1
|
Bouma RG, Wang AZ, den Haan JMM. Exploring CD169 + Macrophages as Key Targets for Vaccination and Therapeutic Interventions. Vaccines (Basel) 2025; 13:330. [PMID: 40266235 PMCID: PMC11946325 DOI: 10.3390/vaccines13030330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
CD169 is a sialic acid-binding immunoglobulin-like lectin (Siglec-1, sialoadhesin) that is expressed by subsets of tissue-resident macrophages and circulating monocytes. This receptor interacts with α2,3-linked Neu5Ac on glycoproteins as well as glycolipids present on the surface of immune cells and pathogens. CD169-expressing macrophages exert tissue-specific homeostatic functions, but they also have opposing effects on the immune response. CD169+ macrophages act as a pathogen filter, protect against infectious diseases, and enhance adaptive immunity, but at the same time pathogens also exploit them to enable further dissemination. In cancer, CD169+ macrophages in tumor-draining lymph nodes are correlated with better clinical outcomes. In inflammatory diseases, CD169 expression is upregulated on monocytes and on monocyte-derived macrophages and this correlates with the disease state. Given their role in promoting adaptive immunity, CD169+ macrophages are currently investigated as targets for vaccination strategies against cancer. In this review, we describe the studies investigating the importance of CD169 and CD169+ macrophages in several disease settings and the vaccination strategies currently under investigation.
Collapse
Affiliation(s)
- Rianne G. Bouma
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Cancer Immunology, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
| | - Aru Z. Wang
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Cancer Immunology, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
| | - Joke M. M. den Haan
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Cancer Immunology, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
2
|
Pérez-Chacón G, Santamaría PG, Redondo-Pedraza J, González-Suárez E. RANK/RANKL Signaling Pathway in Breast Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:309-345. [PMID: 39821032 DOI: 10.1007/978-3-031-70875-6_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
RANK pathway has attracted increasing interest as a promising target in breast cancer, given the availability of denosumab, an anti-RANKL drug. RANK signaling mediates progesterone-driven regulation of mammary gland development and favors breast cancer initiation by controlling mammary cell proliferation and stem cell fate. RANK activation promotes luminal mammary epithelial cell senescence, acting as an initial barrier to tumorigenesis but ultimately facilitating tumor progression and metastasis. Comprehensive analyses have demonstrated that RANK protein expression is an independent biomarker of poor prognosis in postmenopausal and estrogen receptor-negative breast cancer patients. RANK pathway also has multiple roles in immunity and inflammation, regulating innate and adaptive responses. In the tumor microenvironment, RANK and RANKL are expressed by different immune cell populations and contribute to the regulation of tumor immune surveillance, mainly driving immunosuppressive effects.Herein, we discuss the preventive and therapeutic potential of targeting RANK signaling in breast cancer given its tumor cell intrinsic and extrinsic effects. RANKL inhibition has been shown to induce mammary tumor cell differentiation and an antitumor immune response. Moreover, loss of RANK signaling increases sensitivity of breast cancer cells to chemotherapy, targeted therapies such as HER2 and CDK4/6 inhibitors, and immunotherapy. Finally, we describe clinical trials of denosumab for breast cancer prevention, such as those ongoing in women with high risk of developing breast cancer, large phase III clinical trials where the impact of adjuvant denosumab on disease-free survival has been assessed, and window trials to evaluate the immunomodulatory effects of denosumab in breast cancer and other solid tumors.
Collapse
Affiliation(s)
- Gema Pérez-Chacón
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | | | - Eva González-Suárez
- Molecular Oncology, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
- Oncobell, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
3
|
Qi W, Liu C, Shi L, Li H, Hou X, Du H, Chen L, Gao X, Cao X, Guo N, Dong Y, Li C, Yuan F, Teng Z, Hu H, Zhu F, Zhou X, Guo L, Zhao M, Xia M. CD169+ Macrophages Mediate the Immune Response of Allergic Rhinitis Through the Keap1/Nrf2/HO-1 Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309331. [PMID: 39435598 PMCID: PMC11615775 DOI: 10.1002/advs.202309331] [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: 12/01/2023] [Revised: 08/21/2024] [Indexed: 10/23/2024]
Abstract
CD169+ macrophages are a newly defined macrophage subpopulation that can recognize and bind with other cells through related ligands, playing an essential role in antigen presentation and immune tolerance. However, its role in Allergic Rhinitis (AR) is still unclear. To investigate the characteristics of CD169+ macrophages in AR, this work first detects their expression patterns in the nasal mucosa of clinical patients. These results show a significant increase in CD169+ macrophages in the nasal mucosa of patients with AR. Subsequently, this work establishes an animal AR model using CD169 transgenic mice and compared the advantages of the two models. Moreover, this work also demonstrates the effects of CD169 knockout on eosinophils, Th cells, Treg cells, and the migration of dendritic cells (DCs). In addition, this metabolomic data shows that CD169+ macrophages can upregulate alanine production and increase reactive oxygen species (ROS) levels. This process may be mediated through the Keap1/Nrf2/HO-1 signaling pathway. In addition, this work also finds that SLC38A2 plays an essential role in the process of CD169+ macrophages promoting alanine uptake by DCs. This study confirms that CD169+ macrophages can upregulate their internal alanine production and increase ROS levels through the Keap1/Nrf2/HO-1 axis, playing an irreplaceable role in AR.
