1
|
Takahashi H, Perez-Villarroel P, Falahat R, Mulé JJ. Targeting MARCO in combination with anti-CTLA-4 leads to enhanced melanoma regression and immune cell infiltration via macrophage reprogramming. J Immunother Cancer 2025; 13:e011030. [PMID: 40081947 PMCID: PMC11907082 DOI: 10.1136/jitc-2024-011030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Strategies to improve the therapeutic efficacy of cancer immunotherapy with immune checkpoint inhibitors include targeting additional immunosuppressive compartments in the tumor microenvironment (TME). Inhibitory macrophages (Mφ) can be one of the most abundant immune cells in the TME associated with poor prognosis. However, to date, selective Mφ depletion strategies as a cancer immunotherapy have not been successful in clinical trials. Macrophage Receptor with Collagenous Structure (MARCO) is one of a family of class-A scavenger receptors expressed by Mφ in the TME and is one of the most upregulated transcripts in dendritic cells (DC) following their ex vivo uptake of dead tumor cells. The clinical significance of MARCO expression in the TME is not fully understood. METHODS The therapeutic potential of targeting MARCO by an anti-murine MARCO (ED31, clone ED31) monoclonal antibody, which inhibits ligand-binding to MARCO, was explored in combination with anti-cytotoxic T-lymphocyte associated protein 4 (anti-CTLA-4) or anti-programmed cell death protein-1 (anti-PD-1) in C57BL/6J mice bearing B16F10 or Pan02 tumors. The mechanism by which ED31 impacts the TME was investigated by flow cytometry in the different treatment arms. The contribution of Mφ was assessed by both in vivo depletion and in vitro functional assays. Chemokine production was measured by a bead-based multiplex assay. RESULTS ED31 enhanced antitumor efficacy of anti-CTLA-4, but not of anti-PD-1. Analysis of the TME revealed that adding ED31 to anti-CTLA-4 substantially increased immune cell infiltration, including mature conventional DC recruitment, that was due to a switch to M1-pattern chemokines by Mφ. Mφ depletion completely abrogated both the increase in immune cell infiltration and chemokine production, and abolished the antitumor efficacy of the combination therapy. CONCLUSIONS Targeting MARCO as an additional checkpoint in the TME can offer a strategy to improve the antitumor efficacy of anti-CTLA-4 through a mechanism involving Mφ reprogramming rather than their depletion.
Collapse
Affiliation(s)
| | | | - Rana Falahat
- Immunology, Moffitt Cancer Center, Tampa, Florida, USA
| | - James J Mulé
- Immunology, Moffitt Cancer Center, Tampa, Florida, USA
- Cutaneous Oncology Program, Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
2
|
Damigos S, Caliskan A, Wajant G, Giddins S, Moldovan A, Kuhn S, Putz E, Dandekar T, Rudel T, Westermann AJ, Zdzieblo D. A Multicellular In Vitro Model of the Human Intestine with Immunocompetent Features Highlights Host-Pathogen Interactions During Early Salmonella Typhimurium Infection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411233. [PMID: 39807570 PMCID: PMC11884561 DOI: 10.1002/advs.202411233] [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: 09/13/2024] [Revised: 12/24/2024] [Indexed: 01/16/2025]
Abstract
Studying the molecular basis of intestinal infections caused by enteric pathogens at the tissue level is challenging, because most human intestinal infection models have limitations, and results obtained from animals may not reflect the human situation. Infections with Salmonella enterica serovar Typhimurium (STm) have different outcomes between organisms. 3D tissue modeling of primary human material provides alternatives to animal experimentation, but epithelial co-culture with immune cells remains difficult. Macrophages, for instance, contribute to the immunocompetence of native tissue, yet their incorporation into human epithelial tissue models is challenging. A 3D immunocompetent tissue model of the human small intestine based on decellularized submucosa enriched with monocyte-derived macrophages (MDM) is established. The multicellular model recapitulated in vivo-like cellular diversity, especially the induction of GP2 positive microfold (M) cells. Infection studies with STm reveal that the pathogen physically interacts with these M-like cells. MDMs show trans-epithelial migration and phagocytosed STm within the model and the levels of inflammatory cytokines are induced upon STm infection. Infected epithelial cells are shed into the supernatant, potentially reflecting an intracellular reservoir of invasion-primed STm. Together, the 3D model of the human intestinal epithelium bears potential as an alternative to animals to identify human-specific processes underlying enteric bacterial infections.
Collapse
Affiliation(s)
- Spyridon Damigos
- Department for Functional Materials in Medicine and DentistryUniversity Hospital WürzburgWürzburgGermany
| | - Aylin Caliskan
- Department of BioinformaticsBiocenterUniversity of WürzburgWürzburgGermany
| | - Gisela Wajant
- Department for Functional Materials in Medicine and DentistryUniversity Hospital WürzburgWürzburgGermany
| | - Sara Giddins
- Department for Functional Materials in Medicine and DentistryUniversity Hospital WürzburgWürzburgGermany
| | - Adriana Moldovan
- Department of MicrobiologyBiocenterUniversity of WürzburgWürzburgGermany
| | - Sabine Kuhn
- Institute of Clinical Transfusion Medicine and HemotherapyUniversity of WuerzburgWuerzburgGermany
| | - Evelyn Putz
- Institute of Clinical Transfusion Medicine and HemotherapyUniversity of WuerzburgWuerzburgGermany
| | - Thomas Dandekar
- Department of BioinformaticsBiocenterUniversity of WürzburgWürzburgGermany
| | - Thomas Rudel
- Department of MicrobiologyBiocenterUniversity of WürzburgWürzburgGermany
| | - Alexander J. Westermann
- Department of MicrobiologyBiocenterUniversity of WürzburgWürzburgGermany
- Helmholtz‐Institute for RNA‐based Infection Research (HIRI)Helmholtz Centre for Infection Research (HZI)WürzburgGermany
| | - Daniela Zdzieblo
- Department for Functional Materials in Medicine and DentistryUniversity Hospital WürzburgWürzburgGermany
- Translational Center for Regenerative Therapies (TLC‐RT)Fraunhofer Institute for Silicate Research (ISC)97070WürzburgGermany
| |
Collapse
|
3
|
Palacios PA, Santibañez Á, Aguirre-Muñoz F, Gutiérrez-Vera C, Niño de Zepeda-Carrizo V, Góngora-Pimentel M, Müller M, Cáceres M, Kalergis AM, Carreño LJ. Can invariant Natural Killer T cells drive B cell fate? a look at the humoral response. Front Immunol 2025; 16:1505883. [PMID: 40040714 PMCID: PMC11876049 DOI: 10.3389/fimmu.2025.1505883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/21/2025] [Indexed: 03/06/2025] Open
Abstract
Invariant Natural Killer T (NKT) cells represent a unique subset of innate-like T cells that express both NK cell and T cell receptors. These cells are rapidly activated by glycolipid antigens presented via CD1d molecules on antigen-presenting cells (APCs), including B cells, dendritic cells (DCs), and macrophages, or through cytokine-dependent mechanisms. Their ability to produce a wide range of cytokines and express costimulatory molecules underscores their critical role in bridging innate and adaptive immunity. B cells, traditionally recognized for their role in antibody production, also act as potent APCs due to their high expression of CD1d, enabling direct interactions with iNKT cells. This interaction has significant implications for humoral immunity, influencing B cell activation, class-switch recombination (CSR), germinal center formation, and memory B cell differentiation, thus expanding the conventional paradigm of T cell-B cell interactions. While the influence of iNKT cells on B cell biology and humoral responses is well-supported, many aspects of their interaction remain unresolved. Key questions include the roles of different iNKT cell subsets, the diversity of APCs, the spatiotemporal dynamics of these interactions, especially during early activation, and the potential for distinct glycolipid ligands to modulate immune outcomes. Understanding these factors could provide valuable insights into how iNKT cells regulate B cell-mediated immunity and offer opportunities to harness these interactions in immunotherapeutic applications, such as vaccine development. In this review, we examine these unresolved aspects and propose a novel perspective on the regulatory potential of iNKT cells in humoral immunity, emphasizing their promise as a target for innovative vaccine strategies.
