1
|
Wu K, Xu Y, Liu P, Chen K, Zhao Y. STING inhibitors and degraders: Potential therapeutic agents in inflammatory diseases. Eur J Med Chem 2025; 291:117632. [PMID: 40262301 DOI: 10.1016/j.ejmech.2025.117632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/30/2025] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
The regulation of the STING (stimulator of interferon genes) pathway represents a promising target for a range of inflammatory diseases. This review provides an overview of the structure of STING and discusses the mechanisms by which the cyclic GMP-AMP synthase (cGAS)-STING pathway is associated with various autoinflammatory and autoimmune diseases. We explore how targeting STING inhibition or degradation can alleviate excessive inflammatory signaling and improve efficacy. Emerging strategies include inhibiting STING expression by covalently binding compounds or using ligands that target the binding pocket. In addition, selective degradation of STING via the ubiquitin-proteasome system or the lysosomal pathway shows promise. In addition, we explore the implications of modulating the cGAS-STING pathway in the context of various inflammatory diseases. Finally, we summarize the chemical properties of recently developed STING compounds and their potential clinical applications. By comprehensively reviewing the current understanding of the role of STING in inflammation and the therapeutic potential of targeting STING, we aim to identify new avenues of intervention that could improve outcomes for patients with inflammatory diseases. This review highlights the important role of STING in the regulation of inflammation and its potential as a target for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Kerong Wu
- Department of General Surgery, Clinical Translational Research Center for Surgical Infection and Immunity of Nanjing Medical University, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yiwen Xu
- Department of General Surgery, Clinical Translational Research Center for Surgical Infection and Immunity of Nanjing Medical University, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Peizhao Liu
- Research Institute of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | - Kexin Chen
- Department of General Surgery, Clinical Translational Research Center for Surgical Infection and Immunity of Nanjing Medical University, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yun Zhao
- Department of General Surgery, Clinical Translational Research Center for Surgical Infection and Immunity of Nanjing Medical University, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
2
|
Haas G, Seiler M, Nguyen J, Troxler L, Pennarun S, Lefebvre E, Benamrouche Y, Loizeau L, Reinbolt C, Liang M, Lin X, Li W, Xia Z, Marques JT, Imler JL. Regulation of detoxifying enzymes expression and restriction of picorna-like virus infection by natural polysaccharide extracts in Drosophila cells. Virology 2025; 607:110513. [PMID: 40163969 DOI: 10.1016/j.virol.2025.110513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
The world is currently witnessing a rise in viral infections, while the availability of antiviral drugs remains limited. Traditional Chinese medicine (TCM) has historically served as a valuable source of novel compounds for disease treatment. In this study, we assessed the antiviral potential of various TCM compounds using Drosophila melanogaster as a model organism. Our findings reveal that natural polysaccharide extracts, prepared from 10 commonly used medicinal herbs or fungi, exhibit antiviral activity against two picorna-like viruses. Importantly, the antiviral effect is not directly attributable to the compound itself but is instead mediated by cellular responses induced by treatment with the extract. We observed that the polysaccharide extract triggers a broad transcriptional response, which partially overlaps with NF-κB pathway activation in Drosophila. However, the antiviral activity of the extract was independent of classical innate immune pathways, such as RNA interference or NF-κB signaling. Instead, the extract appears to uniquely stimulate detoxification pathways, including upregulation of cytochrome P450 and glutathione S-transferase genes, which correlates with its antiviral effects.
Collapse
Affiliation(s)
- Gabrielle Haas
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Mélodie Seiler
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Jenny Nguyen
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Laurent Troxler
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Samuel Pennarun
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Elise Lefebvre
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | | | - Loriane Loizeau
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Cody Reinbolt
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France
| | - Ming Liang
- Infinitus (China) Company Ltd., Guangzhou, China
| | | | - Wenzhi Li
- Infinitus (China) Company Ltd., Guangzhou, China
| | - Zumeng Xia
- Infinitus (China) Company Ltd., Guangzhou, China
| | - Joao T Marques
- Université de Strasbourg, INSERM U1257, CNRS UPR9022, 67084, Strasbourg, France; Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Jean-Luc Imler
- Université de Strasbourg, CNRS UPR9022, 67084, Strasbourg, France.
| |
Collapse
|
3
|
Zhang X, Fam KT, Dai T, Hang HC. Microbiota mechanisms in cancer progression and therapy. Cell Chem Biol 2025; 32:653-677. [PMID: 40334660 DOI: 10.1016/j.chembiol.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/19/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025]
Abstract
The composition of the microbiota in patients has been shown to correlate with cancer progression and response to therapy, highlighting unique opportunities to improve patient outcomes. In this review, we discuss the challenges and advancements in understanding the chemical mechanisms of specific microbiota species, pathways, and molecules involved in cancer progression and treatment. We also describe the modulation of cancer and immunotherapy by the microbiota, along with approaches for investigating microbiota enzymes and metabolites. Elucidating these specific microbiota mechanisms and molecules should offer new opportunities for developing enhanced diagnostics and therapeutics to improve outcomes for cancer patients. Nonetheless, many microbiota mechanisms remain to be determined and require innovative chemical genetic approaches.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Kyong Tkhe Fam
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Tingting Dai
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Howard C Hang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA; Department of Chemistry, Scripps Research, La Jolla, CA 92037, USA.
| |
Collapse
|
4
|
Wadhonkar K, Singh Y, Rughetti A, Das S, Yangdol R, Sk MH, Baig MS. Role of cancer cell-derived exosomal glycoproteins in macrophage polarization. Mol Biol Rep 2025; 52:451. [PMID: 40347313 DOI: 10.1007/s11033-025-10535-x] [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: 03/16/2025] [Accepted: 04/22/2025] [Indexed: 05/12/2025]
Abstract
Cancer is a deadly disease marked by abnormal cell growth, proliferation, and metastasis-the spread of cancer from its origin to distant sites. A key factor in tumor progression is the tumor microenvironment (TME), which significantly influences tumor behavior and response to treatment. Within the TME, interactions between cancer cells and surrounding immune cells, particularly tumor-associated macrophages (TAMs), play a critical role in shaping immune responses. This review focuses on recent findings from a systematic PubMed search regarding cancer cell-derived exosomal glycoproteins and their role in modulating macrophage phenotypes. Tumor-derived exosomes, a type of extracellular vesicle (EV), carry glycoproteins-proteins with attached sugar chains-that can influence macrophage polarization. These glycoproteins can reprogram macrophages into either the M1 phenotype (proinflammatory and anti-tumor) or the M2 phenotype (anti-inflammatory and tumor-supportive). The M1 macrophages inhibit tumor progression, while M2 macrophages support tumor growth by promoting immune suppression and tissue remodeling. Understanding how exosomal glycoproteins drive this polarization offers critical insight into cancer immunology and may pave the way for novel therapeutic strategies targeting the TME.