Collapse
Affiliation(s)
- Wenwen Qi
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Chengcheng Liu
- Department of Central LaboratoryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Lei Shi
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Hui Li
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Xiaozhi Hou
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Hongjie Du
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Luqiu Chen
- Department of Pediatric SurgeryQilu HospitalCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Xiaochen Gao
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Xue Cao
- Department of OtolaryngologyShandong Provincial HospitalShandong UniversityJinanChina
| | - Na Guo
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Yuhan Dong
- Department of OtolaryngologyShandong Provincial HospitalShandong UniversityJinanChina
| | - Chengzhilin Li
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Fanyu Yuan
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Zhenxiao Teng
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Houyang Hu
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Fangyuan Zhu
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Xuanchen Zhou
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Lulu Guo
- Advanced Medical Research InstituteCheeloo College of MedicineNHC Key Laboratory of OtorhinolaryngologyShandong UniversityJinanChina
| | - Miaoqing Zhao
- Department of PathologyShandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Ming Xia
- Department of OtolaryngologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
- Department of OtolaryngologyShandong Provincial HospitalShandong UniversityJinanChina
- NHC Key Laboratory of OtorhinolaryngologyJinanChina
| |
Collapse
|
4
|
Croft M, Salek-Ardakani S, Ware CF. Targeting the TNF and TNFR superfamilies in autoimmune disease and cancer. Nat Rev Drug Discov 2024; 23:939-961. [PMID: 39448880 DOI: 10.1038/s41573-024-01053-9] [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: 09/09/2024] [Indexed: 10/26/2024]
Abstract
The first anti-tumour necrosis factor (TNF) monoclonal antibody, infliximab (Remicade), celebrated its 25th anniversary of FDA approval in 2023. Inhibitors of TNF have since proved clinically efficacious at reducing inflammation associated with several autoimmune diseases, including rheumatoid arthritis, psoriasis and Crohn's disease. The success of TNF inhibitors raised unrealistic expectations for targeting other members of the TNF superfamily (TNFSF) of ligands and their receptors, with difficulties in part related to their more limited, variable expression and potential redundancy. However, there has been a resurgence of interest and investment, with many of these cytokines or their cognate receptors now under clinical investigation as targets for modulation of autoimmune and inflammatory diseases, as well as cancer. This Review assesses TNFSF-targeted biologics currently in clinical development for immune system-related diseases, highlighting ongoing challenges and future directions.
Collapse
Affiliation(s)
- Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA.
| | | | - Carl F Ware
- Laboratory of Molecular Immunology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA.