Collapse
Affiliation(s)
- Pablo A. Palacios
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Álvaro Santibañez
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Fernanda Aguirre-Muñoz
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Cristián Gutiérrez-Vera
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Valentina Niño de Zepeda-Carrizo
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Martín Góngora-Pimentel
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marioly Müller
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mónica Cáceres
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leandro J. Carreño
- Millennium Institute on Immunology and Immunotherapy, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
4
|
Zhong G, Chang X, Xie W, Zhou X. Targeted protein degradation: advances in drug discovery and clinical practice. Signal Transduct Target Ther 2024; 9:308. [PMID: 39500878 PMCID: PMC11539257 DOI: 10.1038/s41392-024-02004-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/19/2024] [Accepted: 09/28/2024] [Indexed: 11/08/2024] Open
Abstract
Targeted protein degradation (TPD) represents a revolutionary therapeutic strategy in disease management, providing a stark contrast to traditional therapeutic approaches like small molecule inhibitors that primarily focus on inhibiting protein function. This advanced technology capitalizes on the cell's intrinsic proteolytic systems, including the proteasome and lysosomal pathways, to selectively eliminate disease-causing proteins. TPD not only enhances the efficacy of treatments but also expands the scope of protein degradation applications. Despite its considerable potential, TPD faces challenges related to the properties of the drugs and their rational design. This review thoroughly explores the mechanisms and clinical advancements of TPD, from its initial conceptualization to practical implementation, with a particular focus on proteolysis-targeting chimeras and molecular glues. In addition, the review delves into emerging technologies and methodologies aimed at addressing these challenges and enhancing therapeutic efficacy. We also discuss the significant clinical trials and highlight the promising therapeutic outcomes associated with TPD drugs, illustrating their potential to transform the treatment landscape. Furthermore, the review considers the benefits of combining TPD with other therapies to enhance overall treatment effectiveness and overcome drug resistance. The future directions of TPD applications are also explored, presenting an optimistic perspective on further innovations. By offering a comprehensive overview of the current innovations and the challenges faced, this review assesses the transformative potential of TPD in revolutionizing drug development and disease management, setting the stage for a new era in medical therapy.
Collapse
Affiliation(s)
- Guangcai Zhong
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Xiaoyu Chang
- School of Pharmaceutical Sciences, Pingyuan Laboratory, Zhengzhou University, Zhengzhou, 450001, China
| | - Weilin Xie
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| |
Collapse
|
5
|
Ma H, Zhu M, Chen M, Li X, Feng X. The role of macrophage plasticity in neurodegenerative diseases. Biomark Res 2024; 12:81. [PMID: 39135084 PMCID: PMC11321226 DOI: 10.1186/s40364-024-00624-7] [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: 04/28/2024] [Accepted: 07/22/2024] [Indexed: 08/15/2024] Open
Abstract
Tissue-resident macrophages and recruited macrophages play pivotal roles in innate immunity and the maintenance of brain homeostasis. Investigating the involvement of these macrophage populations in eliciting pathological changes associated with neurodegenerative diseases has been a focal point of research. Dysregulated states of macrophages can compromise clearance mechanisms for pathological proteins such as amyloid-β (Aβ) in Alzheimer's disease (AD) and TDP-43 in Amyotrophic lateral sclerosis (ALS). Additionally, recent evidence suggests that abnormalities in the peripheral clearance of pathological proteins are implicated in the pathogenesis and progression of neurodegenerative diseases. Furthermore, numerous genome-wide association studies have linked genetic risk factors, which alter the functionality of various immune cells, to the accumulation of pathological proteins. This review aims to unravel the intricacies of macrophage biology in both homeostatic conditions and neurodegenerative disorders. To this end, we initially provide an overview of the modifications in receptor and gene expression observed in diverse macrophage subsets throughout development. Subsequently, we outlined the roles of resident macrophages and recruited macrophages in neurodegenerative diseases and the progress of targeted therapy. Finally, we describe the latest advances in macrophage imaging methods and measurement of inflammation, which may provide information and related treatment strategies that hold promise for informing the design of future investigations and therapeutic interventions.
Collapse
Affiliation(s)
- Hongyue Ma
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Mingxia Zhu
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Mengjie Chen
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Xiuli Li
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Xinhong Feng
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China.
| |
Collapse
|
6
|
Zhou G, Zhang L, Shao S. The application of MARCO for immune regulation and treatment. Mol Biol Rep 2024; 51:246. [PMID: 38300385 DOI: 10.1007/s11033-023-09201-x] [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: 10/27/2023] [Accepted: 12/30/2023] [Indexed: 02/02/2024]
Abstract
Macrophage receptor with collagen structure (MARCO) is a member of scavenger receptor class A (SR-A) and shares structural and functional similarities with SR-A1. In recent years, many studies have shown that MARCO can trigger an immune response and has therapeutic potential as a target for immunotherapy. Studies have shown that alterations in MARCO expression following pathogen infection cause changes in the functions of innate and adaptive immune cells, including macrophages, dendritic cells, B cells, and T cells, affecting the body's immune response to invading pathogens; thus, MARCO plays a crucial role in triggering the immune response, bridging innate and adaptive immunity, and eliminating pathogens. This paper is a comprehensive summary of the recent research on MARCO. This review focuses on the multiple functions of MARCO, including adhesion, migration, phagocytosis, and cytokine secretion with special emphasis on the complex interactions between MARCO and various types of cells involved in the immune response, as well as possible immune-related mechanisms. In summary, in this review, we discuss the structure and function of MARCO and its role in the immune response and highlight the therapeutic potential of MARCO as a target for immunotherapy. We hope that this review provides a theoretical basis for future research on MARCO.
Collapse
Affiliation(s)
- Guiyuan Zhou
- Department of Histology and Embryology, Hebei Medical University, No. 361, Zhongshan East Road, Chang'an District, Shijiazhuang, 050017, China
| | - Lei Zhang
- Shijiazhuang Vocational College of City Economy, No. 12, Wenming Road, Economic and Technological Development Zone, Shijiazhuang, 050017, China.
| | - Suxia Shao
- Department of Histology and Embryology, Hebei Medical University, No. 361, Zhongshan East Road, Chang'an District, Shijiazhuang, 050017, China.
| |
Collapse
|
7
|
Sharma P, Venkatachalam K, Binesh A. Decades Long Involvement of THP-1 Cells as a Model for Macrophage Research: A Comprehensive Review. Antiinflamm Antiallergy Agents Med Chem 2024; 23:85-104. [PMID: 38676532 DOI: 10.2174/0118715230294413240415054610] [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: 01/03/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/29/2024]
Abstract
Over the years, researchers have endeavored to identify dependable and reproducible in vitro models for examining macrophage behavior under controlled conditions. The THP-1 cell line has become a significant and widely employed tool in macrophage research within these models. Originating from the peripheral blood of individuals with acute monocytic leukemia, this human monocytic cell line can undergo transformation into macrophage-like cells, closely mirroring primary human macrophages when exposed to stimulants. Macrophages play a vital role in the innate immune system, actively regulating inflammation, responding to infections, and maintaining tissue homeostasis. A comprehensive understanding of macrophage biology and function is crucial for gaining insights into immunological responses, tissue healing, and the pathogenesis of diseases such as viral infections, autoimmune disorders, and neoplastic conditions. This review aims to thoroughly evaluate and emphasize the extensive history of THP-1 cells as a model for macrophage research. Additionally, it will delve into the significance of THP-1 cells in advancing our comprehension of macrophage biology and their invaluable contributions to diverse scientific domains.
Collapse
Affiliation(s)
- Prakhar Sharma
- Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, 603103, Tamil Nadu, India
| | - Kaliyamurthi Venkatachalam
- Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, 603103, Tamil Nadu, India
| | - Ambika Binesh
- Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, 603103, Tamil Nadu, India
| |
Collapse
|
8
|
Meurman JH, Goldberg M. Irma Thesleff-Orthodontist who became a developmental biologist. Oral Dis 2023; 29 Suppl 1:883-885. [PMID: 36149325 DOI: 10.1111/odi.14382] [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: 08/30/2022] [Revised: 09/29/2022] [Accepted: 09/17/2022] [Indexed: 11/27/2022]
Abstract
Irma Thesleff is one of the leading scholars in developmental biology. She and her research group have clarified the mysteries of tooth development. For several decades, her research of very high quality has focused on morphogenesis and resulted in an understanding of the highly complex signaling networks. Irma Thesleff has been duly recognized both in the domestic and international context. Her research continues despite her retirement.