Collapse
Affiliation(s)
- Khandu Wadhonkar
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Yashi Singh
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Aurelia Rughetti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, University of Rome "Sapienza", Rome, Italy
| | - Soumalya Das
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rigzin Yangdol
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - M Hassan Sk
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India.
| |
Collapse
|
5
|
Mu L, Li J, Lin Z, Zeng Q, Deng L, Wu S, Li J, Yin X, Ye J. MBL regulates phagocytosis and bactericidal activity of macrophages by triggering AKT/NF-κB/Rab5A axis occurred early in vertebrate evolution. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf028. [PMID: 40280188 DOI: 10.1093/jimmun/vkaf028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/05/2025] [Indexed: 04/29/2025]
Abstract
Mannose-binding lectin (MBL) is a vital and versatile component of innate immunity, which plays an essential role in host defense. In mammals, MBL performs multiple functions, including pattern recognition, complement activation, and phagocytosis. Previous studies revealed that OnMBL can promote the opsonophagocytosis of macrophages through its interaction with calreticulin (CRT) in a complement activation-independent manner in Nile tilapia (Oreochromis niloticus). However, the oligomer structural characteristics of MBL and the pathways involved in immune defense mechanisms remain poorly understood. In this study, we identified different oligomer forms of OnMBL in tilapia serum, with significant increases in trimer and tetramer levels present following immunization with Streptococcus agalactiae. Further investigation demonstrated that a higher degree of OnMBL oligomerization enhanced its ability to bind and phagocytose bacteria. Notably, OnMBL promoted the formation of phagolysosomes, which are responsible for degrading and eliminating ingested bacteria. Additionally, OnMBL knockdown caused a marked downregulation of the CD91a postinfection. Moreover, it is confirmed that OnMBL interacted with OnCRT and OnCD91a, with OnCD91a being essential for the OnMBL/CRT complex to facilitate phagocytosis and bacterial clearance. Mechanistic studies revealed that OnMBL/CRT complex enhanced phagocytosis through collaboration with OnCD91a, triggering a positive feedback loop mediated by the AKT/NF-κB/Rab5A signaling axis, thereby boosting macrophage activities and antibacterial immune responses. Therefore, this study elucidates the antibacterial response mechanism of oligomer OnMBL and its receptor in early vertebrates. These findings add to the knowledge regarding the regulatory mechanisms of C-type lectins in fish and provide valuable insights into the evolution of innate immunity.
Collapse
Affiliation(s)
- Liangliang Mu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, P. R. China
| | - Jiadong Li
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, P. R. China
| | - Zhanyao Lin
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, P. R. China
| | - Qingliang Zeng
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, P. R. China
| | - Lu Deng
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, P. R. China
| | - Siqi Wu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, P. R. China
| | - Jun Li
- School of Biological Science, Lake Superior State University, Sault Ste. Marie, MI, United States
| | - Xiaoxue Yin
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, P. R. China
| | - Jianmin Ye
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, P. R. China
- Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, School of Life Sciences, South China Normal University, Guangzhou, P. R. China
| |
Collapse
|
6
|
Vela Navarro N, De Nadai Mundim G, Cudic M. Implications of Mucin-Type O-Glycosylation in Alzheimer's Disease. Molecules 2025; 30:1895. [PMID: 40363702 PMCID: PMC12073284 DOI: 10.3390/molecules30091895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders linked to aging. Major hallmarks of AD pathogenesis include amyloid-β peptide (Aβ) plaques, which are extracellular deposits originating from the processing of the amyloid precursor protein (APP), and neurofibrillary tangles (NFTs), which are intracellular aggregates of tau protein. Recent evidence indicates that disruptions in metal homeostasis and impaired immune recognition of these aggregates trigger neuroinflammation, ultimately driving disease progression. Therefore, a more comprehensive approach is needed to understand the underlying causes of the disease. Patients with AD present abnormal glycan profiles, and most known AD-related molecules are either modified with glycans or involved in glycan regulation. A deeper understanding of how O-glycosylation influences the balance between amyloid-beta peptide production and clearance, as well as microglia's pro- and anti-inflammatory responses, is crucial for deciphering the early pathogenic events of AD. This review aims to provide a comprehensive summary of the extensive research conducted on the role of mucin-type O-glycosylation in the pathogenesis of AD, discussing its role in disease onset and immune recognition.
Collapse
Affiliation(s)
| | | | - Maré Cudic
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Rd., Boca Raton, FL 33431, USA; (N.V.N.); (G.D.N.M.)
| |
Collapse
|
7
|
Li T, Wu X, Li X, Chen M. Cancer-associated fungi: An emerging powerful player in cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2025; 1880:189287. [PMID: 39971202 DOI: 10.1016/j.bbcan.2025.189287] [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: 04/30/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
The role of the human microbiome in cancer has been extensively studied, focusing mainly on bacteria-host interactions and their impact on tumor development and treatment response. However, fungi, an immune-active component of the human microbiome, have received less attention regarding their roles in cancer. Recent studies have identified the widespread and specific colonization and distribution of fungi in multiple sites in patients across various cancer types. Importantly, host-fungal immune interactions significantly influence immune regulation within the tumor microenvironment. The rapid advancement of immune-checkpoint blockade (ICB)-based cancer immunotherapy creates an urgent need for effective biomarkers and synergistic therapeutic targets. Cancer-associated fungi and their associated antifungal immunity demonstrate significant potential and efficacy in enhancing cancer immunotherapy. This review summarizes and discusses the growing evidence of the functions and mechanisms of commensal and pathogenic cancer-associated fungi in cancer immunotherapy. Additionally, we emphasize the potential of fungi as predictive biomarkers and therapeutic targets in cancer immunotherapy.
Collapse
Affiliation(s)
- Tianhang Li
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China; Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.
| | - Xiangyu Wu
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiangyang Li
- Department of Gastrointestinal Tumor Surgery, Nanjing Tianyinshan Hospital, Affiliated Hospital of China Pharmaceutical University, Nanjing, China.
| | - Ming Chen
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China; Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.
| |
Collapse
|
8
|
Ling X, Dong Z, He J, Chen D, He D, Guo R, He Q, Li M. Advances in Polymer-Based Self-Adjuvanted Nanovaccines. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409021. [PMID: 40079071 DOI: 10.1002/smll.202409021] [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: 10/02/2024] [Revised: 02/22/2025] [Indexed: 03/14/2025]
Abstract
Nanovaccines, as a new generation of vaccines, have garnered significant interest due to their exceptional potential in enhancing disease prevention and treatment. Their unique features, such as high stability, antigens protection, prolonged retention, and targeted delivery to lymph nodes, immune cells, and tumors, set them apart as promising candidates in the field of immunotherapy. Polymers, with their superior degradability, capacity to mimic pathogen characteristics, and surface functionality that facilitates modifications, serve as ideal carriers for vaccine components. Polymer-based self-adjuvanted nanovaccines have the remarkable ability to augment immune responses. The inherent adjuvant-like properties of polymers themselves offer a pathway toward more efficient exploitation of nanomaterials and the optimization of nanovaccines. This review article aims to summarize the categorization of polymers and elucidate their mechanisms of action as adjuvants. Additionally, it delves into the advantages and limitations of polymer-based self-adjuvanted nanovaccines in disease management and prevention, providing valuable insights for their design and application. This comprehensive analysis could contribute to the development of more effective and tailored nanovaccines for a wide range of diseases.