| |
Collapse
|
5
|
Wang L, Fan J, Wu S, Cheng S, Zhao J, Fan F, Gao C, Qiao R, Sheng Q, Hu Y, Zhang Y, Liu P, Jiao Z, Wei T, Lei J, Chen Y, Qin H. LTBR acts as a novel immune checkpoint of tumor-associated macrophages for cancer immunotherapy. IMETA 2024; 3:e233. [PMID: 39429877 PMCID: PMC11487550 DOI: 10.1002/imt2.233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 10/22/2024]
Abstract
Tumor-associated macrophages (TAMs) greatly contribute to immune checkpoint inhibitor (ICI) resistance of cancer. However, its underlying mechanisms and whether TAMs can be promising targets to overcome ICI resistance remain to be unveiled. Through integrative analysis of immune multiomics data and single-cell RNA-seq data (iMOS) in lung adenocarcinoma (LUAD), lymphotoxin β receptor (LTBR) is identified as a potential immune checkpoint of TAMs, whose high expression, duplication, and low methylation are correlated with unfavorable prognosis. Immunofluorescence staining shows that the infiltration of LTBR+ TAMs is associated with LUAD stages, immunotherapy failure, and poor prognosis. Mechanistically, LTΒR maintains immunosuppressive activity and M2 phenotype of TAMs by noncanonical nuclear factor kappa B and Wnt/β-catenin signaling pathways. Macrophage-specific knockout of LTBR hinders tumor growth and prolongs survival time via blocking TAM immunosuppressive activity and M2 phenotype. Moreover, TAM-targeted delivery of LTΒR small interfering RNA improves the therapeutic effect of ICI via reversing TAM-mediated immunosuppression, such as boosting cytotoxic CD8+ T cells and inhibiting granulocytic myeloid-derived suppressor cells infiltration. Taken together, we bring forth an immune checkpoint discovery pipeline iMOS, identify LTBR as a novel immune checkpoint of TAMs, and propose a new immunotherapy strategy by targeting LTBR+ TAMs.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Jieyi Fan
- Department of Aerospace MedicineFourth Military Medical UniversityXi'anChina
| | - Sifan Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Shilin Cheng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Junlong Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Fan Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Chunchen Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Rong Qiao
- Department of Clinical Oncology, Xijing HospitalFourth Military Medical UniversityXi'anChina
| | - Qiqi Sheng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Yiyang Hu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Yong Zhang
- Department of Pulmonary Medicine, Xijing HospitalFourth Military Medical UniversityXi'anChina
| | - Pengjun Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Zhe Jiao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Tiaoxia Wei
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| | - Jie Lei
- Department of Thoracic Surgery, Tangdu HospitalFourth Military Medical UniversityXi'anChina
| | - Yan Chen
- Department of Clinical Oncology, Xijing HospitalFourth Military Medical UniversityXi'anChina
| | - Hongyan Qin
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Medical Genetics and Developmental BiologyFourth Military Medical UniversityXi'anChina
| |
Collapse
|
6
|
Stader F, Liu C, Derbalah A, Momiji H, Pan X, Gardner I, Jamei M, Sepp A. A Physiologically Based Pharmacokinetic Model Relates the Subcutaneous Bioavailability of Monoclonal Antibodies to the Saturation of FcRn-Mediated Recycling in Injection-Site-Draining Lymph Nodes. Antibodies (Basel) 2024; 13:70. [PMID: 39189241 PMCID: PMC11348173 DOI: 10.3390/antib13030070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024] Open
Abstract
The bioavailability of a monoclonal antibody (mAb) or another therapeutic protein after subcutaneous (SC) dosing is challenging to predict from first principles, even if the impact of injection site physiology and drug properties on mAb bioavailability is generally understood. We used a physiologically based pharmacokinetic model to predict pre-systemic clearance after SC administration mechanistically by incorporating the FcRn salvage pathway in antigen-presenting cells (APCs) in peripheral lymph nodes, draining the injection site. Clinically observed data of the removal rate of IgG from the arm as well as its plasma concentration after SC dosing were mostly predicted within the 95% confidence interval. The bioavailability of IgG was predicted to be 70%, which mechanistically relates to macropinocytosis in the draining lymph nodes and transient local dose-dependent partial saturation of the FcRn receptor in the APCs, resulting in higher catabolism and consequently less drug reaching the systemic circulation. The predicted free FcRn concentration was reduced to 40-45%, reaching the minimum 1-2 days after the SC administration of IgG, and returned to baseline after 8-12 days, depending on the site of injection. The model predicted the uptake into APCs, the binding affinity to FcRn, and the dose to be important factors impacting the bioavailability of a mAb.
Collapse
Affiliation(s)
- Felix Stader
- Simcyp Division, Certara UK Ltd., Level 2 Acero, 1 Concourse Way, Sheffield S1 2BJ, UK (X.P.); (I.G.); (A.S.)
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Iliopoulou M, Bajur AT, McArthur HCW, Gabai M, Coyle C, Ajao F, Köchl R, Cope AP, Spillane KM. Extracellular matrix rigidity modulates physical properties of subcapsular sinus macrophage-B cell immune synapses. Biophys J 2024; 123:2282-2300. [PMID: 37840242 PMCID: PMC11331050 DOI: 10.1016/j.bpj.2023.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/17/2023] [Accepted: 10/11/2023] [Indexed: 10/17/2023] Open
Abstract
Subcapsular sinus macrophages (SSMs) play a key role in immune defense by forming immunological barriers that control the transport of antigens from lymph into lymph node follicles. SSMs participate in antibody responses by presenting antigens directly to naive B cells and by supplying antigens to follicular dendritic cells to propagate germinal center reactions. Despite the prominent roles that SSMs play during immune responses, little is known about their cell biology because they are technically challenging to isolate and study in vitro. Here, we used multicolor fluorescence microscopy to identify lymph node-derived SSMs in culture. We focused on the role of SSMs as antigen-presenting cells, and found that their actin cytoskeleton regulates the spatial organization and mobility of multivalent antigens (immune complexes [ICs]) displayed on the cell surface. Moreover, we determined that SSMs are mechanosensitive cells that respond to changes in extracellular matrix rigidity by altering the architecture of the actin cytoskeleton, leading to changes in cell morphology, membrane topography, and IC mobility. Changes to extracellular matrix rigidity also modulate actin remodeling by both SSMs and B cells when they form an immune synapse. This alters synapse duration but not IC internalization nor NF-κB activation in the B cell. Taken together, our data reveal that the mechanical microenvironment may influence B cell responses by modulating physical characteristics of antigen presentation by SSMs.