Collapse
Affiliation(s)
- Jukka H Meurman
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Michel Goldberg
- Biomédicale des Saints Pères, Université Paris Descartes, Paris, France
| |
Collapse
|
9
|
Activation of macrophages mediates dietary restriction-induced splenic involution. Life Sci 2022; 310:121068. [DOI: 10.1016/j.lfs.2022.121068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 09/29/2022] [Accepted: 10/07/2022] [Indexed: 11/09/2022]
|
10
|
Zhong A, Gao T. Transcriptome analysis reveals similarities and differences in immune responses in the head and trunk kidneys of yellow catfish (Pelteobagrus fulvidraco) stimulated with Aeromonas hydrophila. FISH & SHELLFISH IMMUNOLOGY 2022; 130:155-163. [PMID: 36055554 DOI: 10.1016/j.fsi.2022.08.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/19/2022] [Accepted: 08/15/2022] [Indexed: 06/15/2023]
Abstract
Teleosts have a unique immune system because their head kidney (HK) and trunk kidney (TK) are sites for hematopoiesis. However, the immune functions of the HK and TKs require further elucidation in yellow catfish (Pelteobagrus fulvidraco). In the present study, imprints of the HK and TK were examined using the Wright's-Giemsa staining method. Morphological characteristics of the blood cell lineages revealed that the HK and TK were hematopoietic organs. To explore its immune function, transcriptome sequencing was performed after infection with Aeromonas hydrophila. A total of 1139 genes showed significant alterations in their expression in the kidney; these genes included 737 upregulated and 402 downregulated genes. Furthermore, 1117 differentially expressed genes were observed in the HK, which included 784 upregulated and 333 downregulated genes. Both organs showed 357 upregulated genes and 85 downregulated genes. Some immune-related genes were only expressed in the TK, such as ATP-dependent RNA helicase DDX58, the gene encoding the immunoglobulin heavy chain and light chain. The immune responses in the HK and TK were differential and the TK played a critical role in the mechanism underlying the immune response. The purpose of the present study was to facilitate the elucidation of the immune defense mechanism of yellow catfish and other teleosts.
Collapse
Affiliation(s)
- Aihua Zhong
- Aquaculture Department, College of Fishery, Zhejiang Ocean University, No.1, Haida South Road, Changzhi Island, Zhoushan, Zhejiang Province, 316022, China.
| | - Tianxiang Gao
- Aquaculture Department, College of Fishery, Zhejiang Ocean University, No.1, Haida South Road, Changzhi Island, Zhoushan, Zhejiang Province, 316022, China
| |
Collapse
|
11
|
Guo X, Liu Y, Liu J, Xu D, Chi C, Lv Z, Liu H. Sequence and functional features of a novel scavenger receptor homolog, SCARA5 from Yellow drum (Nibea albiflora). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 135:104463. [PMID: 35690228 DOI: 10.1016/j.dci.2022.104463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
As an important member in SR-As, member 5 (SCARA5) can swallow apoptotic cells and foreign bodies, and participate multiple signaling pathways to inhibit tumor occurrence, development growth and metastasis. To explore its immune function, SCARA5 was identified from the yellow drum (Nibea albiflora) according to its transcriptome data, and its full-length cDNA was 6968 bp (named as NaSCARA5, GenBank accession no: MW070211) encoding 497 amino acids with a calculated molecular weight of 55.12 kDa, which had the typical motifs of SR family, such as transmembrane helix region, coil region, Pfam collagens region and SR region. BLASTp and the phylogenetic relationship analysis illustrated that the sequences shared high similarity with known SCARA5 of teleosts. Quantitative real time RT-PCR analysis showed that NaSCARA5 was expressed in intestine, stomach, liver, kidney, gill, heart and spleen, with the highest in the spleen (24.42-fold compared with that in heart). After being infected with Polyinosinic:polycytidylic acid (PolyI:C), Vibrio alginolyticus and Vibrio parahaemolyticus, NaSCARA5 mRNA were up-regulated with time dependent mode in spleen, which suggested that NaSCARA5 might play an important role in the immune process of fish. The extracellular domain of NaSCARA5 was successfully expressed in BL21 (DE3), and yielded the target protein of the expected size with many active sites for their conferring protein-protein interaction functions. After being purified by Ni-NAT Superflow resin and renatured, it was found to bind all the tested bacteria (V.parahaemolyticus,V.alginolyticus and Vibrio harveyi). The eukaryotic expression vector of the NaSCARA5-EGFP fusion protein was constructed and transferred into epithelioma papulosum cyprini (EPC) cells, and it was mainly expressed on the cell membrane indicating that NaSCARA5 was a typical transmembrane protein. The aforementioned results indicated that NaSCARA5 played a significant role in the defense against pathogenic bacteria infection as PRRs, which may provide some further understandings of the regulatory mechanisms in the fish innate immune system for SR family.
Collapse
Affiliation(s)
- Xiaoxian Guo
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, PR China
| | - Yue Liu
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, PR China
| | - Jiaxin Liu
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, PR China
| | - Dongdong Xu
- Marine Fishery Institute of Zhejiang Province, Key Lab of Mariculture and Enhancement of Zhejiang Province, Zhoushan, 316100, China
| | - Changfeng Chi
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, PR China
| | - Zhenming Lv
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, PR China
| | - Huihui Liu
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, PR China.
| |
Collapse
|
12
|
Kimura S, Yokoyama S, Pilon AL, Kurotani R. Emerging role of an immunomodulatory protein secretoglobin 3A2 in human diseases. Pharmacol Ther 2022; 236:108112. [PMID: 35016921 PMCID: PMC9271138 DOI: 10.1016/j.pharmthera.2022.108112] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/30/2021] [Accepted: 01/06/2022] [Indexed: 12/27/2022]
Abstract
Secretoglobin (SCGB) 3A2 was first identified in 2001 as a protein exhibiting similarities in amino acid sequence and gene structure to SCGB1A1, a multi-functional cytokine-like molecule highly expressed in airway epithelial Club cells that was the first identified and extensively studied member of the SCGB gene superfamily. SCGB3A2 is a small secretory protein of ~10 kDa that forms a dimer and a tetramer. SCGB3A2 is predominantly expressed in airway epithelial Club cells, and has anti-inflammatory, growth factor, anti-fibrotic, and anti-cancer activities that influence various lung diseases. This review summarizes the current understanding of SCGB3A2 biological functions and its role in human diseases with emphasis on its mechanisms of actions and signaling pathway.
Collapse
Affiliation(s)
- Shioko Kimura
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Shigetoshi Yokoyama
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | | | - Reiko Kurotani
- Biochemical Engineering, Graduate School of Science and Engineering, Yamagata University, Yonezawa, Yamagata 992-8510, Japan
| |
Collapse
|
13
|
Dienst EGT, Kremer EJ. Adenovirus receptors on antigen-presenting cells of the skin. Biol Cell 2022; 114:297-308. [PMID: 35906865 DOI: 10.1111/boc.202200043] [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: 05/16/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 12/01/2022]
Abstract
Skin, the largest human organ, is part of the first line of physical and immunological defense against many pathogens. Understanding how skin antigen-presenting cells (APCs) respond to viruses or virus-based vaccines is crucial to develop antiviral pharmaceutics, and efficient and safe vaccines. Here, we discuss the way resident and recruited skin APCs engage adenoviruses and the impact on innate immune responses. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS, Montpellier, France
| |
Collapse
|
14
|
Chinn HK, Gardell JL, Matsumoto LR, Labadie KP, Mihailovic TN, Lieberman NAP, Davis A, Pillarisetty VG, Crane CA. Hypoxia-inducible lentiviral gene expression in engineered human macrophages. J Immunother Cancer 2022; 10:jitc-2021-003770. [PMID: 35728871 PMCID: PMC9214393 DOI: 10.1136/jitc-2021-003770] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background Human immune cells, including monocyte-derived macrophages, can be engineered to deliver proinflammatory cytokines, bispecific antibodies, and chimeric antigen receptors to support immune responses in different disease settings. When gene expression is regulated by constitutively active promoters, lentiviral payload gene expression is unregulated, and can result in potentially toxic quantities of proteins. Regulated delivery of lentivirally encoded proteins may allow localized or conditional therapeutic protein expression to support safe delivery of adoptively transferred, genetically modified cells with reduced capacity for systemic toxicities. Methods In this study, we engineered human macrophages to express genes regulated by hypoxia responsive elements included in the lentiviral promoter region to drive conditional lentiviral gene expression only under hypoxic conditions. We tested transduced macrophages cultured in hypoxic conditions for the transient induced expression of reporter genes and the secreted cytokine, interleukin-12. Expression of hypoxia-regulated genes was investigated both transcriptionally and translationally, and in the presence of human tumor cells in a slice culture system. Finally, hypoxia-regulated gene expression was evaluated in a subcutaneous humanized-mouse cancer model. Results Engineered macrophages were shown to conditionally and tranisently express lentivirally encoded gene protein products, including IL-12 in hypoxic conditions in vitro. On return to normoxic conditions, lentiviral payload expression returned to basal levels. Reporter genes under the control of hypoxia response elements were upregulated under hypoxic conditions in the presence of human colorectal carcinoma cells and in the hypoxic xenograft model of glioblastoma, suggesting utility for systemic engineered cell delivery capable of localized gene delivery in cancer. Conclusions Macrophages engineered to express hypoxia-regulated payloads have the potential to be administered systemically and conditionally express proteins in tissues with hypoxic conditions. In contrast to immune cells that function or survive poorly in hypoxic conditions, macrophages maintain a proinflammatory phenotype that may support continued gene and protein expression when regulated by conditional hypoxia responsive elements and naturally traffic to hypoxic microenvironments, making them ideal vehicles for therapeutic payloads to hypoxic tissues, such as solid tumors. With the ability to fine-tune delivery of potent proteins in response to endogenous microenvironments, macrophage-based cellular therapies may therefore be designed for different disease settings.
