammatory contexts, such as during homeostasis and in cancer. The advent of single-cell technologies has enabled an unbiased, high-dimensional characterization of various DC states, including mature DCs. This approach has clarified the molecular programmes associated with DC maturation and also revealed how cancers exploit these pathways to subvert immune surveillance. In this Review, we discuss the mechanisms by which cancer disrupts DC maturation and highlight emerging therapeutic opportunities to modulate DC states. These insights could inform the development of DC-centric immunotherapies, expanding the arsenal of strategies to enhance antitumour immunity.
Collapse
Affiliation(s)
- Chang Yoon Moon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meriem Belabed
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Park
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raphaël Mattiuz
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Puleston
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
2
|
Bosteels V, Janssens S. Striking a balance: new perspectives on homeostatic dendritic cell maturation. Nat Rev Immunol 2025; 25:125-140. [PMID: 39289483 DOI: 10.1038/s41577-024-01079-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2024] [Indexed: 09/19/2024]
Abstract
Dendritic cells (DCs) are crucial gatekeepers of the balance between immunity and tolerance. They exist in two functional states, immature or mature, that refer to an information-sensing versus an information-transmitting state, respectively. Historically, the term DC maturation was used to describe the acquisition of immunostimulatory capacity by DCs following their triggering by pathogens or tissue damage signals. As such, immature DCs were proposed to mediate tolerance, whereas mature DCs were associated with the induction of protective T cell immunity. Later studies have challenged this view and unequivocally demonstrated that two distinct modes of DC maturation exist, homeostatic and immunogenic DC maturation, each with a distinct functional outcome. Therefore, the mere expression of maturation markers cannot be used to predict immunogenicity. How DCs become activated in homeostatic conditions and maintain tolerance remains an area of intense debate. Several recent studies have shed light on the signals driving the homeostatic maturation programme, especially in the conventional type 1 DC (cDC1) compartment. Here, we highlight our growing understanding of homeostatic DC maturation and the relevance of this process for immune tolerance.
Collapse
Affiliation(s)
- Victor Bosteels
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sophie Janssens
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.
| |
Collapse
|
3
|
Bu S, Ling JY, Wu X, Zhang L, Shi X, Huang L, Zhao Z, Yang Y, Xiang Z, Liu YU, Liu Y, Zhang Y. Downregulation of MerTK in circulating T cells of patients with non-proliferative diabetic retinopathy. Front Endocrinol (Lausanne) 2025; 15:1509445. [PMID: 39845889 PMCID: PMC11750652 DOI: 10.3389/fendo.2024.1509445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
Objective To explore the differential gene expression in peripheral blood immune cells of individuals with type 2 diabetes mellitus (DM), comparing those with and without non-proliferative diabetic retinopathy (NPDR). Methods From a pool of 126 potential participants, 60 were selected for detailed analysis. This group included 12 healthy donors (HDs), 22 individuals with DM, and 26 with NPDR. We analyzed peripheral blood mononuclear cells (PBMCs) using RNA sequencing and quantitative PCR (qPCR) to pinpoint differentially expressed genes (DEGs). Western blot and flow cytometry were also employed to evaluate the protein expression of specific genes. Results In patients with NPDR compared to those with DM alone, MerTK-a gene implicated in inherited retinal dystrophies due to its mutations-was notably downregulated in PBMCs. Through flow cytometry, we assessed the protein levels and cellular distribution of MerTK, finding a predominant expression in monocytes and myeloid-derived suppressor cells (MDSCs), with a marked reduction in CD4+ and CD8+ T cells, as well as in natural killer T (NKT) cells. Patients with DM demonstrated a significant deviation in the PBMCs composition, particularly in B cells, CD4+ T cells, and NK cells, when compared to HDs. Conclusions The study indicates that MerTK expression in T cells within PBMCs could act as a viable blood biomarker for NPDR risk in patients with DM. Furthermore, the regulation of T cells by MerTK might represent a critical pathway through which DM evolves into NPDR.