Collapse
Affiliation(s)
- Maro Iliopoulou
- Department of Physics, King's College London, London, United Kingdom
| | - Anna T Bajur
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell & Molecular Biophysics, King's College London, London, United Kingdom
| | | | - Michael Gabai
- Department of Physics, King's College London, London, United Kingdom
| | - Carl Coyle
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, United Kingdom
| | - Favour Ajao
- Department of Physics, King's College London, London, United Kingdom
| | - Robert Köchl
- Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom
| | - Andrew P Cope
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, United Kingdom
| | - Katelyn M Spillane
- Department of Physics, King's College London, London, United Kingdom; Randall Centre for Cell & Molecular Biophysics, King's College London, London, United Kingdom.
| |
Collapse
|
8
|
Tang Y, Liu B, Zhang Y, Liu Y, Huang Y, Fan W. Interactions between nanoparticles and lymphatic systems: Mechanisms and applications in drug delivery. Adv Drug Deliv Rev 2024; 209:115304. [PMID: 38599495 DOI: 10.1016/j.addr.2024.115304] [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: 12/12/2023] [Revised: 03/08/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
The lymphatic system has garnered significant attention in drug delivery research due to the advantages it offers, such as enhancing systemic exposure and enabling lymph node targeting for nanomedicines via the lymphatic delivery route. The journey of drug carriers involves transport from the administration site to the lymphatic vessels, traversing the lymph before entering the bloodstream or targeting specific lymph nodes. However, the anatomical and physiological barriers of the lymphatic system play a pivotal role in influencing the behavior and efficiency of carriers. To expedite research and subsequent clinical translation, this review begins by introducing the composition and classification of the lymphatic system. Subsequently, we explore the routes and mechanisms through which nanoparticles enter lymphatic vessels and lymph nodes. The review further delves into the interactions between nanomedicine and body fluids at the administration site or within lymphatic vessels. Finally, we provide a comprehensive overview of recent advancements in lymphatic delivery systems, addressing the challenges and opportunities inherent in current systems for delivering macromolecules and vaccines.
Collapse
Affiliation(s)
- Yisi Tang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; NHC Key Laboratory of Comparative Medicine, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China
| | - Bao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuting Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China.
| | - Wufa Fan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
9
|
Horsnell HL, Cao WH, Belz GT, Mueller SN, Alexandre YO. The transcription factor SpiB regulates the fibroblastic reticular cell network and CD8 + T-cell responses in lymph nodes. Immunol Cell Biol 2024; 102:269-279. [PMID: 38441326 DOI: 10.1111/imcb.12740] [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: 10/13/2023] [Revised: 02/02/2024] [Accepted: 02/14/2024] [Indexed: 04/04/2024]
Abstract
Fibroblastic reticular cells (FRCs) construct microanatomical niches that support lymph node (LN) homeostasis and coordination of immune responses. Transcription factors regulating the functionality of FRCs remain poorly understood. Here, we investigated the role of the transcription factor SpiB that is expressed in LN FRCs. Conditional ablation of SpiB in FRCs impaired the FRC network in the T-cell zone of LNs, leading to reduced numbers of FRCs and altered homeostatic functions including reduced CCL21 and interleukin-7 expression. The size and cellularity of LNs remained intact in the absence of SpiB but the space between the reticular network increased, indicating that although FRCs were reduced in number they stretched to maintain network integrity. Following virus infection, antiviral CD8+ T-cell responses were impaired, suggesting a role for SpiB expression in FRCs in orchestrating immune responses. Together, our findings reveal a new role for SpiB as an important regulator of FRC functions and immunity in LNs.