Collapse
Affiliation(s)
- Harrison K Chinn
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Jennifer L Gardell
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington, USA.,Mozart Therapeutics, Seattle, Washington, USA
| | - Lisa R Matsumoto
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Kevin P Labadie
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Tara N Mihailovic
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Nicole A P Lieberman
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Amira Davis
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | | | | |
Collapse
|
15
|
Lama R, Pereiro P, Figueras A, Novoa B. Zebrafish as a Vertebrate Model for Studying Nodavirus Infections. Front Immunol 2022; 13:863096. [PMID: 35401537 PMCID: PMC8987509 DOI: 10.3389/fimmu.2022.863096] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
Nervous necrosis virus (NNV) is a neurotropic pathogenic virus affecting a multitude of marine and freshwater fish species that has a high economic impact on aquaculture farms worldwide. Therefore, the development of new tools and strategies aimed at reducing the mortality caused by this virus is a pivotal need. Although zebrafish is not considered a natural host for NNV, the numerous experimental advantages of this species make zebrafish an attractive model for studying different aspects of the disease caused by NNV, viral encephalopathy and retinopathy (VER). In this work, we established the best way and age to infect zebrafish larvae with NNV, obtaining significant mortalities in 3-day-postfertilization larvae when the virus was inoculated directly into the brain or by intramuscular microinjection. As occurs in naturally susceptible fish species, we confirmed that after intramuscular injection the virus was able to migrate to the central nervous system (CNS). As expected, due to the severe damage that this virus causes to the CNS, alterations in the swimming behavior of the zebrafish larvae were also observed. Taking advantage of the existence of transgenic fluorescent zebrafish lines, we were able to track the migration of different innate immune cells, mainly neutrophils, to the site of infection with NNV via the brain. However, we did not observe colocalization between the viral particles and neutrophils. RNA-Seq analysis of NNV-infected and uninfected larvae at 1, 3 and 5 days postinfection (dpi) revealed a powerful modulation of the antiviral immune response, especially at 5 dpi. We found that this response was dominated by, though not restricted to, the type I interferon system, the major defence mechanism in the innate immune response against viral pathogens. Therefore, as zebrafish larvae are able to develop the main characteristic of NNV infection and respond with an efficient immune arsenal, we confirmed the suitability of zebrafish larvae for modelling VER disease and studying different aspects of NNV pathogenesis, immune response and screening of antiviral drugs.
Collapse
|
16
|
Schaefer REM, Callahan RC, Atif SM, Orlicky DJ, Cartwright IM, Fontenot AP, Colgan SP, Onyiah JC. Disruption of monocyte-macrophage differentiation and trafficking by a heme analog during active inflammation. Mucosal Immunol 2022; 15:244-256. [PMID: 34916594 PMCID: PMC8881314 DOI: 10.1038/s41385-021-00474-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 09/23/2021] [Accepted: 11/23/2021] [Indexed: 02/04/2023]
Abstract
Heme metabolism is a key regulator of inflammatory responses. Cobalt protoporphyrin IX (CoPP) is a heme analog and mimic that potently activates the NRF2/heme oxygenase-1 (HO-1) pathway, especially in monocytes and macrophages. We investigated the influence of CoPP on inflammatory responses using a murine model of colitis. Surprisingly, conditional deletion of myeloid HO-1 did not impact the colonic inflammatory response or the protective influence of CoPP in the setting of dextran sodium sulfate-induced colitis. Rather, we reveal that CoPP elicits a contradictory shift in blood myeloid populations relative to the colon during active intestinal inflammation. Major population changes include markedly diminished trafficking of CCR2+Ly6Chi monocytes to the inflamed colon, despite significant mobilization of this population into circulation. This resulted in significantly diminished colonic expansion of monocyte-derived macrophages and inflammatory cytokine expression. These findings were linked with significant induction of systemic CCL2 leading to a disrupted CCL2 chemoattractant gradient toward the colon and concentration-dependent suppression of circulating monocyte CCR2 expression. Administration of CoPP also induced macrophage differentiation toward a MarcohiHmox1hi anti-inflammatory erythrophagocytic phenotype, contributing to an overall decreased inflammatory profile. Such findings redefine protective influences of heme metabolism during inflammation, and highlight previously unreported immunosuppressive mechanisms of endogenous CCL2 induction.
Collapse
Affiliation(s)
- Rachel E. M. Schaefer
- Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO,Department of Medicine, University of Colorado School of Medicine, Aurora, CO,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Rosemary C. Callahan
- Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO,Department of Medicine, University of Colorado School of Medicine, Aurora, CO,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Shaikh M. Atif
- Division of Allergy, Asthma and Clinical Immunology, University of Colorado School of Medicine, Aurora, CO
| | - David J. Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO
| | - Ian M. Cartwright
- Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO,Department of Medicine, University of Colorado School of Medicine, Aurora, CO,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Andrew P. Fontenot
- Division of Allergy, Asthma and Clinical Immunology, University of Colorado School of Medicine, Aurora, CO
| | - Sean P. Colgan
- Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO,Department of Medicine, University of Colorado School of Medicine, Aurora, CO,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Joseph C. Onyiah
- Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO,Department of Medicine, University of Colorado School of Medicine, Aurora, CO,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, Corresponding author: Joseph C. Onyiah, M.D., University of Colorado School of Medicine, Rocky Mountain Regional VA Medical Center, 12700 East 19th Ave. MS B-146, Aurora, CO 80045,
| |
Collapse
|
17
|
Linares-Alcántara E, Mendlovic F. Scavenger Receptor A1 Signaling Pathways Affecting Macrophage Functions in Innate and Adaptive Immunity. Immunol Invest 2022; 51:1725-1755. [PMID: 34986758 DOI: 10.1080/08820139.2021.2020812] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
First discovered on macrophages by Goldstein and Brown in 1979, Scavenger Receptors have since been shown to participate in a diverse number of cell functions; equally diverse are their structures and the ligands they bind. Macrophage activation is crucial in the outcome of an immune response. SR-A1 is highly abundant on macrophages and recognizes both host- and microorganism-derived molecules that impact processes that are initiated, perpetuated, or modified. This review summarizes the involvement of SR-A1 in both inflammatory and anti-inflammatory responses, the multiple-ligand internalization mechanisms and the diversity of signaling pathways that impact macrophage function and activation. Engagement of SR-A1 results in the stimulation of differential signaling pathways and patterns of cytokine expression, kinetics, magnitude of response and activation status. SR-A1 plays essential roles in phagocytosis and efferocytosis, interacting with other receptors and promoting tolerance in response to apoptotic cell uptake. In cell adhesion, tissue remodeling, and cell migration, SR-A1 signals through different pathways engaging different cytoplasmic motifs. We describe the role of SR-A1 during innate and adaptive immune responses, such as participation in macrophage polarization and interaction with other innate receptors, as well as in antigen uptake, processing, and presentation, regulating T and B cell activation. The dichotomous contribution of SR-A1 on macrophage functions is discussed. A better understanding of the role SR-A1 plays through molecular mechanisms and crosstalk with other receptors may provide insights into developing novel therapeutic strategies to modulate immune responses and immunopathologies.
Collapse
Affiliation(s)
- Elizabeth Linares-Alcántara
- Facultad de Ciencias, UNAM, Av. Universidad 3000, Col. Copilco-Universidad, Ciudad de Mexico, Mexico.,Departamento de Microbiología y Parasitología, Facultad de Medicina, UNAM, Av. Universidad 3000, Col. Copilco-Universidad, Ciudad de Mexico, Mexico
| | - Fela Mendlovic
- Departamento de Microbiología y Parasitología, Facultad de Medicina, UNAM, Av. Universidad 3000, Col. Copilco-Universidad, Ciudad de Mexico, Mexico.,Facultad de Ciencias de la Salud, Universidad Anahuac Mexico Norte, Huixquilucan, Mexico
| |
Collapse
|
18
|
Cardoso MS, Santos RF, Almeida S, Sá M, Pérez-Cabezas B, Oliveira L, Tavares J, Carmo AM. Physical Interactions With Bacteria and Protozoan Parasites Establish the Scavenger Receptor SSC4D as a Broad-Spectrum Pattern Recognition Receptor. Front Immunol 2021; 12:760770. [PMID: 35003072 PMCID: PMC8739261 DOI: 10.3389/fimmu.2021.760770] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/29/2021] [Indexed: 12/19/2022] Open
Abstract
Since the pioneering discoveries, by the Nobel laureates Jules Hoffmann and Bruce Beutler, that Toll and Toll-like receptors can sense pathogenic microorganisms and initiate, in vertebrates and invertebrates, innate immune responses against microbial infections, many other families of pattern recognition receptors (PRRs) have been described. One of such receptor clusters is composed by, if not all, at least several members of the scavenger receptor cysteine-rich (SRCR) superfamily. Many SRCR proteins are plasma membrane receptors of immune cells; however, a small subset consists of secreted receptors that are therefore in circulation. We here describe the first characterization of biological and functional roles of the circulating human protein SSC4D, one of the least scrutinized members of the family. Within leukocyte populations, SSC4D was found to be expressed by monocytes/macrophages, neutrophils, and B cells, but its production was particularly evident in epithelial cells of several organs and tissues, namely, in the kidney, thyroid, lung, placenta, intestinal tract, and liver. Similar to other SRCR proteins, SSC4D shows the capacity of physically binding to different species of bacteria, and this opsonization can increase the phagocytic capacity of monocytes. Importantly, we have uncovered the capacity of SSC4D of binding to several protozoan parasites, a singular feature seldom described for PRRs in general and here demonstrated for the first time for an SRCR family member. Overall, our study is pioneer in assigning a PRR role to SSC4D.