Collapse
Affiliation(s)
- Shimiao Bu
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiang-Yue Ling
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiaojun Wu
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Liting Zhang
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiangyu Shi
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Lang Huang
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zheng Zhao
- Department of Neurology, The First Affiliated Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Ying Yang
- Department of Neurology, The First Affiliated Hospital, School of Medicine, Jinan University, Guangzhou, China
| | - Zongqin Xiang
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yong U. Liu
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Yufeng Liu
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuehong Zhang
- Department of Ophthalmology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory for Neuroimmunology in Health and Diseases, Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
4
|
Mor G, Singh A, Yang J, Adzibolosu N, Cai S, Kauf E, Yang L, Li Q, Li H, Werner A, Parthasarathy S, Ding J, Fortier J, Rodriguez-Garcia M, Diao LH. Decoding Functional and Developmental Trajectories of Tissue-Resident Uterine Dendritic Cells Through Integrative Omics. RESEARCH SQUARE 2024:rs.3.rs-5424920. [PMID: 39606471 PMCID: PMC11601813 DOI: 10.21203/rs.3.rs-5424920/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Uterine dendritic cells (uDCs) are critical for endometrial function, yet their origin, molecular characteristics, and specific roles during the pre- and post-implantation periods in the human endometrium remain largely unknown. The complexity of the endometrial environment makes defining the contributions of uDCs subtypes challenging. We hypothesize that distinct uDC subsets carry out specialized functions, and that resident progenitor DCs generate these subtypes. Employing single-cell RNA sequencing on uterine tissues collected across different menstrual phases and during early pregnancy, we identify several uDCs subtypes, including resident progenitor DCs. CITE-seq was performed on endometrial single-cell suspensions to link surface protein expression with key genes identified by the RNAseq analysis. Our analysis revealed the developmental trajectory of the uDCs along with the distinct functional roles of each uDC subtype, including immune regulation, antigen presentation, and creating a conducive environment for embryo implantation. This study provides a comprehensive characterization of uDCs, serving as a foundational reference for future studies for better understanding female reproductive disorders such as infertility and pregnancy complications.
Collapse
Affiliation(s)
| | | | - Jing Yang
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen 361102, China
| | | | - Songchen Cai
- Shenzhen Zhongshan Obstetrics & Gynecology Hospital
| | | | | | - Qiyuan Li
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Hanjie Li
- Shenzhen Institutes of Advanced Technology
| | | | | | | | | | | | | |
Collapse
|
5
|
Zewdie EY, Edwards GM, Hunter DM, Earp HS, Holtzhausen A. MerTK Induces Dysfunctional Dendritic Cells by Metabolic Reprogramming. Cancer Immunol Res 2024; 12:1268-1285. [PMID: 38976507 PMCID: PMC11371516 DOI: 10.1158/2326-6066.cir-23-0666] [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: 08/15/2023] [Revised: 01/05/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024]
Abstract
Checkpoint inhibitors, specifically anti-programmed cell death protein 1 (PD1), have shown success in treating metastatic melanoma; however, some patients develop resistance. Dendritic cells (DC) play a key role in initiating an immune response, but in certain circumstances they become ineffective. We investigated the role of MerTK, a receptor tyrosine kinase responsible for myeloid cell clearance of dead cells, in the regulation of DC function and metabolism in the tumor microenvironment. Tumors resistant to anti-PD1 exhibited increased levels of MerTK+ DCs. Treating wild-type DCs with apoptotic melanoma cells in vitro resulted in increased MerTK expression, elevated mitochondrial respiration and fatty acid oxidation, and reduced T-cell stimulatory capacity, all characteristics of dysfunctional DCs. In contrast, dead cells had only limited effect on the metabolism of MerTK-deficient DCs, which instead maintained an antigen-presenting, stimulatory phenotype. The efficacy of anti-PD1 to slow tumor progression and induce antigen specific T-cell infiltration was markedly increased in mice with selective ablation of MerTK in the DC compartment, suggesting the possibility of therapeutically targeting MerTK to modulate DC metabolism and function and enhance anti-PD1 therapy.