Collapse
Affiliation(s)
- Harry L Horsnell
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Wang Hj Cao
- Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- University of Queensland Frazer Institute, University of Queensland, Brisbane, QLD, Australia
| | - Gabrielle T Belz
- Walter and Eliza Hall Institute of Medical Research (WEHI), Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- University of Queensland Frazer Institute, University of Queensland, Brisbane, QLD, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Yannick O Alexandre
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
10
|
M. S. Barron A, Fabre T, De S. Distinct fibroblast functions associated with fibrotic and immune-mediated inflammatory diseases and their implications for therapeutic development. F1000Res 2024; 13:54. [PMID: 38681509 PMCID: PMC11053351 DOI: 10.12688/f1000research.143472.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/28/2023] [Indexed: 05/01/2024] Open
Abstract
Fibroblasts are ubiquitous cells that can adopt many functional states. As tissue-resident sentinels, they respond to acute damage signals and shape the earliest events in fibrotic and immune-mediated inflammatory diseases. Upon sensing an insult, fibroblasts produce chemokines and growth factors to organize and support the response. Depending on the size and composition of the resulting infiltrate, these activated fibroblasts may also begin to contract or relax thus changing local stiffness within the tissue. These early events likely contribute to the divergent clinical manifestations of fibrotic and immune-mediated inflammatory diseases. Further, distinct changes to the cellular composition and signaling dialogue in these diseases drive progressive fibroblasts specialization. In fibrotic diseases, fibroblasts support the survival, activation and differentiation of myeloid cells, granulocytes and innate lymphocytes, and produce most of the pathogenic extracellular matrix proteins. Whereas, in immune-mediated inflammatory diseases, sequential accumulation of dendritic cells, T cells and B cells programs fibroblasts to support local, destructive adaptive immune responses. Fibroblast specialization has clear implications for the development of effective induction and maintenance therapies for patients with these clinically distinct diseases.
Collapse
Affiliation(s)
- Alexander M. S. Barron
- Inflammation & Immunology Research Unit, Pfizer, Inc., Cambridge, Massachusetts, 02139, USA
| | - Thomas Fabre
- Inflammation & Immunology Research Unit, Pfizer, Inc., Cambridge, Massachusetts, 02139, USA
| | - Saurav De
- Inflammation & Immunology Research Unit, Pfizer, Inc., Cambridge, Massachusetts, 02139, USA
| |
Collapse
|
11
|
Wu Y, Zhao S, Guo W, Liu Y, Requena Mullor MDM, Rodrìguez RA, Wei R. Systematic analysis of the prognostic value and immunological function of LTBR in human cancer. Aging (Albany NY) 2024; 16:129-152. [PMID: 38175686 PMCID: PMC10817409 DOI: 10.18632/aging.205356] [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: 04/12/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2024]
Abstract
Lymphotoxin beta receptor (LTBR) is a positive T cell proliferation regulator gene. It is closely associated with the tumor immune microenvironment. However, its role in cancer and immunotherapy is unclear. Firstly, the expression level and prognostic value of LTBR were analyzed. Secondly, the expression of LTBR in clinical stages, immune subtypes, and molecular subtypes was analyzed. The correlation between LTBR and immune regulatory genes, immune checkpoint genes, and RNA modification genes was then analyzed. Correlations between LTBR and immune cells, scores, cancer-related functional status, tumor stemness index, mismatch repair (MMR) genes, and DNA methyltransferase were also analyzed. In addition, we analyzed the role of LTBR in DNA methylation, mutational status, tumor mutation burden (TMB), and microsatellite instability (MSI). Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) were used to explore the role of LTBR in pan-cancer. Finally, the drugs associated with LTBR were analyzed. The expression of LTBR was confirmed using quantitative real-time PCR and Western blot. LTBR is significantly overexpressed in most cancers and is associated with low patient survival. In addition, LTBR expression was strongly correlated with immune cells, score, cancer-related functional status, tumor stemness index, MMR genes, DNA methyltransferase, DNA methylation, mutational status, TMB, and MSI. Enrichment analysis revealed that LTBR was associated with apoptosis, necroptosis, and immune-related pathways. Finally, multiple drugs targeting LTBR were identified. LTBR is overexpressed in several tumors and is associated with a poor prognosis. It is related to immune-related genes and immune cell infiltration.