Collapse
Affiliation(s)
- Marcos S. Cardoso
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Rita F. Santos
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Sarah Almeida
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
- Departamento de Biologia, Universidade de Aveiro, Aveiro, Portugal
| | - Mónica Sá
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
- Doutoramento em Ciências Farmacêuticas (especialidade Microbiologia), Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Begoña Pérez-Cabezas
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Liliana Oliveira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Joana Tavares
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Alexandre M. Carmo
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Porto, Portugal
| |
Collapse
|
19
|
Siggins MK, Sriskandan S. Bacterial Lymphatic Metastasis in Infection and Immunity. Cells 2021; 11:33. [PMID: 35011595 PMCID: PMC8750085 DOI: 10.3390/cells11010033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
Lymphatic vessels permeate tissues around the body, returning fluid from interstitial spaces back to the blood after passage through the lymph nodes, which are important sites for adaptive responses to all types of pathogens. Involvement of the lymphatics in the pathogenesis of bacterial infections is not well studied. Despite offering an obvious conduit for pathogen spread, the lymphatic system has long been regarded to bar the onward progression of most bacteria. There is little direct data on live virulent bacteria, instead understanding is largely inferred from studies investigating immune responses to viruses or antigens in lymph nodes. Recently, we have demonstrated that extracellular bacterial lymphatic metastasis of virulent strains of Streptococcus pyogenes drives systemic infection. Accordingly, it is timely to reconsider the role of lymph nodes as absolute barriers to bacterial dissemination in the lymphatics. Here, we summarise the routes and mechanisms by which an increasing variety of bacteria are acknowledged to transit through the lymphatic system, including those that do not necessarily require internalisation by host cells. We discuss the anatomy of the lymphatics and other factors that influence bacterial dissemination, as well as the consequences of underappreciated bacterial lymphatic metastasis on disease and immunity.
Collapse
Affiliation(s)
- Matthew K. Siggins
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
| | - Shiranee Sriskandan
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2DD, UK
| |
Collapse
|
20
|
Waaijer SJH, Suurs FV, Hau CS, Vrijland K, de Visser KE, de Groot DJA, de Vries EGE, Lub-de Hooge MN, Schröder CP. Radiolabeled Monoclonal Antibody Against Colony-Stimulating Factor 1 Receptor Specifically Distributes to the Spleen and Liver in Immunocompetent Mice. Front Oncol 2021; 11:786191. [PMID: 34976826 PMCID: PMC8716378 DOI: 10.3389/fonc.2021.786191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Abstract
Macrophages can promote tumor development. Preclinically, targeting macrophages by colony-stimulating factor 1 (CSF1)/CSF1 receptor (CSF1R) monoclonal antibodies (mAbs) enhances conventional therapeutics in combination treatments. The physiological distribution and tumor uptake of CSF1R mAbs are unknown. Therefore, we radiolabeled a murine CSF1R mAb and preclinically visualized its biodistribution by PET. CSF1R mAb was conjugated to N-succinyl-desferrioxamine (N-suc-DFO) and subsequently radiolabeled with zirconium-89 (89Zr). Optimal protein antibody dose was first determined in non-tumor-bearing mice to assess physiological distribution. Next, biodistribution of optimal protein dose and 89Zr-labeled isotype control was compared with PET and ex vivo biodistribution after 24 and 72 h in mammary tumor-bearing mice. Tissue autoradiography and immunohistochemistry determined radioactivity distribution and tissue macrophage presence, respectively. [89Zr]Zr-DFO-N-suc-CSF1R-mAb optimal protein dose was 10 mg/kg, with blood pool levels of 10 ± 2% injected dose per gram tissue (ID/g) and spleen and liver uptake of 17 ± 4 and 11 ± 4%ID/g at 72 h. In contrast, 0.4 mg/kg of [89Zr]Zr-DFO-N-suc-CSF1R mAb was eliminated from circulation within 24 h; spleen and liver uptake was 126 ± 44% and 34 ± 7%ID/g, respectively. Tumor-bearing mice showed higher uptake of [89Zr]Zr-DFO-N-suc-CSF1R-mAb in the liver, lymphoid tissues, duodenum, and ileum, but not in the tumor than did 89Zr-labeled control at 72 h. Immunohistochemistry and autoradiography showed that 89Zr was localized to macrophages within lymphoid tissues. Following [89Zr]Zr-DFO-N-suc-CSF1R-mAb administration, tumor macrophages were almost absent, whereas isotype-group tumors contained over 500 cells/mm2. We hypothesize that intratumoral macrophage depletion by [89Zr]Zr-DFO-N-suc-CSF1R-mAb precluded tumor uptake higher than 89Zr-labeled control. Translation of molecular imaging of macrophage-targeting therapeutics to humans may support macrophage-directed therapeutic development.
Collapse
Affiliation(s)
- Stijn J. H. Waaijer
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Frans V. Suurs
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Cheei-Sing Hau
- Division of Tumor Biology & Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Kim Vrijland
- Division of Tumor Biology & Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Karin E. de Visser
- Division of Tumor Biology & Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Derk Jan A. de Groot
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Elisabeth G. E. de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marjolijn N. Lub-de Hooge
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Carolina P. Schröder
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- *Correspondence: Carolina P. Schröder,
| |
Collapse
|
21
|
Scavenger receptor MARCO contributes to macrophage phagocytosis and clearance of tumor cells. Exp Cell Res 2021; 408:112862. [PMID: 34626585 DOI: 10.1016/j.yexcr.2021.112862] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 09/04/2021] [Accepted: 10/04/2021] [Indexed: 11/22/2022]
Abstract
Macrophage receptor with collagenous structure (MARCO) is a member of the class A scavenger receptor family which is expressed on the cell surface of macrophages. It is well known that MARCO avidly binds to unopsonized pathogens, leading to its ingestion by macrophages. However, the role of MARCO in the recognition and phagocytosis of tumor cells by macrophages remains poorly understood. In this study, we used lentiviral technology to knockdown and overexpress MARCO and investigated the ability of macrophages to phagocytose tumor cells. Our results showed that MARCO expression was correlated with the ability of macrophages to carry on phagocytosis. MARCO knockdown led to significant decreases in the number of engulfment pseudopodia of macrophages and inhibition of the phagocytosis of tumor cells. On the other hand, MARCO overexpression elevated activity of SYK, PI3K and Rac1 in macrophages, which led to changes in macrophage morphology and enhanced phagocytosis by promoting formation of stress fibers and pseudopodia. By Co-IP analysis we showed that MARCO directly binds to the β5 integrin of SL4 tumor cells. In summary, these results demonstrated the important role for MARCO in demonstrated tumor cells uptake and clearance by macrophages.
Collapse
|
22
|
Carpentier KS, Sheridan RM, Lucas CJ, Davenport BJ, Li FS, Lucas ED, McCarthy MK, Reynoso GV, May NA, Tamburini BAJ, Hesselberth JR, Hickman HD, Morrison TE. MARCO + lymphatic endothelial cells sequester arthritogenic alphaviruses to limit viremia and viral dissemination. EMBO J 2021; 40:e108966. [PMID: 34618370 PMCID: PMC8591538 DOI: 10.15252/embj.2021108966] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 02/02/2023] Open
Abstract
Viremia in the vertebrate host is a major determinant of arboviral reservoir competency, transmission efficiency, and disease severity. However, immune mechanisms that control arboviral viremia are poorly defined. Here, we identify critical roles for the scavenger receptor MARCO in controlling viremia during arthritogenic alphavirus infections in mice. Following subcutaneous inoculation, arthritogenic alphavirus particles drain via the lymph and are rapidly captured by MARCO+ lymphatic endothelial cells (LECs) in the draining lymph node (dLN), limiting viral spread to the bloodstream. Upon reaching the bloodstream, alphavirus particles are cleared from the circulation by MARCO-expressing Kupffer cells in the liver, limiting viremia and further viral dissemination. MARCO-mediated accumulation of alphavirus particles in the draining lymph node and liver is an important host defense mechanism as viremia and viral tissue burdens are elevated in MARCO-/- mice and disease is more severe. In contrast to prior studies implicating a key role for lymph node macrophages in limiting viral dissemination, these findings exemplify a previously unrecognized arbovirus-scavenging role for lymphatic endothelial cells and improve our mechanistic understanding of viremia control during arthritogenic alphavirus infection.