Collapse
Affiliation(s)
- Xiaoli Ling
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Ziyan Dong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Jiao He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Dong Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Dan He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Rong Guo
- West China College of Basic Medical Sciences and Forensic Science, Sichuan University, Chengdu, 610041, P. R. China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
9
|
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: 2] [Impact Index Per Article: 2.0] [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
|
10
|
Zhang J, Wu H, Huang Y, Yang Y, Yekefenhazi D, Zou W, Han F. Characterization and Immune Functions of LcβLectin from Large Yellow Croaker ( Larimichthys crocea): A Potential Antiviral Defense Molecule. Int J Mol Sci 2025; 26:3251. [PMID: 40244096 PMCID: PMC11989850 DOI: 10.3390/ijms26073251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/28/2025] [Accepted: 03/30/2025] [Indexed: 04/18/2025] Open
Abstract
Large yellow croaker iridovirus (LYCIV) poses a significant threat to the large yellow croaker (Larimichthys crocea) aquaculture industry due to its rapid transmission and high lethality. Galectins, as evolutionarily conserved carbohydrate-binding lectins and pattern recognition receptors (PRRs) in the innate immune system, play crucial roles in immune responses. In this study, we characterized the beta-galactoside-binding lectin from large yellow croaker (LcβLectin) and explored its potential as a disease resistance gene against LYCIV. The full-length cDNA of LcβLectin was cloned and found to contain conserved elements, such as β-galactoside-binding motifs, HNPR, and WCEEHR domains. Using L. crocea head-kidney macrophages (LCM10), we demonstrated that recombinant LcβLectin significantly inhibits LYCIV-induced cytopathic effects and reduces macrophage apoptosis, highlighting its key role in viral defense. Moreover, the overexpression of LcβLectin in LCM10 cells followed by transcriptomic analysis revealed its substantial regulatory effects on key immune-related signaling pathways, including C-type lectin signaling, p53 signaling, and Toll-like receptor signaling pathways. Collectively, our findings suggest that LcβLectin enhances fish resistance to viral diseases by augmenting immune system function and activating immune-related pathways, providing valuable insights into the innate immune mechanisms of aquatic species and potential strategies for disease prevention in aquaculture.
Collapse
Affiliation(s)
- Jiawei Zhang
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Fisheries College, Jimei University, Xiamen 361000, China; (J.Z.); (H.W.); (Y.H.); (Y.Y.); (D.Y.)
| | - Hongling Wu
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Fisheries College, Jimei University, Xiamen 361000, China; (J.Z.); (H.W.); (Y.H.); (Y.Y.); (D.Y.)
| | - Ying Huang
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Fisheries College, Jimei University, Xiamen 361000, China; (J.Z.); (H.W.); (Y.H.); (Y.Y.); (D.Y.)
| | - Yao Yang
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Fisheries College, Jimei University, Xiamen 361000, China; (J.Z.); (H.W.); (Y.H.); (Y.Y.); (D.Y.)
| | - Dinaer Yekefenhazi
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Fisheries College, Jimei University, Xiamen 361000, China; (J.Z.); (H.W.); (Y.H.); (Y.Y.); (D.Y.)
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH8 9YL, UK
| | - Wenzheng Zou
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Fisheries College, Jimei University, Xiamen 361000, China; (J.Z.); (H.W.); (Y.H.); (Y.Y.); (D.Y.)
| | - Fang Han
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Fisheries College, Jimei University, Xiamen 361000, China; (J.Z.); (H.W.); (Y.H.); (Y.Y.); (D.Y.)
| |
Collapse
|
11
|
Ito T, Guenther C, Ishikawa E, Yabuki T, Nagae M, Nakatani Y, Yamasaki S. Phylogenetic and structural insights into the origin of C-type lectin Mincle in vertebrates. Immunogenetics 2025; 77:18. [PMID: 40119899 PMCID: PMC11929736 DOI: 10.1007/s00251-025-01375-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025]
Abstract
Our bodies are continuously exposed to injurious insults by infection and tissue damage, which are primarily sensed by innate immune receptors to maintain homeostasis. Among such receptors is macrophage-inducible C-type lectin (Mincle, gene symbol CLEC4E), a member of the C-type lectin receptor (CLR) family, which functions as an immune sensor for both pathogens and damaged self. To monitor these injurious stimuli, Mincle recognizes disaccharide-based pathogen-derived glycolipids and monosaccharide-based intracellular metabolites, such as β-glucosylceramide. Mincle is well-conserved among mammals; however, there are questions that remain unclear, such as from which lower vertebrate did it arise and whether the original ligand was self or non-self. Here, we found homologues of Mincle and its signaling subunit Fc receptor γ chain (FcRγ) in lower vertebrates, such as reptiles, amphibians, and fishes. The crystal structure of a Mincle homologue revealed that fish Mincle possesses a narrower sugar-binding pocket than that of mammalian Mincle, and accommodates only monosaccharide moieties. These results suggest that Mincle may have evolved from a self-recognizing receptor, and its sugar-binding pocket widened during evolution, presumably to adapt to disaccharide-based glycolipids derived from life-threatening pathogens.
Collapse
Affiliation(s)
- Taiki Ito
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
| | - Carla Guenther
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
| | - Eri Ishikawa
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
| | - Takae Yabuki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and Drug Delivery Systems (CAMaD), Osaka University, Suita, Osaka, Japan
| | - Masamichi Nagae
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
| | - Yoichiro Nakatani
- Laboratory of Medical and Evolutionary Genomics, Department of Biological Informatics, Bioinformatics Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.
- Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan.
- Center for Advanced Modalities and Drug Delivery Systems (CAMaD), Osaka University, Suita, Osaka, Japan.