Collapse
Affiliation(s)
- Yinteng Wu
- Department of Orthopedic and Trauma Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Shijian Zhao
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Wenliang Guo
- Department of Rehabilitation Medicine, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, Guangxi 537100, China
| | - Ying Liu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | | | | | - Ruqiong Wei
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| |
Collapse
|
12
|
Samborska I, Maievskyi O, Podzihun L, Lavrynenko V. Features of immune reactivity of the spleen and mechanisms of organ damage under the influence of animal venom toxins including scorpions (review). WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:120-125. [PMID: 38431816 DOI: 10.36740/wlek202401115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
OBJECTIVE Aim: To establish features of immune reactivity of the spleen and mechanisms of organ damage under the influence of animal venom toxins including scorpions. PATIENTS AND METHODS Materials and Methods: A thorough literature analysis was conducted on the basis of PubMed, Google Scholar, Web of Science, and Scopus databases. When processing the search results, we chose the newest publications up to 5 years old or the most thorough publications that vividly described the essence of our topic. CONCLUSION Conclusions: Spleen plays a leading role in the implementation of the body's defense processes, the elimination of structural elements affected by toxins, and the restoration of immune homeostasis. Its participation in the formation of the immune response can be accompanied by qualitative and quantitative changes in histological organization. Morpho-functional changes in the spleen under the action of animal venom toxins currently require careful study, because from the information available in the literature today, it is not possible to clearly construct a complete picture of lesions of certain components of the organ at the microscopic or submicroscopic levels. Therefore, this direction of research in the medical field is currently relevant, taking into account the existence of a large number of poisonous animals, including scorpions.
Collapse
Affiliation(s)
- Inha Samborska
- NATIONAL PIROGOV MEMORIAL MEDICAL UNIVERSITY, VINNYTSIA, UKRAINE
| | - Oleksandr Maievskyi
- EDUCATIONAL AND SCIENTIFIC CENTER "INSTITUTE OF BIOLOGY AND MEDICINE" OF TARAS SHEVCHENKO NATIONAL UNIVERSITY OF KYIV, KYIV, UKRAINE
| | | | - Victoriia Lavrynenko
- EDUCATIONAL AND SCIENTIFIC CENTER "INSTITUTE OF BIOLOGY AND MEDICINE" OF TARAS SHEVCHENKO NATIONAL UNIVERSITY OF KYIV, KYIV, UKRAINE
| |
Collapse
|
13
|
Fanelli M, Petrone V, Maracchioni C, Chirico R, Cipriani C, Coppola L, Malagnino V, Teti E, Sorace C, Zordan M, Vitale P, Iannetta M, Balestrieri E, Rasi G, Grelli S, Malergue F, Sarmati L, Minutolo A, Matteucci C. Persistence of circulating CD169+monocytes and HLA-DR downregulation underline the immune response impairment in PASC individuals: the potential contribution of different COVID-19 pandemic waves. CURRENT RESEARCH IN MICROBIAL SCIENCES 2023; 6:100215. [PMID: 38187999 PMCID: PMC10767315 DOI: 10.1016/j.crmicr.2023.100215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024] Open
Abstract
The use of CD169 as a marker of viral infection has been widely discussed in the context of COVID-19, and in particular, its crucial role in the early detection of SARS-CoV-2 infection and its association with the severity and clinical outcome of COVID-19 were demonstrated. COVID-19 patients show relevant systemic alteration and immunological dysfunction that persists in individuals with post-acute sequelae of SARS-CoV-2 infection (PASC). It is critical to implement the characterization of the disease, focusing also on the possible impact of the different COVID-19 waves and the consequent effects found after infection. On this basis, we evaluated by flow cytometry the expression of CD169 and HLA-DR on monocytes from COVID-19 patients and PASC individuals to better elucidate their involvement in immunological dysfunction, also evaluating the possible impact of different pandemic waves. The results confirm CD169 RMFI is a good marker of viral infection. Moreover, COVID-19 patients and PASC individuals showed high percentage of CD169+ monocytes, but low percentage of HLA-DR+ monocytes and the alteration of systemic inflammatory indices. We have also observed alterations of CD169 and HLA-DR expression and indices of inflammation upon different COVID-19 waves. The persistence of specific myeloid subpopulations suggests a role of CD169+ monocytes and HLA-DR in COVID-19 disease and chronic post-infection inflammation, opening new opportunities to evaluate the impact of specific pandemic waves on the immune response impairment and systemic alterations with the perspective to provide new tools to monitoring new variants and diseases associated to emerging respiratory viruses.