Collapse
Affiliation(s)
- Kathryn S Carpentier
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraCOUSA
| | - Ryan M Sheridan
- RNA Bioscience InitiativeUniversity of Colorado School of MedicineAuroraCOUSA
| | - Cormac J Lucas
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraCOUSA
| | - Bennett J Davenport
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraCOUSA
| | - Frances S Li
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraCOUSA
| | - Erin D Lucas
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraCOUSA
| | - Mary K McCarthy
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraCOUSA
| | - Glennys V Reynoso
- Viral Immunity and Pathogenesis UnitLaboratory of Clinical Microbiology and ImmunologyNational Institutes of Allergy and Infectious DiseasesNIHBethesdaMDUSA
| | - Nicholas A May
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraCOUSA
| | - Beth A J Tamburini
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraCOUSA
- Division of Gastroenterology and HepatologyDepartment of MedicineUniversity of Colorado Anschutz Medical Campus School of MedicineAuroraCOUSA
| | - Jay R Hesselberth
- RNA Bioscience InitiativeUniversity of Colorado School of MedicineAuroraCOUSA
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado School of MedicineAuroraCOUSA
| | - Heather D Hickman
- Viral Immunity and Pathogenesis UnitLaboratory of Clinical Microbiology and ImmunologyNational Institutes of Allergy and Infectious DiseasesNIHBethesdaMDUSA
| | - Thomas E Morrison
- Department of Immunology and MicrobiologyUniversity of Colorado School of MedicineAuroraCOUSA
| |
Collapse
|
23
|
Devanathan AS, Kashuba AD. Human Immunodeficiency Virus Persistence in the Spleen: Opportunities for Pharmacologic Intervention. AIDS Res Hum Retroviruses 2021; 37:725-735. [PMID: 33499746 DOI: 10.1089/aid.2020.0266] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The persistence of HIV in the spleen, despite combination antiretroviral therapy, is not well understood. Sustained immune dysregulation and delayed immune recovery, in addition to immune cell exhaustion, may contribute to persistence of infection in the spleen. Eliminating HIV from this secondary lymphoid organ will require a thorough understanding of antiretroviral (ARV) pharmacology in the spleen, which has been minimally investigated. Low ARV exposure within the spleen may hinder the achievement of a functional or sterilizing cure if cells are not protected from HIV infection. In this study, we provide an overview of the anatomy and physiology of the spleen, review the evidence of the spleen as a site for persistence of HIV, discuss the consequences of persistence of HIV in the spleen, address challenges to eradicating HIV in the spleen, and examine opportunities for future curative efforts.
Collapse
Affiliation(s)
| | - Angela D.M. Kashuba
- UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
- Division of Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
24
|
Transcriptomics Underlying Pulmonary Ozone Pathogenesis Regulated by Inflammatory Mediators in Mice. Antioxidants (Basel) 2021; 10:antiox10091489. [PMID: 34573120 PMCID: PMC8466999 DOI: 10.3390/antiox10091489] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 11/17/2022] Open
Abstract
Ozone (O3) is the predominant oxidant air pollutant associated with airway inflammation, lung dysfunction, and the worsening of preexisting respiratory diseases. We previously demonstrated the injurious roles of pulmonary immune receptors, tumor necrosis factor receptor (TNFR), and toll-like receptor 4, as well as a transcription factor NF-κB, in response to O3 in mice. In the current study, we profiled time-dependent and TNFR- and NF-κB-regulated lung transcriptome changes by subacute O3 to illuminate the underlying molecular events and downstream targets. Mice lacking Tnfr1/Tnfr2 (Tnfr-/-) or Nfkb1 (Nfkb1-/-) were exposed to air or O3. Lung RNAs were prepared for cDNA microarray analyses, and downstream and upstream mechanisms were predicted by pathway analyses of the enriched genes. O3 significantly altered the genes involved in inflammation and redox (24 h), cholesterol biosynthesis and vaso-occlusion (48 h), and cell cycle and DNA repair (48–72 h). Transforming growth factor-β1 was a predicted upstream regulator. Lack of Tnfr suppressed the immune cell proliferation and lipid-related processes and heightened epithelial cell integrity, and Nfkb1 deficiency markedly suppressed lung cell cycle progress during O3 exposure. Common differentially regulated genes by TNFR and NF-κB1 (e.g., Casp8, Il6, and Edn1) were predicted to protect the lungs from cell death, connective tissue injury, and inflammation. Il6-deficient mice were susceptible to O3-induced protein hyperpermeability, indicating its defensive role, while Tnf-deficient mice were resistant to overall lung injury caused by O3. The results elucidated transcriptome dynamics and provided new insights into the molecular mechanisms regulated by TNFR and NF-κB1 in pulmonary subacute O3 pathogenesis.
Collapse
|
25
|
Lukácsi S, Farkas Z, Saskői É, Bajtay Z, Takács-Vellai K. Conserved and Distinct Elements of Phagocytosis in Human and C. elegans. Int J Mol Sci 2021; 22:ijms22168934. [PMID: 34445642 PMCID: PMC8396242 DOI: 10.3390/ijms22168934] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Endocytosis provides the cellular nutrition and homeostasis of organisms, but pathogens often take advantage of this entry point to infect host cells. This is counteracted by phagocytosis that plays a key role in the protection against invading microbes both during the initial engulfment of pathogens and in the clearance of infected cells. Phagocytic cells balance two vital functions: preventing the accumulation of cell corpses to avoid pathological inflammation and autoimmunity, whilst maintaining host defence. In this review, we compare elements of phagocytosis in mammals and the nematode Caenorhabditis elegans. Initial recognition of infection requires different mechanisms. In mammals, pattern recognition receptors bind pathogens directly, whereas activation of the innate immune response in the nematode rather relies on the detection of cellular damage. In contrast, molecules involved in efferocytosis—the engulfment and elimination of dying cells and cell debris—are highly conserved between the two species. Therefore, C. elegans is a powerful model to research mechanisms of the phagocytic machinery. Finally, we show that both mammalian and worm studies help to understand how the two phagocytic functions are interconnected: emerging data suggest the activation of innate immunity as a consequence of defective apoptotic cell clearance.
Collapse
Affiliation(s)
- Szilvia Lukácsi
- MTA-ELTE Immunology Research Group, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (S.L.); (Z.B.)
| | - Zsolt Farkas
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (Z.F.); (É.S.)
| | - Éva Saskői
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (Z.F.); (É.S.)
| | - Zsuzsa Bajtay
- MTA-ELTE Immunology Research Group, Eötvös Loránd Research Network (ELKH), Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (S.L.); (Z.B.)
- Department of Immunology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary
| | - Krisztina Takács-Vellai
- Department of Biological Anthropology, Eötvös Loránd University, Pázmány Péter s. 1/C, 1117 Budapest, Hungary; (Z.F.); (É.S.)
- Correspondence:
| |
Collapse
|
26
|
Ding Y, Labitzky V, Legler K, Qi M, Schumacher U, Schmalfeldt B, Stürken C, Oliveira-Ferrer L. Molecular characteristics and tumorigenicity of ascites-derived tumor cells: mitochondrial oxidative phosphorylation as a novel therapy target in ovarian cancer. Mol Oncol 2021; 15:3578-3595. [PMID: 34060699 PMCID: PMC8637562 DOI: 10.1002/1878-0261.13028] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/16/2021] [Accepted: 05/28/2021] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer disseminates primarily intraperitoneally. Detached tumor cell aggregates (spheroids) from the primary tumor are regarded as ‘metastatic units’ that exhibit a low sensitivity to classical chemotherapy, probably due to their unique molecular characteristics. We have analyzed the cellular composition of ascites from OvCa patients, using flow cytometry, and studied their behavior in vitro and in vivo. We conclude that ascites‐derived cultured cells from OvCa patients give rise to two subpopulations: adherent cells and non‐adherent cells. Here, we found that the AD population includes mainly CD90+ cells with highly proliferative rates in vitro but no tumorigenic potential in vivo, whereas the NAD population contains principally tumor cell spheroids (EpCAM+/CD24+) with low proliferative potential in vitro. Enriched tumor cell spheroids from the ascites of high‐grade serous OvCA patients, obtained using cell strainers, were highly tumorigenic in vivo and their metastatic spread pattern precisely resembled the tumor dissemination pattern found in the corresponding patients. Comparative transcriptome analyses from ascites‐derived tumor cell spheroids (n = 10) versus tumor samples from different metastatic sites (n = 30) revealed upregulation of genes involved in chemoresistance (TGM1, HSPAs, MT1s), cell adhesion and cell‐barrier integrity (PKP3, CLDNs, PPL), and the oxidative phosphorylation process. Mitochondrial markers (mass and membrane potential) showed a reduced mitochondrial function in tumoroids from tumor tissue compared with ascites‐derived tumor spheroids in flow cytometry analysis. Interestingly, response to OXPHOS inhibition by metformin and IACS010759 in tumor spheroids correlated with the extent of mitochondrial membrane potential measured by fluorescence‐activated cell sorting. Our data contribute to a better understanding of the biology of ovarian cancer spheroids and identify the OXPHOS pathway as new potential treatment option in advanced ovarian cancer.