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
12
|
Liu X, Wang S, Du X, Wang Y, Mo L, Li H, Qu Z, Wang X, Sun J, Li Y, Wang J. Identification of Disulfidptosis-Related Genes and Molecular Subgroups in Rheumatoid Arthritis for Diagnostic Model and Patient Stratification. J Inflamm Res 2025; 18:4157-4175. [PMID: 40125081 PMCID: PMC11930242 DOI: 10.2147/jir.s505746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
Introduction Cell death contributes to the pathogenesis of rheumatoid arthritis (RA) through various pathways. Disulfidptosis is a recently discovered novel form of cell death characterized by the abnormal accumulation of intracellular disulfide bonds. It remains unclear for the association between RA and disulfidptosis. Methods A comprehensive analysis of three GEO datasets was presented in this study. First, the analysis involved the use of weighted gene co-expression network analysis (WGCNA) and differential analysis and were employed to identify the key module genes related to RA and disulfidptosis-related genes. The machine learning algorithms were used to identify the hub genes. Second, a diagnostic model was constructed for RA based on the hub genes. The nomogram and receiver operating characteristic (ROC) curves were utilized to evaluate the diagnostic value of the model. Third, two RA subtypes were identified based on hub genes by using consensus clustering analysis. Then, the disease activity scores, clinical markers, and immune cells were compared between the two RA subgroups. Finally, the differential expression of hub genes was validated between healthy controls and RA patients by qPCR. Results Four hub genes (KLHL2, POLK, CLEC4D, NXT2) were identified. The expression of the four hub genes was verified to be significantly higher in RA patients compared with healthy controls. The superior diagnostic value of the model was validated, which demonstrated that the model outperforms each hub gene individually. Two subtypes of RA were determined. Patients in cluster A exhibited relatively lower levels of DAS28-CRP, DAS28-ESR, CDAI, SDAI, RF, CRP, and MMP3. In contrast, patients in cluster B had significantly higher levels of the above markers. Conclusion Four hub genes were identified to provide unique insights into the role of disulfidptosis in RA. Additionally, a promising diagnosis model and patient stratification were established based on the hub genes to assess the risk of RA onset and RA disease activity.
Collapse
Affiliation(s)
- Xinyi Liu
- Department of Rheumatology and Immunology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Siyao Wang
- Department of Gastroenterology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Xinru Du
- Department of Rheumatology and Immunology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yulu Wang
- Xi’an Jiaotong University College of Medicine, Xi’an, People’s Republic of China
| | - Lingfei Mo
- Department of Rheumatology and Immunology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Hanchao Li
- Department of Rheumatology and Immunology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Zechao Qu
- Department of Spine Surgery, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Xiaohao Wang
- Department of Spine Surgery, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Jian Sun
- Institute of Endemic Diseases, School of Public Health & Key Laboratory of Trace Elements and Endemic Diseases, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yuanyuan Li
- Department of Rheumatology and Immunology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Jing Wang
- Department of Rheumatology and Immunology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| |
Collapse
|
13
|
Fleischer AB, Amann B, von Toerne C, Degroote RL, Schmalen A, Weißer T, Hauck SM, Deeg CA. Differential Expression of ARG1 and MRC2 in Retinal Müller Glial Cells During Autoimmune Uveitis. Biomolecules 2025; 15:288. [PMID: 40001591 PMCID: PMC11853277 DOI: 10.3390/biom15020288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Retinal Müller glial cells (RMG) play a crucial role in retinal neuroinflammation, including autoimmune uveitis. Increasing evidence supports their function as active modulators of immune responses and potential atypical antigen-presenting cells (APCs). To further investigate this hypothesis, we conducted a differential proteome analysis of primary equine RMG from healthy controls and horses with equine recurrent uveitis (ERU), a spontaneous model of autoimmune uveitis. This analysis identified 310 proteins with differential abundance. Among these, the Major Histocompatibility Complex (MHC) class II and the enzyme Arginase 1 (ARG1) were significantly enriched in RMG from uveitis-affected horses, whereas Mannose Receptor C-type 2 (MRC2) and its interactor Thrombospondin 1 (THBS1) were more abundant in healthy RMG. The detection of MHC class II in equine RMG, consistent with previous studies, validates the robustness of our approach. Furthermore, the identification of ARG1 and MRC2, together with THBS1, provides new insights into the immunomodulatory and antigen-presenting properties of RMG. Immunohistochemical analyses confirmed the proteomic findings and revealed the spatial distribution of ARG1 and MRC2. ARG1 and MRC2 are thus markers for RMG in the neuroinflammatory or physiological milieu and highlight potential differences in the immune function of RMG, particularly in antigen presentation.
Collapse
Affiliation(s)
- Amelie B. Fleischer
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany (T.W.)
| | - Barbara Amann
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany (T.W.)
| | - Christine von Toerne
- Metabolomics and Proteomics Core, Helmholtz Center Munich, German Research Center for Environmental Health, D-80939 Munich, Germany
| | - Roxane L. Degroote
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany (T.W.)
| | - Adrian Schmalen
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany (T.W.)
| | - Tanja Weißer
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany (T.W.)
| | - Stefanie M. Hauck
- Metabolomics and Proteomics Core, Helmholtz Center Munich, German Research Center for Environmental Health, D-80939 Munich, Germany
| | - Cornelia A. Deeg
- Chair of Physiology, Department of Veterinary Sciences, LMU Munich, D-82152 Martinsried, Germany (T.W.)
| |
Collapse
|
14
|
Tuor M, Stappers MHT, Desgardin A, Ruchti F, Sparber F, Orr SJ, Gow NAR, LeibundGut-Landmann S. Card9 and MyD88 differentially regulate Th17 immunity to the commensal yeast Malassezia in the murine skin. Mucosal Immunol 2025; 18:205-219. [PMID: 39579986 DOI: 10.1016/j.mucimm.2024.11.004] [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: 06/19/2024] [Revised: 10/28/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
The fungal community of the skin microbiome is dominated by a single genus, Malassezia. Besides its symbiotic lifestyle at the host interface, this commensal yeast has also been associated with diverse inflammatory skin diseases in humans and pet animals. Stable colonization is maintained by antifungal type 17 immunity. The mechanisms driving Th17 responses to Malassezia remain, however, unclear. Here, we show that the C-type lectin receptors Mincle, Dectin-1, and Dectin-2 recognize conserved patterns in the cell wall of Malassezia and induce dendritic cell activation in vitro, while only Dectin-2 is required for Th17 activation during experimental skin colonization in vivo. In contrast, Toll-like receptor recognition was redundant in this context. Instead, inflammatory IL-1 family cytokines signaling via MyD88 were also implicated in Th17 activation in a T cell-intrinsic manner. Taken together, we characterized the pathways contributing to protective immunity against the most abundant member of the skin mycobiome. This knowledge contributes to the understanding of barrier immunity and its regulation by commensals and is relevant considering how aberrant immune responses are associated with severe skin pathologies.