Collapse
Affiliation(s)
- Marialaura Fanelli
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1 - 00133, Rome, 00133, Italy
| | - Vita Petrone
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1 - 00133, Rome, 00133, Italy
| | - Christian Maracchioni
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1 - 00133, Rome, 00133, Italy
| | - Rossella Chirico
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1 - 00133, Rome, 00133, Italy
| | - Chiara Cipriani
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1 - 00133, Rome, 00133, Italy
| | - Luigi Coppola
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Infectious Diseases Clinic, Policlinic of Tor Vergata, Rome, 00133, Italy
| | - Vincenzo Malagnino
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Infectious Diseases Clinic, Policlinic of Tor Vergata, Rome, 00133, Italy
| | - Elisabetta Teti
- Infectious Diseases Clinic, Policlinic of Tor Vergata, Rome, 00133, Italy
| | - Chiara Sorace
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Infectious Diseases Clinic, Policlinic of Tor Vergata, Rome, 00133, Italy
| | - Marta Zordan
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Infectious Diseases Clinic, Policlinic of Tor Vergata, Rome, 00133, Italy
| | - Pietro Vitale
- Infectious Diseases Clinic, Policlinic of Tor Vergata, Rome, 00133, Italy
| | - Marco Iannetta
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Infectious Diseases Clinic, Policlinic of Tor Vergata, Rome, 00133, Italy
| | - Emanuela Balestrieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1 - 00133, Rome, 00133, Italy
| | - Guido Rasi
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1 - 00133, Rome, 00133, Italy
| | - Sandro Grelli
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1 - 00133, Rome, 00133, Italy
- Virology Unit, Policlinic of Tor Vergata, Rome, 00133, Italy
| | - Fabrice Malergue
- Global Research Organization, Beckman Coulter Life Sciences, Marseille, 13009, France
| | - Loredana Sarmati
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Infectious Diseases Clinic, Policlinic of Tor Vergata, Rome, 00133, Italy
| | - Antonella Minutolo
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1 - 00133, Rome, 00133, Italy
| | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1 - 00133, Rome, 00133, Italy
| |
Collapse
|
14
|
Alexandre YO, Mueller SN. Splenic stromal niches in homeostasis and immunity. Nat Rev Immunol 2023; 23:705-719. [PMID: 36973361 DOI: 10.1038/s41577-023-00857-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 03/29/2023]
Abstract
The spleen is a gatekeeper of systemic immunity where immune responses against blood-borne pathogens are initiated and sustained. Non-haematopoietic stromal cells construct microanatomical niches in the spleen that make diverse contributions to physiological spleen functions and regulate the homeostasis of immune cells. Additional signals from spleen autonomic nerves also modify immune responses. Recent insight into the diversity of the splenic fibroblastic stromal cells has revised our understanding of how these cells help to orchestrate splenic responses to infection and contribute to immune responses. In this Review, we examine our current understanding of how stromal niches and neuroimmune circuits direct the immunological functions of the spleen, with a focus on T cell immunity.
Collapse
Affiliation(s)
- Yannick O Alexandre
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| |
Collapse
|
15
|
Gerberick A, Rinaldo CR, Sluis-Cremer N. Antigen Presenting Cell-Mediated HIV-1 Trans Infection in the Establishment and Maintenance of the Viral Reservoir. MEDICAL RESEARCH ARCHIVES 2023; 11:10.18103/mra.v11i7.1.4064. [PMID: 39634038 PMCID: PMC11616617 DOI: 10.18103/mra.v11i7.1.4064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Despite potent antiretroviral therapy, an HIV-1 reservoir persists that represents a major barrier to a cure. Understanding the mechanisms by which the HIV-1 reservoir is established and maintained is critical for the discovery of effective treatments to significantly reduce or eliminate the viral reservoir. In addition to cis infection, in which HIV-1 directly infects target CD4+ T cells, cell-to-cell transmission, or trans infection, can also occur. HIV-1 trans infection is significantly more efficient than cis infection, mostly due to the occurrence of multiple infections per cell during transfer. Additionally, trans infection is efficient even in the presence of ART and/or neutralizing antibodies. Cell-to-cell transmission is mediated by CD4+ T cells and professional antigen presenting cells (APC). Here we focus on APC, i.e., myeloid dendritic cells, B lymphocytes, and monocytes/macrophages, that bind, internalize, and transfer HIV-1 to target CD4+ T cells via various proposed mechanisms. We assess the potential impact of trans infection on the establishment and maintenance of the HIV-1 reservoir including its role in disease progression. We consider the natural interactions between APC and CD4+ T cells in vivo that HIV-1 may hijack, allowing for the highly efficient trans infection of CD4+ T cells, maintaining the viral reservoirs in tissue despite undetectable plasma viral loads in peripheral blood. We propose that these modes of viral pathogenesis need to be addressed in potential cure strategies to ensure eradication of the viral reservoir.