Collapse
Affiliation(s)
- Yi Ding
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Germany
| | - Vera Labitzky
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Germany
| | - Karen Legler
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Germany
| | - Minyue Qi
- Bioinformatic Core Facility, University Medical Center Hamburg-Eppendorf, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Germany
| | - Barbara Schmalfeldt
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Germany
| | - Christine Stürken
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Germany
| | | |
Collapse
|
27
|
Rasmussen RK, Etzerodt A. Therapeutic targeting of tumor-associated macrophages. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 91:185-211. [PMID: 34099108 DOI: 10.1016/bs.apha.2021.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Tumor-associated macrophages are among the most abundant non-cancerous cells in the tumor microenvironment and in many cancers macrophage infiltration into the tumor is associated with poor prognosis. Macrophages contribute to tumor development by promoting angiogenesis and immune suppression, and display remarkable phenotypic heterogeneity in the tumor microenvironment. Therapeutic strategies targeting macrophages that currently are in clinical development are mainly focused on a general depletion of tumor-associated macrophages, either by targeting the CSF-1/CSF-1R axis or by inhibiting macrophage recruitment by blocking CCR2/CCL2 signaling. Despite good pre-clinical response rates the treatment strategies focusing on general macrophage targeting have only shown limited clinical success and new approaches that target specific subsets of tumo-associated macrophages are emerging. This chapter will briefly present the functions and heterogeneity of tumor-associated macrophages and provide an overview of the current state of clinical development for pan-targeting strategies as well as discuss new strategies for targeting specific macrophage subsets for future anti-tumor immunotherapies.
Collapse
Affiliation(s)
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
28
|
Yirsaw AW, Gillespie A, Britton E, Doerle A, Johnson L, Marston S, Telfer J, Baldwin CL. Goat γδ T cell subpopulations defined by WC1 expression, responses to pathogens and cytokine production. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 118:103984. [PMID: 33352199 DOI: 10.1016/j.dci.2020.103984] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 06/12/2023]
Abstract
The major functions of γδ T cells in mammals overlap with those of αβ T cells but differ in that γδ T cells are rapid responders and see different types of antigens. While γδ T cells have been shown to be a major population of circulating lymphocytes in artiodactyl species such as cattle, sheep, and pigs, less is known about these cells in goats, an important agricultural species. We have recently shown that WC1, a γδ T cell-specific family of hybrid pattern recognition receptors/co-receptors, is a multigenic family in goats expanded beyond what occurs in cattle. This study was conducted to address some of the limitations of previous studies in determining the proportions of γδ T cells, WC1+ γδ T cells as well as the WC1.1+ and WC1.2+ subpopulations in blood and to evaluate their responses to various pathogens. Previously, the proportion of caprine γδ T cells was determined using a monoclonal antibody (mAb) 86D that we show here does not react with all γδ T cells thereby underestimating their contribution to the lymphocyte population. Using a mAb reactive with the TCRδ constant region we found the proportion of γδ T cells in blood was not significantly less than that of either CD4 or CD8 T cells and did not decrease with age after 6 months. γδ T cells that expressed WC1 ranged from ~20 to 85% of the total γδ T cells. Less than half of those were classified as WC1.1+ or WC1.2+ by mAb staining thus indicating a third major WC1+ population. We found that naïve γδ T cells proliferated in cultures of PBMC stimulated with antigens of Leptospira or Mycobacterium avium paratuberculosis (MAP) more than they did in control medium cultures or in those stimulated with M. bovis BCG antigens and that the responding γδ T cells included both WC1+ and WC1- cells. In ex vivo PMA/ionomycin-stimulated cultures of WC1- γδ T cells but not WC1+ cells produced both IL-17 and IFNγ. In longterm cultures with Leptospira or MAP both WC1- and WC1+ cells proliferated but only WC1- γδ T cells produced IL-17. In conclusion, goats have a substantial number of WC1- and WC1+ γδ T cells in PBMC that do not decrease with animal age after 6 months; both populations respond to bacterial antigens as naïve cells but in these cultures only the WC1- γδ cells produc IL-17 and IFNγ .
Collapse
Affiliation(s)
- Alehegne W Yirsaw
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Alexandria Gillespie
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Emily Britton
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Alyssa Doerle
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Lisa Johnson
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Susan Marston
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Janice Telfer
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA; Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, 01003, USA
| | - Cynthia L Baldwin
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA; Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
29
|
Fu YL, Harrison RE. Microbial Phagocytic Receptors and Their Potential Involvement in Cytokine Induction in Macrophages. Front Immunol 2021; 12:662063. [PMID: 33995386 PMCID: PMC8117099 DOI: 10.3389/fimmu.2021.662063] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/30/2021] [Indexed: 11/13/2022] Open
Abstract
Phagocytosis is an essential process for the uptake of large (>0.5 µm) particulate matter including microbes and dying cells. Specialized cells in the body perform phagocytosis which is enabled by cell surface receptors that recognize and bind target cells. Professional phagocytes play a prominent role in innate immunity and include macrophages, neutrophils and dendritic cells. These cells display a repertoire of phagocytic receptors that engage the target cells directly, or indirectly via opsonins, to mediate binding and internalization of the target into a phagosome. Phagosome maturation then proceeds to cause destruction and recycling of the phagosome contents. Key subsequent events include antigen presentation and cytokine production to alert and recruit cells involved in the adaptive immune response. Bridging the innate and adaptive immunity, macrophages secrete a broad selection of inflammatory mediators to orchestrate the type and magnitude of an inflammatory response. This review will focus on cytokines produced by NF-κB signaling which is activated by extracellular ligands and serves a master regulator of the inflammatory response to microbes. Macrophages secrete pro-inflammatory cytokines including TNFα, IL1β, IL6, IL8 and IL12 which together increases vascular permeability and promotes recruitment of other immune cells. The major anti-inflammatory cytokines produced by macrophages include IL10 and TGFβ which act to suppress inflammatory gene expression in macrophages and other immune cells. Typically, macrophage cytokines are synthesized, trafficked intracellularly and released in response to activation of pattern recognition receptors (PRRs) or inflammasomes. Direct evidence linking the event of phagocytosis to cytokine production in macrophages is lacking. This review will focus on cytokine output after engagement of macrophage phagocytic receptors by particulate microbial targets. Microbial receptors include the PRRs: Toll-like receptors (TLRs), scavenger receptors (SRs), C-type lectin and the opsonic receptors. Our current understanding of how macrophage receptor stimulation impacts cytokine production is largely based on work utilizing soluble ligands that are destined for endocytosis. We will instead focus this review on research examining receptor ligation during uptake of particulate microbes and how this complex internalization process may influence inflammatory cytokine production in macrophages.
Collapse
Affiliation(s)
- Yan Lin Fu
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, Canada
| | - Rene E. Harrison
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, Canada
| |
Collapse
|
30
|
In Sickness and in Health: The Immunological Roles of the Lymphatic System. Int J Mol Sci 2021; 22:ijms22094458. [PMID: 33923289 PMCID: PMC8123157 DOI: 10.3390/ijms22094458] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/15/2021] [Accepted: 04/18/2021] [Indexed: 02/06/2023] Open
Abstract
The lymphatic system plays crucial roles in immunity far beyond those of simply providing conduits for leukocytes and antigens in lymph fluid. Endothelial cells within this vasculature are distinct and highly specialized to perform roles based upon their location. Afferent lymphatic capillaries have unique intercellular junctions for efficient uptake of fluid and macromolecules, while expressing chemotactic and adhesion molecules that permit selective trafficking of specific immune cell subsets. Moreover, in response to events within peripheral tissue such as inflammation or infection, soluble factors from lymphatic endothelial cells exert “remote control” to modulate leukocyte migration across high endothelial venules from the blood to lymph nodes draining the tissue. These immune hubs are highly organized and perfectly arrayed to survey antigens from peripheral tissue while optimizing encounters between antigen-presenting cells and cognate lymphocytes. Furthermore, subsets of lymphatic endothelial cells exhibit differences in gene expression relating to specific functions and locality within the lymph node, facilitating both innate and acquired immune responses through antigen presentation, lymph node remodeling and regulation of leukocyte entry and exit. This review details the immune cell subsets in afferent and efferent lymph, and explores the mechanisms by which endothelial cells of the lymphatic system regulate such trafficking, for immune surveillance and tolerance during steady-state conditions, and in response to infection, acute and chronic inflammation, and subsequent resolution.