Collapse
Affiliation(s)
- Meret Tuor
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Mark H T Stappers
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Alice Desgardin
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Fiorella Ruchti
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Florian Sparber
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Selinda J Orr
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Neil A R Gow
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland; Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK.
| |
Collapse
|
15
|
Linnemann L, Antwi-Ekwuruke J, Gnanapragassam V, Bang C, Rühlemann M, Ruland J, Hartmann W, Heepmann L, Dörken S, Yunus SM, Viebrock B, Schlosser A, Lepenies B, Breloer M. The C-type lectin receptor MINCLE interferes with eosinophil function and protective intestinal immunity in Strongyloides ratti-infected mice. Mucosal Immunol 2025; 18:220-231. [PMID: 39581231 DOI: 10.1016/j.mucimm.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/23/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
Strongyloides ratti is a helminth parasite that displays tissue-migrating and intestinal life stages. Myeloid C-type lectin receptors (CLRs) are pattern recognition receptors that recognize pathogen-derived ligands and initiate immune responses. To date, the role of CLRs in S. ratti infection has not been investigated. Here, we show that S. ratti-derived ligands are recognized by the CLR Macrophage inducible Ca2+-dependent lectin receptor (MINCLE). While MINCLE-deficiency did not affect initiation of a protective anti-S. ratti type 2 immunity, MINCLE-deficient mice had a transient advantage in intestinal immunity. Unravelling the underlying mechanism, we show that next to macrophages, dendritic cells and neutrophils, a fraction of eosinophils express MINCLE and expand during S. ratti infection. MINCLE-deficient eosinophils exhibited a more active phenotype and prolonged expansion in vivo and displayed increased capacity to reduce S. ratti motility and produce reactive oxygen species in vitro, compared to wild-type (WT) eosinophils. Depletion of eosinophils in S. ratti-infected mice after the tissue-migration phase elevated intestinal worm burden in MINCLE-deficient mice to the WT level. Thus, our findings establish a central contribution of eosinophils to parasite ejection from the intestine and suggest that S. ratti-triggered signalling via MINCLE interferes with eosinophil mediated ejection of S. ratti from the intestine.
Collapse
Affiliation(s)
- Lara Linnemann
- Section Interface, Bernhard Nocht Institute for Tropical Medicine, 20359, Hamburg, Germany
| | | | - Vinayaga Gnanapragassam
- Institute for Immunology, University of Veterinary Medicine Hannover, 30559, Hanover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559, Hanover, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, University Kiel, 24118, Kiel, Germany
| | - Malte Rühlemann
- Institute of Clinical Molecular Biology, University Kiel, 24118, Kiel, Germany
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, 81675, Munich, Germany; Center for Translational Cancer Research (TranslaTUM), 81675, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Germany
| | - Wiebke Hartmann
- Section Interface, Bernhard Nocht Institute for Tropical Medicine, 20359, Hamburg, Germany
| | - Lennart Heepmann
- Section Interface, Bernhard Nocht Institute for Tropical Medicine, 20359, Hamburg, Germany
| | - Sara Dörken
- Section Interface, Bernhard Nocht Institute for Tropical Medicine, 20359, Hamburg, Germany
| | - Saleh M Yunus
- Section Interface, Bernhard Nocht Institute for Tropical Medicine, 20359, Hamburg, Germany
| | - Birte Viebrock
- Section Interface, Bernhard Nocht Institute for Tropical Medicine, 20359, Hamburg, Germany
| | - Annette Schlosser
- Section Interface, Bernhard Nocht Institute for Tropical Medicine, 20359, Hamburg, Germany
| | - Bernd Lepenies
- Institute for Immunology, University of Veterinary Medicine Hannover, 30559, Hanover, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559, Hanover, Germany
| | - Minka Breloer
- Section Interface, Bernhard Nocht Institute for Tropical Medicine, 20359, Hamburg, Germany; Department for Biology, University Hamburg, 20148, Hamburg, Germany.
| |
Collapse
|
16
|
Ronchese F, Webb GR, Ochiai S, Lamiable O, Brewerton M. How type-2 dendritic cells induce Th2 differentiation: Instruction, repression, or fostering T cell-T cell communication? Allergy 2025; 80:395-407. [PMID: 39324367 PMCID: PMC11804308 DOI: 10.1111/all.16337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 09/03/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
Allergic disease is caused by the activation of allergen-specific CD4+ type-2 T follicular helper cells (Tfh2) and T helper 2 (Th2) effector cells that secrete the cytokines IL-4, IL-5, IL-9, and IL-13 upon allergen encounter, thereby inducing IgE production by B cells and tissue inflammation. While it is accepted that the priming and differentiation of naïve CD4+ T cells into Th2 requires allergen presentation by type 2 dendritic cells (DC2s), the underlying signals remain unidentified. In this review we focus on the interaction between allergen-presenting DC2s and naïve CD4+ T cells in lymph node (LN), and the potential mechanisms by which DC2s might instruct Th2 differentiation. We outline recent advances in characterizing DC2 development and heterogeneity. We review mechanisms of allergen sensing and current proposed mechanisms of Th2 differentiation, with specific consideration of the role of DC2s and how they might contribute to each mechanism. Finally, we assess recent publications reporting a detailed analysis of DC-T cell interactions in LNs and how they support Th2 differentiation. Together, these studies are starting to shape our understanding of this key initial step of the allergic immune response.
Collapse
Affiliation(s)
| | - Greta R. Webb
- Malaghan Institute of Medical ResearchWellingtonNew Zealand
| | - Sotaro Ochiai
- Malaghan Institute of Medical ResearchWellingtonNew Zealand
| | | | - Maia Brewerton
- Malaghan Institute of Medical ResearchWellingtonNew Zealand
- Department of Clinical Immunology and AllergyAuckland City HospitalAucklandNew Zealand
| |
Collapse
|
17
|
Li X, Wang X, Shang Z, Yang S, Tang Y, Xu W. Non-Immune Functions of Innate Immunity Acting on Physiological Processes: Insights from Drosophila. Int J Mol Sci 2025; 26:1087. [PMID: 39940855 PMCID: PMC11817114 DOI: 10.3390/ijms26031087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
As the first line of host immune defense, innate immunity plays a key role in warding off foreign pathogens and damage. Drosophila melanogaster, as a classical model animal for more than 100 years, is an important research model for studying innate immunity. In recent years, scientists have made remarkable progress in the recognition mechanisms of innate immunity, the mechanisms of effector molecules, and the modes of their response at the cellular and tissue levels. However, the interaction between innate immunity and other physiological functions remains relatively novel and has yet to be systematically explored. Here, we first briefly discuss the link between the innate immunity system and physiological regulation, from several representative perspectives such as sleep, insulin, and brain function. Then, using Drosophila as a model, we provide an overview of the physiological system and specifically summarize the research on the regulation of physiology by innate immunity, covering sleep, lipid metabolism, development, neurodegenerative diseases, memory, feeding, lifespan, movement, and antioxidation. This review provides valuable perspectives into how innate immunity influences other physiological processes, providing a deeper understanding of the complex roles underlying innate immunity.