Collapse
Affiliation(s)
- Eden Y. Zewdie
- UNC Lineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, U.S.A
- UNC Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, U.S.A
| | - George M. Edwards
- UNC Lineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, U.S.A
| | - Debra M. Hunter
- UNC Lineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, U.S.A
| | - H. Shelton Earp
- UNC Lineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, U.S.A
- UNC Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, U.S.A
- UNC Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, U.S.A
| | - Alisha Holtzhausen
- UNC Lineberger Comprehensive Cancer Center, Department of Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, U.S.A
- UNC Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, U.S.A
| |
Collapse
|
6
|
Bian W, Hu X, Xiao R, Yao R, Zhang B, Zhu M, Liu T, Liu Y, Li J, Lin P, Xie A, Li F, Ling D. Catalytic Dual-Mode Immunotherapy: Anisotropic AuPt Heterostructure Decorated with Starry Pt Nanoclusters for Robust Cancer Photometalloimmunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403116. [PMID: 38816935 PMCID: PMC11267392 DOI: 10.1002/advs.202403116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/05/2024] [Indexed: 06/01/2024]
Abstract
To overcome current limitations in photoimmunotherapy, such as insufficient tumor antigen generation and a subdued immune response, a novel photo-/metallo dual-mode immunotherapeutic agent (PMIA) is introduced for potent near-infrared (NIR) light-triggered cancer therapy. PMIA features a dumbbell-like AuPt heterostructure decorated with starry Pt nanoclusters, meticulously engineered for enhancing plasmonic catalysis through multi-dimensional regulation of Pt growth on Au nanorods. Under NIR laser exposure, end-tipped Pt nanoclusters induce efficient electron-hole spatial separation along the longitudinal axis, resulting in radial and axial electron distribution polarization, conferring unique anisotropic properties to PMIA. Additionally, starry Pt nanoclusters on the sides of Au nanorods augment the local electron enrichment field. Validated through finite-difference time-domain analysis and Raman scattering, this configuration fosters local electron enrichment, facilitating robust reactive oxygen species generation for potent photoimmunotherapy. Moreover, Pt nanoclusters facilitate Pt2+ ion release, instigating intranuclear DNA damage and inducing synergistic immunogenic cell death (ICD) for metalloimmunotherapy. Consequently, PMIA elicits abundant danger-associated molecular patterns, promotes T cell infiltration, and triggers systemic immune responses, effectively treating primary and distant tumors, inhibiting metastasis in vivo. This study unveils a pioneering dual-mode ICD amplification strategy driven by NIR light, synergistically integrating photoimmunotherapy and metalloimmunotherapy, culminating in potent cancer photometalloimmunotherapy.
Collapse
Affiliation(s)
- Wei Bian
- Department of Breast SurgeryFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- Key Laboratory of Cellular Physiology at Shanxi Medical UniversityMinistry of EducationTaiyuan030000China
| | - Xi Hu
- School of PharmacyAnhui University of Chinese MedicineHefei230012China
| | - Ruixue Xiao
- Frontiers Science Center for Transformative MoleculesSchool of Chemistry and Chemical EngineeringSchool of Biomedical EngineeringNational Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240China
| | - Rui Yao
- Key Laboratory of Cellular Physiology at Shanxi Medical UniversityMinistry of EducationTaiyuan030000China
| | - Bo Zhang
- Frontiers Science Center for Transformative MoleculesSchool of Chemistry and Chemical EngineeringSchool of Biomedical EngineeringNational Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240China
- WLA LaboratoriesShanghai201203China
| | - Mingjian Zhu
- Institute of PharmaceuticsHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Tianqi Liu
- Key Laboratory of Cellular Physiology at Shanxi Medical UniversityMinistry of EducationTaiyuan030000China
| | - Yamin Liu
- Frontiers Science Center for Transformative MoleculesSchool of Chemistry and Chemical EngineeringSchool of Biomedical EngineeringNational Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240China
| | - Jing Li
- Department of Breast SurgeryFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Peihua Lin
- Frontiers Science Center for Transformative MoleculesSchool of Chemistry and Chemical EngineeringSchool of Biomedical EngineeringNational Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240China
| | - An Xie
- School of PharmacyAnhui University of Chinese MedicineHefei230012China
| | - Fangyuan Li
- Songjiang Institute and Songjiang HospitalShanghai Key Laboratory of Emotions and Affective DisordersShanghai Jiao Tong University School of MedicineShanghai200025China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang ProvinceHangzhou310009China
| | - Daishun Ling
- Frontiers Science Center for Transformative MoleculesSchool of Chemistry and Chemical EngineeringSchool of Biomedical EngineeringNational Center for Translational MedicineShanghai Jiao Tong UniversityShanghai200240China
- WLA LaboratoriesShanghai201203China
| |
Collapse
|
7
|
Ma Y, Jiang T, Zhu X, Xu Y, Wan K, Zhang T, Xie M. Efferocytosis in dendritic cells: an overlooked immunoregulatory process. Front Immunol 2024; 15:1415573. [PMID: 38835772 PMCID: PMC11148234 DOI: 10.3389/fimmu.2024.1415573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 05/09/2024] [Indexed: 06/06/2024] Open
Abstract
Efferocytosis, the process of engulfing and removing apoptotic cells, plays an essential role in preserving tissue health and averting undue inflammation. While macrophages are primarily known for this task, dendritic cells (DCs) also play a significant role. This review delves into the unique contributions of various DC subsets to efferocytosis, highlighting the distinctions in how DCs and macrophages recognize and handle apoptotic cells. It further explores how efferocytosis influences DC maturation, thereby affecting immune tolerance. This underscores the pivotal role of DCs in orchestrating immune responses and sustaining immune equilibrium, providing new insights into their function in immune regulation.