Collapse
Affiliation(s)
- Abigail Gerberick
- University of Pittsburgh School of Medicine, Department of Medicine, Division of Infectious Diseases, Pittsburgh, PA, 15261, USA
| | - Charles R Rinaldo
- University of Pittsburgh School of Medicine, Department of Medicine, Division of Infectious Diseases, Pittsburgh, PA, 15261, USA
| | - Nicolas Sluis-Cremer
- University of Pittsburgh School of Medicine, Department of Medicine, Division of Infectious Diseases, Pittsburgh, PA, 15261, USA
| |
Collapse
|
16
|
Raïch-Regué D, Resa-Infante P, Gallemí M, Laguia F, Muñiz-Trabudua X, Muñoz-Basagoiti J, Perez-Zsolt D, Chojnacki J, Benet S, Clotet B, Martinez-Picado J, Izquierdo-Useros N. Role of Siglecs in viral infections: A double-edged sword interaction. Mol Aspects Med 2023; 90:101113. [PMID: 35981912 PMCID: PMC9923124 DOI: 10.1016/j.mam.2022.101113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 01/21/2023]
Abstract
Sialic-acid-binding immunoglobulin-like lectins are cell surface immune receptors known as Siglecs that play a paramount role as modulators of immunity. In recent years, research has underscored how the underlaying biology of this family of receptors influences the outcome of viral infections. While Siglecs are needed to promote effective antiviral immune responses, they can also pave the way to viral dissemination within tissues. Here, we review how recent preclinical findings focusing on the interplay between Siglecs and viruses may translate into promising broad-spectrum therapeutic interventions or key biomarkers to monitor the course of viral infections.
Collapse
Affiliation(s)
- Dàlia Raïch-Regué
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain
| | - Patricia Resa-Infante
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain; University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Spain
| | - Marçal Gallemí
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain
| | - Fernando Laguia
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain
| | - Xabier Muñiz-Trabudua
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain
| | | | - Daniel Perez-Zsolt
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain
| | - Jakub Chojnacki
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain; Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Spain
| | - Susana Benet
- Fundació lluita contra la SIDA, Infectious Diseases Department, Hospital Germans Trias i Pujol, 08916, Badalona, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain; University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Spain; Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Spain; Fundació lluita contra la SIDA, Infectious Diseases Department, Hospital Germans Trias i Pujol, 08916, Badalona, Spain; Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain; University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Spain; Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Spain; Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029, Madrid, Spain; Catalan Institution for Research and Advanced Studies (ICREA), 08010, Barcelona, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain; Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Spain; Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
17
|
Herzog S, Fragkou PC, Arneth BM, Mkhlof S, Skevaki C. Myeloid CD169/Siglec1: An immunoregulatory biomarker in viral disease. Front Med (Lausanne) 2022; 9:979373. [PMID: 36213653 PMCID: PMC9540380 DOI: 10.3389/fmed.2022.979373] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
CD169, also known as Siglec1 or Sialoadhesin (Sn), is a surface adhesion molecule on human myeloid cells. Being part of the Siglec family, it acts as a receptor for sialylated molecular structures, which are found among various pathogenic and non-pathogenic ligands. Recent data suggest that CD169 may represent a promising new biomarker in acute respiratory and non-respiratory viral infections, such as SARS-CoV-2, Respiratory syncytial virus (RSV) and Human immunodeficiency virus (HIV). Therein lies a great potential to sufficiently differentiate viral from bacterial infection, which has been an incessant challenge in the clinical management of infectious disease. CD169 equips myeloid cells with functions, reaching far beyond pathogen elimination. In fact, CD169 seems to crosslink innate and adaptive immunity by antigen presentation and consecutive pathogen elimination, embodying a substantial pillar of immunoregulation. Yet, our knowledge about the kinetics, mechanisms of induction, signaling pathways and its precise role in host-pathogen interaction remains largely obscure. In this review, we describe the role of CD169 as a potentially novel diagnostic biomarker for respiratory viral infection by evaluating its strengths and weaknesses and considering host factors that are involved in pathogenesis of virus infection. Finally, this brief review aims to point out shortcomings of available evidence, thus, guiding future work revolving the topic.
Collapse
Affiliation(s)
- Silva Herzog
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Justus Liebig University Giessen, Giessen, Germany
- The European Society of Clinical Microbiology and Infection (ESCMID) Study Group for Respiratory Viruses (ESGREV), Basel, Switzerland
| | - Paraskevi C. Fragkou
- The European Society of Clinical Microbiology and Infection (ESCMID) Study Group for Respiratory Viruses (ESGREV), Basel, Switzerland
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, Evangelismos Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Borros M. Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Justus Liebig University Giessen, Giessen, Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps-University Marburg, Marburg, Germany
| | - Samr Mkhlof
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Justus Liebig University Giessen, Giessen, Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps-University Marburg, Marburg, Germany
| | - Chrysanthi Skevaki
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Justus Liebig University Giessen, Giessen, Germany
- The European Society of Clinical Microbiology and Infection (ESCMID) Study Group for Respiratory Viruses (ESGREV), Basel, Switzerland
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps-University Marburg, Marburg, Germany
- Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Marburg, Germany
- *Correspondence: Chrysanthi Skevaki,
| |
Collapse
|