Collapse
Affiliation(s)
| | | | | | | | | | - Wenhua Xu
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao 266071, China
| |
Collapse
|
18
|
Carroll KA, Sawden M, Sharma S. DAMPs, PAMPs, NLRs, RIGs, CLRs and TLRs - Understanding the Alphabet Soup in the Context of Bone Biology. Curr Osteoporos Rep 2025; 23:6. [PMID: 39808398 DOI: 10.1007/s11914-024-00900-3] [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: 12/09/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the current understanding of cell-autonomous innate immune pathways that contribute to bone homeostasis and disease. RECENT FINDINGS Germ-line encoded pattern recognition receptors (PRRs) are the first line of defense against danger and infections. In the bone microenvironment, PRRs and downstream signaling pathways, that mount immune defense, interface intimately with the core cellular processes in bone cells to alter bone formation and resorption. The role of PRR engagement on bone remodeling has been best described as a result of activated macrophages secreting effector molecules that reshape the characteristics of bone-resident cells. However, it is being increasingly recognized that local bone resident-cells like osteoclasts and osteoblasts possess an arsenal of PRRs. The engagement of these PRRs by stimuli in the bone niche can drive cell-autonomous (aka cell-intrinsic) responses that, in turn, impact bone-remodeling dramatically, irrespective of immune cell effectors. Indeed, this vital role for cell-autonomous innate immune responses is evident in how reduced PRR activity within osteoclast progenitors correlates with their reduced differentiation and abnormal bone remodeling. Further, cell-intrinsic PRR activity has now been shown to influence the behavior of osteoblasts, osteocytes and other local immune/non-immune cell populations. However, distinct PRR families have varying impact on bone homeostasis and inflammation, emphasizing the importance of investigating these different nodes of innate immune signaling in bone cells to better identify how they synergistically and/or antagonistically regulate bone remodeling in the course of an immune response. Innate immune sensing within bone resident cells is a critical determinant for bone remodeling in health and disease.
Collapse
Affiliation(s)
- K A Carroll
- Department of Immunology, Tufts University, Boston, MA, 02111, USA
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA, 02111, USA
| | - M Sawden
- Department of Immunology, Tufts University, Boston, MA, 02111, USA
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA, 02111, USA
| | - S Sharma
- Department of Immunology, Tufts University, Boston, MA, 02111, USA.
| |
Collapse
|
19
|
Kroemer G, Montégut L, Kepp O, Zitvogel L. The danger theory of immunity revisited. Nat Rev Immunol 2024; 24:912-928. [PMID: 39511426 DOI: 10.1038/s41577-024-01102-9] [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: 10/09/2024] [Indexed: 11/15/2024]
Abstract
The danger theory of immunity, introduced by Polly Matzinger in 1994, posits that tissue stress, damage or infection has a decisive role in determining immune responses. Since then, a growing body of evidence has supported the idea that the capacity to elicit cognate immune responses (immunogenicity) relies on the combination of antigenicity (the ability to be recognized by T cell receptors or antibodies) and adjuvanticity (additional signals arising owing to tissue damage). Here, we discuss the molecular foundations of the danger theory while focusing on immunologically relevant damage-associated molecular patterns, microorganism-associated molecular patterns, and neuroendocrine stress-associated immunomodulatory molecules, as well as on their receptors. We critically evaluate patient-relevant evidence, examining how cancer cells and pathogenic viruses suppress damage-associated molecular patterns to evade immune recognition, how intestinal dysbiosis can reduce immunostimulatory microorganism-associated molecular patterns and compromise immune responses, and which hereditary immune defects support the validity of the danger theory. Furthermore, we incorporate the danger hypothesis into a close-to-fail-safe hierarchy of immunological tolerance mechanisms that also involve the clonal deletion and inactivation of immune cells.
Collapse
Affiliation(s)
- Guido Kroemer
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Léa Montégut
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, INSERM U1138, Équipe Labellisée - Ligue Nationale contre le Cancer, Université Paris Cité, Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France.
- INSERM UMR 1015, ClinicObiome, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France.
- Université Paris-Saclay, Ile-de-France, Paris, France.
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS), Villejuif, France.
| |
Collapse
|
20
|
Chen F, Tang H, Cai X, Lin J, Kang R, Tang D, Liu J. DAMPs in immunosenescence and cancer. Semin Cancer Biol 2024; 106-107:123-142. [PMID: 39349230 DOI: 10.1016/j.semcancer.2024.09.005] [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: 08/30/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/02/2024]
Abstract
Damage-associated molecular patterns (DAMPs) are endogenous molecules released by cells in response to injury or stress, recognized by host pattern recognition receptors that assess the immunological significance of cellular damage. The interaction between DAMPs and innate immune receptors triggers sterile inflammation, which serves a dual purpose: promoting tissue repair and contributing to pathological conditions, including age-related diseases. Chronic inflammation mediated by DAMPs accelerates immunosenescence and influences both tumor progression and anti-tumor immunity, underscoring the critical role of DAMPs in the nexus between aging and cancer. This review explores the characteristics of immunosenescence and its impact on age-related cancers, investigates the various types of DAMPs, their release mechanisms during cell death, and the immune activation pathways they initiate. Additionally, we examine the therapeutic potential of targeting DAMPs in age-related diseases. A detailed understanding of DAMP-induced signal transduction could provide critical insights into immune regulation and support the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Fangquan Chen
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Hu Tang
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Xiutao Cai
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Junhao Lin
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China.
| |
Collapse
|
21
|
Nardini E, Rodriguez E, van Kooyk Y. The tissue glycome as regulator of immune activation and tolerance mediated by C-type lectins and Siglecs. Semin Immunol 2024; 76:101913. [PMID: 39602867 DOI: 10.1016/j.smim.2024.101913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
The immune system is a complex network of highly specialized microenvironments, denominated niches, which arise from dynamic interactions between immune and parenchymal cells as well as acellular components such as structural elements and local molecular signals. A critical, yet underexplored, layer shaping these niches is the glycome, the complete repertoire of glycans and glycoconjugates produced by cells. The glycome is prevalent in the outer membrane of cells and their secreted components, and can be sensed by glycan binding receptors on immune cells. These receptors detect changes in glycosylation and consequently modulate immune cell activity, trafficking, and signalling, altering homeostasis. Tissues like the brain and the placenta are prone to accommodate tolerance, while the gut and the thymus are sensitive to inflammation. We provide here an overview of current literature that shows the impact of altered glycosylation of tissues on host immune cells and how interference in this process may lead to new diagnostics and immune therapeutics, aiming to restore the immune balance in autoimmunity and cancer.
Collapse
Affiliation(s)
- Eleonora Nardini
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, Amsterdam 1117, The Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands; Amsterdam institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Ernesto Rodriguez
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, Amsterdam 1117, The Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands; Amsterdam institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, Amsterdam 1117, The Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands; Amsterdam institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands.
| |
Collapse
|
22
|
Heimburg-Molinaro J, Mehta AY, Tilton CA, Cummings RD. Insights Into Glycobiology and the Protein-Glycan Interactome Using Glycan Microarray Technologies. Mol Cell Proteomics 2024; 23:100844. [PMID: 39307422 PMCID: PMC11585810 DOI: 10.1016/j.mcpro.2024.100844] [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: 06/13/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 11/11/2024] Open
Abstract
Glycans linked to proteins and lipids and also occurring in free forms have many functions, and these are partly elicited through specific interactions with glycan-binding proteins (GBPs). These include lectins, adhesins, toxins, hemagglutinins, growth factors, and enzymes, but antibodies can also bind glycans. While humans and other animals generate a vast repertoire of GBPs and different glycans in their glycomes, other organisms, including phage, microbes, protozoans, fungi, and plants also express glycans and GBPs, and these can also interact with their host glycans. This can be termed the protein-glycan interactome, and in nature is likely to be vast, but is so far very poorly described. Understanding the breadth of the protein-glycan interactome is also a key to unlocking our understanding of infectious diseases involving glycans, and immunology associated with antibodies binding to glycans. A key technological advance in this area has been the development of glycan microarrays. This is a display technology in which minute quantities of glycans are attached to the surfaces of slides or beads. This allows the arrayed glycans to be interrogated by GBPs and antibodies in a relatively high throughput approach, in which a protein may bind to one or more distinct glycans. Such binding can lead to novel insights and hypotheses regarding both the function of the GBP, the specificity of an antibody and the function of the glycan within the context of the protein-glycan interactome. This article focuses on the types of glycan microarray technologies currently available to study animal glycobiology and examples of breakthroughs aided by these technologies.