Collapse
Affiliation(s)
- Yanyan Ma
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tangxing Jiang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xun Zhu
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yizhou Xu
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ke Wan
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tingxuan Zhang
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Miaorong Xie
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Parsons EL, Kim JS, Malloy AMW. Development of innate and adaptive immunity to RSV in young children. Cell Immunol 2024; 399-400:104824. [PMID: 38615612 DOI: 10.1016/j.cellimm.2024.104824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/29/2024] [Accepted: 03/25/2024] [Indexed: 04/16/2024]
Abstract
Infection of the respiratory tract with respiratory syncytial virus (RSV) is common and occurs repeatedly throughout life with most severe disease occurring at the extremes of age: in young infants and the elderly. Effective anti-viral therapeutics are not available and therefore prevention has been the primary strategy for reducing the disease burden. Our current understanding of respiratory mucosal cell biology and the immune response within the respiratory tract is inadequate to prevent infection caused by a pathogen like RSV that does not disseminate outside of this environment. Gaps in our understanding of the activation of innate and adaptive immunity in response to RSV and the role of age upon infection also limit improvements in the design of therapeutics and vaccines for young infants. However, advancements in structural biology have improved our ability to characterize antibodies against viral proteins and in 2023 the first vaccines for those over 60 years and pregnant women became available, potentially reducing the burden of disease. This review will examine our current understanding of the critical facets of anti-RSV immune responses in infants and young children as well as highlight areas where more research is needed.
Collapse
Affiliation(s)
| | - Jisung S Kim
- Uniformed Services University, Bethesda, MD, USA; Henry M. Jackson Foundation, Bethesda, MD, USA
| | | |
Collapse
|
9
|
Hildreth AD, Padilla ET, Gupta M, Wong YY, Sun R, Legala AR, O'Sullivan TE. Adipose cDC1s contribute to obesity-associated inflammation through STING-dependent IL-12 production. Nat Metab 2023; 5:2237-2252. [PMID: 37996702 DOI: 10.1038/s42255-023-00934-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
Obesity is associated with chronic low-grade white adipose tissue (WAT) inflammation that can contribute to the development of insulin resistance in mammals. Previous studies have identified interleukin (IL)-12 as a critical upstream regulator of WAT inflammation and metabolic dysfunction during obesity. However, the cell types and mechanisms that initiate WAT IL-12 production remain unclear. Here we show that conventional type 1 dendritic cells (cDC1s) are the cellular source of WAT IL-12 during obesity through analysis of mouse and human WAT single-cell transcriptomic datasets, IL-12 reporter mice and IL-12p70 protein levels by enzyme-linked immunosorbent assay. We demonstrate that cDC1s contribute to obesity-associated inflammation by increasing group 1 innate lymphocyte interferon-γ production and inflammatory macrophage accumulation. Inducible depletion of cDC1s increased WAT insulin sensitivity and systemic glucose tolerance during diet-induced obesity. Mechanistically, endocytosis of apoptotic bodies containing self-DNA by WAT cDC1s drives stimulator of interferon genes (STING)-dependent IL-12 production. Together, these results suggest that WAT cDC1s act as critical regulators of adipose tissue inflammation and metabolic dysfunction during obesity.
Collapse
Affiliation(s)
- Andrew D Hildreth
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Eddie T Padilla
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Meha Gupta
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yung Yu Wong
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ryan Sun
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Akshara R Legala
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|