Collapse
Affiliation(s)
- Jamie Heimburg-Molinaro
- Department of Surgery Beth Israel Deaconess Medical Center, National Center for Functional Glycomics (NCFG), Harvard Medical School, Boston, Massachusetts, USA
| | - Akul Y Mehta
- Department of Surgery Beth Israel Deaconess Medical Center, National Center for Functional Glycomics (NCFG), Harvard Medical School, Boston, Massachusetts, USA
| | - Catherine A Tilton
- Department of Surgery Beth Israel Deaconess Medical Center, National Center for Functional Glycomics (NCFG), Harvard Medical School, Boston, Massachusetts, USA
| | - Richard D Cummings
- Department of Surgery Beth Israel Deaconess Medical Center, National Center for Functional Glycomics (NCFG), Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
23
|
Stegmann F, Lepenies B. Myeloid C-type lectin receptors in host-pathogen interactions and glycan-based targeting. Curr Opin Chem Biol 2024; 82:102521. [PMID: 39214069 DOI: 10.1016/j.cbpa.2024.102521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/30/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Lectin-glycan interactions play a crucial role in the immune system. An important class of lectins in the innate immune system is myeloid C-type lectin receptors (CLRs). Myeloid CLRs act as pattern recognition receptors and are predominantly expressed by myeloid cells, such as macrophages, dendritic cells, and neutrophils. In innate immunity, CLRs contribute to self/non-self discrimination. While the recognition of pathogen-associated molecular patterns (PAMPs) by CLRs may contribute to a protective immune response, CLR engagement can also be exploited by pathogens for immune evasion. Since various CLRs act as endocytic receptors and trigger distinct signaling pathways in myeloid cells, CLR targeting has proven useful for drug/antigen delivery into antigen-presenting cells and the modulation of immune responses. This review covers recent discoveries of pathogen/CLR interactions and novel approaches for CLR targeting within the period of the past two years.
Collapse
Affiliation(s)
- Felix Stegmann
- Institute for Immunology, University of Veterinary Medicine Hannover, 30559 Hannover, Lower Saxony, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Lower Saxony, Germany
| | - Bernd Lepenies
- Institute for Immunology, University of Veterinary Medicine Hannover, 30559 Hannover, Lower Saxony, Germany; Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Lower Saxony, Germany.
| |
Collapse
|
24
|
Iliev ID, Brown GD, Bacher P, Gaffen SL, Heitman J, Klein BS, Lionakis MS. Focus on fungi. Cell 2024; 187:5121-5127. [PMID: 39303681 PMCID: PMC11722117 DOI: 10.1016/j.cell.2024.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 09/22/2024]
Abstract
Fungi play critical roles in the homeostasis of ecosystems globally and have emerged as significant causes of an expanding repertoire of devastating diseases in plants, animals, and humans. In this Commentary, we highlight the importance of fungal pathogens and argue for concerted research efforts to enhance understanding of fungal virulence, antifungal immunity, novel drug targets, antifungal resistance, and the mycobiota to improve human health.
Collapse
Affiliation(s)
- Iliyan D Iliev
- Joan and Sanford I. Weill Department of Medicine, The Jill Roberts Institute for Research in Inflammatory Bowel Disease, and Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| | - Gordon D Brown
- MRC Centre for Medical Mycology at the University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Petra Bacher
- Institute of Immunology and Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Sarah L Gaffen
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Bruce S Klein
- Departments of Pediatrics, Medicine, and Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
25
|
Rojekar S, Gholap AD, Togre N, Bhoj P, Haeck C, Hatvate N, Singh N, Vitore J, Dhoble S, Kashid S, Patravale V. Current status of mannose receptor-targeted drug delivery for improved anti-HIV therapy. J Control Release 2024; 372:494-521. [PMID: 38849091 DOI: 10.1016/j.jconrel.2024.06.002] [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: 03/12/2024] [Revised: 05/22/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024]
Abstract
In the pursuit of achieving better therapeutic outcomes in the treatment of HIV, innovative drug delivery strategies have been extensively explored. Mannose receptors, which are primarily found on macrophages and dendritic cells, offer promising targets for drug delivery due to their involvement in HIV pathogenesis. This review article comprehensively evaluates recent drug delivery system advancements targeting the mannose receptor. We have systematically described recent developments in creating and utilizing drug delivery platforms, including nanoparticles, liposomes, micelles, noisomes, dendrimers, and other nanocarrier systems targeted at the mannose receptor. These strategies aim to enhance drug delivery specificity, bioavailability, and therapeutic efficacy while decreasing off-target effects and systemic toxicity. Furthermore, the article delves into how mannose receptors and HIV interact, highlighting the potential for exploiting this interaction to enhance drug delivery to infected cells. The review covers essential topics, such as the rational design of nanocarriers for mannose receptor recognition, the impact of physicochemical properties on drug delivery performance, and how targeted delivery affects the pharmacokinetics and pharmacodynamics of anti-HIV agents. The challenges of these novel strategies, including immunogenicity, stability, and scalability, and future research directions in this rapidly growing area are discussed. The knowledge synthesis presented in this review underscores the potential of mannose receptor-based targeted drug delivery as a promising avenue for advancing HIV treatment. By leveraging the unique properties of mannose receptors, researchers can design drug delivery systems that cater to individual needs, overcome existing limitations, and create more effective and patient-friendly treatments in the ongoing fight against HIV/AIDS.
Collapse
Affiliation(s)
- Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Namdev Togre
- Department of Pathology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Priyanka Bhoj
- Department of Pathology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Clement Haeck
- Population Council, , Center for Biomedical Research, 1230 York Avenue, New York, NY 10065, USA
| | - Navnath Hatvate
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India
| | - Nidhi Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata 700054, India
| | - Jyotsna Vitore
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gujarat 382355, India
| | - Sagar Dhoble
- Department of Pharmacology and Toxicology, R. K. Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Snehal Kashid
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gujarat 382355, India
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India.
| |
Collapse
|
26
|
Monti M, Ferrari G, Gazzurelli L, Bugatti M, Facchetti F, Vermi W. Plasmacytoid dendritic cells at the forefront of anti-cancer immunity: rewiring strategies for tumor microenvironment remodeling. J Exp Clin Cancer Res 2024; 43:196. [PMID: 39020402 PMCID: PMC11253500 DOI: 10.1186/s13046-024-03121-9] [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: 05/10/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are multifaceted immune cells executing various innate immunological functions. Their first line of defence consists in type I interferons (I-IFN) production upon nucleic acids sensing through endosomal Toll-like receptor (TLR) 7- and 9-dependent signalling pathways. Type I IFNs are a class of proinflammatory cytokines that have context-dependent functions on cancer immunosurveillance and immunoediting. In the last few years, different studies have reported that pDCs are also able to sense cytosolic DNA through cGAS-STING (stimulator of interferon genes) pathway eliciting a potent I-IFN production independently of TLR7/9. Human pDCs are also endowed with direct effector functions via the upregulation of TRAIL and production of granzyme B, the latter modulated by cytokines abundant in cancer tissues. pDCs have been detected in a wide variety of human malignant neoplasms, including virus-associated cancers, recruited by chemotactic stimuli. Although the role of pDCs in cancer immune surveillance is still uncompletely understood, their spontaneous activation has been rarely documented; moreover, their presence in the tumor microenvironment (TME) has been associated with a tolerogenic phenotype induced by immunosuppressive cytokines or oncometabolites. Currently tested treatment options can lead to pDCs activation and disruption of the immunosuppressive TME, providing a relevant clinical benefit. On the contrary, the antibody-drug conjugates targeting BDCA-2 on immunosuppressive tumor-associated pDCs (TA-pDCs) could be proposed as novel immunomodulatory therapies to achieve disease control in patients with advance stage hematologic malignancies or solid tumors. This Review integrate recent evidence on the biology of pDCs and their pharmacological modulation, suggesting their relevant role at the forefront of cancer immunity.
Collapse
Affiliation(s)
- Matilde Monti
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, P.Le Spedali Civili 1, 25123, Brescia, Italy
| | - Giorgia Ferrari
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, P.Le Spedali Civili 1, 25123, Brescia, Italy
| | - Luisa Gazzurelli
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, P.Le Spedali Civili 1, 25123, Brescia, Italy
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, P.Le Spedali Civili 1, 25123, Brescia, Italy
| | - Fabio Facchetti
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, P.Le Spedali Civili 1, 25123, Brescia, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, P.Le Spedali Civili 1, 25123, Brescia, Italy.
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
27
|
Tuor M, Stappers MH, Ruchti F, Desgardin A, Sparber F, Orr SJ, Gow NA, LeibundGut-Landmann S. Card9 and MyD88 differentially regulate Th17 immunity to the commensal yeast Malassezia in the murine skin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603211. [PMID: 39071334 PMCID: PMC11275786 DOI: 10.1101/2024.07.12.603211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The fungal community of the skin microbiome is dominated by a single genus, Malassezia. Besides its symbiotic lifestyle at the host interface, this commensal yeast has also been associated with diverse inflammatory skin diseases in humans and pet animals. Stable colonization is maintained by antifungal type 17 immunity. The mechanisms driving Th17 responses to Malassezia remain, however, unclear. Here, we show that the C-type lectin receptors Mincle, Dectin-1, and Dectin-2 recognize conserved patterns in the cell wall of Malassezia and induce dendritic cell activation in vitro, while only Dectin-2 is required for Th17 activation during experimental skin colonization in vivo. In contrast, Toll-like receptor recognition was redundant in this context. Instead, inflammatory IL-1 family cytokines signaling via MyD88 were also implicated in Th17 activation in a T cell-intrinsic manner. Taken together, we characterized the pathways contributing to protective immunity against the most abundant member of the skin mycobiome. This knowledge contributes to the understanding of barrier immunity and its regulation by commensals and is relevant considering how aberrant immune responses are associated with severe skin pathologies.
Collapse
Affiliation(s)
- Meret Tuor
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, Switzerland
| | - Mark H.T. Stappers
- Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Fiorella Ruchti
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, Switzerland
| | - Alice Desgardin
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, Switzerland
| | - Florian Sparber
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, Switzerland
| | - Selinda J. Orr
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen’s University Belfast, Belfast BT9 7BL, UK
| | - Neil A.R. Gow
- Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, Switzerland
- Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| |
Collapse
|
28
|
Zheng Y, Feng J, Ling M, Yu Y, Tao Y, Wang X. A comprehensive review on targeting cluster of differentiation: An attractive strategy for inhibiting viruses through host proteins. Int J Biol Macromol 2024; 269:132200. [PMID: 38723834 DOI: 10.1016/j.ijbiomac.2024.132200] [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: 03/04/2024] [Revised: 04/20/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Viral infections continue to pose a significant global public health threat. Targeting host proteins, such as cluster of differentiation (CD) macromolecules, may offer a promising alternative approach to developing antiviral treatments. CDs are cell-surface biological macromolecules mainly expressed on leukocytes that viruses can use to enter cells, thereby evading immune detection and promoting their replication. The manipulation of CDs by viruses may represent an effective and clever means of survival through the prolonged co-evolution of hosts and viruses. Targeting of CDs is anticipated to hinder the invasion of related viruses, modulate the body's immune system, and diminish the incidence of subsequent inflammation. They have become crucial for biomedical diagnosis, and some have been used as valuable tools for resisting viral infections. However, a summary of the structures and functions of CDs involved in viral infection is currently lacking. The development of drugs targeting these biological macromolecules is restricted both in terms of their availability and the number of compounds currently identified. This review provides a comprehensive analysis of the critical role of CD proteins in virus invasion and a list of relevant targeted antiviral agents, which will serve as a valuable reference for future research in this field.
Collapse
Affiliation(s)
- Youle Zheng
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jin Feng
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Min Ling
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yixin Yu
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yanfei Tao
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| |
Collapse
|
29
|
Carroll SL, Pasare C, Barton GM. Control of adaptive immunity by pattern recognition receptors. Immunity 2024; 57:632-648. [PMID: 38599163 PMCID: PMC11037560 DOI: 10.1016/j.immuni.2024.03.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
One of the most significant conceptual advances in immunology in recent history is the recognition that signals from the innate immune system are required for induction of adaptive immune responses. Two breakthroughs were critical in establishing this paradigm: the identification of dendritic cells (DCs) as the cellular link between innate and adaptive immunity and the discovery of pattern recognition receptors (PRRs) as a molecular link that controls innate immune activation as well as DC function. Here, we recount the key events leading to these discoveries and discuss our current understanding of how PRRs shape adaptive immune responses, both indirectly through control of DC function and directly through control of lymphocyte function. In this context, we provide a conceptual framework for how variation in the signals generated by PRR activation, in DCs or other cell types, can influence T cell differentiation and shape the ensuing adaptive immune response.
Collapse
Affiliation(s)
- Shaina L Carroll
- Division of Immunology & Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Chandrashekhar Pasare
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH USA
| | - Gregory M Barton
- Division of Immunology & Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720 USA.
| |
Collapse
|