Collapse
|
31
|
Ma C, Feng K, Yang X, Yang Z, Wang Z, Shang Y, Fan G, Liu L, Yang S, Li X, Han J, Duan Y, Chen Y. Targeting macrophage liver X receptors by hydrogel-encapsulated T0901317 reduces atherosclerosis without effect on hepatic lipogenesis. Br J Pharmacol 2021; 178:1620-1638. [PMID: 33506494 DOI: 10.1111/bph.15387] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 01/11/2021] [Accepted: 01/11/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Targeting macrophage but not hepatocyte liver X receptors (LXRs) can reduce atherosclerosis without effect on hepatic lipogenesis. In this study, we encapsulated LXR ligands with D-Nap-GFFY to form a nanofibre hydrogel (D-Nap-GFFY-T0901317 or GFFY-T0901317) and determined its effect on atherosclerosis, hepatic lipogenesis and the underlying mechanisms involved. EXPERIMENTAL APPROACH D-Nap-GFFY-T0901317 was subcutaneously injected to proatherogenic diet-fed apoE-deficient (Apoe-/- ) mice, followed by determination of the development of atherosclerosis, liver steatosis and the involved mechanisms, with comparison of T0901317 oral administration. KEY RESULTS Subcutaneous injection of D-Nap-GFFY-T0901317 to Apoe-/- mice inhibited atherosclerosis at a comparable level as T0901317 oral administration without effect on hepatic lipogenesis. More importantly, D-Nap-GFFY-T0901317 regressed the advanced lesions. In arterial wall, D-Nap-GFFY-T0901317 reduced macrophage/foam cells, necrotic cores and calcification and increased collagen content. It activated expression of ABCA1/G1 and smooth muscle α-actin, while inhibiting expression of intracellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1). D-Nap-GFFY-T0901317 also reduced serum pro-inflammatory cytokines and facilitated Kupffer cell M2 polarization. Mechanistically, D-Nap-GFFY-T0901317 was selectively taken up by macrophages but not hepatocytes, resulting in activation of macrophage ABCA1/G1 expression, while having no effect on lipogenic genes in hepatocytes. Moreover, the selective uptake of D-Nap-GFFY-T0901317 by macrophages was mainly completed in a scavenger receptor class A-dependent manner. CONCLUSION AND IMPLICATIONS Our study demonstrates that D-Nap-GFFY-T0901317 reduces atherosclerosis without effect on hepatic lipogenesis by targeting macrophage LXRs selectively, indicating its potential application for atherosclerosis treatment.
Collapse
Affiliation(s)
- Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ke Feng
- Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Zhimou Yang
- Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhongyan Wang
- Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yuna Shang
- Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lipei Liu
- Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Shu Yang
- Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoju Li
- Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.,Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| |
Collapse
|
32
|
Salah A, Li Y, Wang H, Qi N, Wu Y. Macrophages as a Double-Edged Weapon: The Use of Macrophages in Cancer Immunotherapy and Understanding the Cross-Talk Between Macrophages and Cancer. DNA Cell Biol 2021; 40:429-440. [PMID: 33481665 DOI: 10.1089/dna.2020.6087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Macrophages (Mϕs) play an essential role in maintaining body homeostasis. They perform dual functions produced by different subtypes. Mϕs not only fight against pathogens and foreign bodies such as bacteria or cancer cells but also participate in healing and repairing damaged tissue since they maintain both proinflammatory and anti-inflammatory effects sequentially. Tumors possess the ability to polarize Mϕs from proinflammatory M1 subtype to anti-inflammatory M2-like Mϕs called tumor-associated macrophages, which, in turn, help the tumors to acquire cancer hallmarks. Consequently, this polarization allows tumors to grow and spread. In this light, Mϕs have been a subject of intense study, and researchers have developed protocols to derive different Mϕs subtypes either as a new state-of-the-art therapeutic approach or to understand the cross-talk between cancer and Mϕs. In this review, we present the use of primary Mϕs in adoptive immunotherapy for cancer, illustrate the reciprocating interplay between cancer and Mϕs, and the resulting structural and functional change on both cell types. Furthermore, we summarize the recent cutting-edge approaches of using Mϕs in cancer immunotherapy.
Collapse
Affiliation(s)
- Ahmed Salah
- Department of Biochemistry and Molecular Biology, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, P.R. China
| | - Yanqin Li
- Department of Biochemistry and Molecular Biology, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, P.R. China
| | - Hao Wang
- Hangzhou Biaomo Biosciences Co., Ltd., Hangzhou, P.R. China.,Asia Stem Cell Therapies Co., Limited, Shanghai, P.R. China
| | - Nianmin Qi
- Hangzhou Biaomo Biosciences Co., Ltd., Hangzhou, P.R. China.,Asia Stem Cell Therapies Co., Limited, Shanghai, P.R. China
| | - Yuehong Wu
- Department of Biochemistry and Molecular Biology, College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou, P.R. China
| |
Collapse
|
33
|
La Fleur L, Botling J, He F, Pelicano C, Zhou C, He C, Palano G, Mezheyeuski A, Micke P, Ravetch JV, Karlsson MCI, Sarhan D. Targeting MARCO and IL37R on Immunosuppressive Macrophages in Lung Cancer Blocks Regulatory T Cells and Supports Cytotoxic Lymphocyte Function. Cancer Res 2020; 81:956-967. [PMID: 33293426 DOI: 10.1158/0008-5472.can-20-1885] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/05/2020] [Accepted: 12/03/2020] [Indexed: 12/30/2022]
Abstract
The progression and metastatic capacity of solid tumors are strongly influenced by immune cells in the tumor microenvironment. In non-small cell lung cancer (NSCLC), accumulation of anti-inflammatory tumor-associated macrophages (TAM) is associated with worse clinical outcome and resistance to therapy. Here we investigated the immune landscape of NSCLC in the presence of protumoral TAMs expressing the macrophage receptor with collagenous structure (MARCO). MARCO-expressing TAM numbers correlated with increased occurrence of regulatory T cells and effector T cells and decreased natural killer (NK) cells in these tumors. Furthermore, transcriptomic data from the tumors uncovered a correlation between MARCO expression and the anti-inflammatory cytokine IL37. In vitro studies subsequently showed that lung cancer cells polarized macrophages to express MARCO and gain an immune-suppressive phenotype through the release of IL37. MARCO-expressing TAMs blocked cytotoxic T-cell and NK-cell activation, inhibiting their proliferation, cytokine production, and tumor killing capacity. Mechanistically, MARCO+ macrophages enhanced regulatory T (Treg) cell proliferation and IL10 production and diminished CD8 T-cell activities. Targeting MARCO or IL37 receptor (IL37R) by antibody or CRISPR knockout of IL37 in lung cancer cell lines repolarized TAMs, resulting in recovered cytolytic activity and antitumoral capacity of NK cells and T cells and downmodulated Treg cell activities. In summary, our data demonstrate a novel immune therapeutic approach targeting human TAMs immune suppression of NK- and T-cell antitumor activities. SIGNIFICANCE: This study defines tumor-derived IL37 and the macrophage scavenger receptor MARCO as potential therapeutic targets to remodel the immune-suppressive microenvironment in patients with lung cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/4/956/F1.large.jpg.
Collapse
Affiliation(s)
- Linnéa La Fleur
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Fei He
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Catarina Pelicano
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Chikai Zhou
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Chenfei He
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Giorgia Palano
- Department of Medicine, Karolinska University Hospital, Huddinge, Sweden
| | - Artur Mezheyeuski
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Dhifaf Sarhan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
34
|
Targeting a scavenger receptor on tumor-associated macrophages activates tumor cell killing by natural killer cells. Proc Natl Acad Sci U S A 2020; 117:32005-32016. [PMID: 33229588 DOI: 10.1073/pnas.2015343117] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tumor-associated macrophages (TAMs) can have protumor properties, including suppressing immune responses, promoting vascularization and, consequently, augmenting tumor progression. To stop TAM-mediated immunosuppression, we use a novel treatment by injecting antibodies specific for scavenger receptor MARCO, which is expressed on a specific subpopulation of TAMs in the tumor. We now report the location of this TAM as well as the pleiotropic mechanism of action of anti-MARCO antibody treatment on tumor progression and further show that this is potentially relevant to humans. Using specific targeting, we observed decreased tumor vascularization, a switch in the metabolic program of MARCO-expressing macrophages, and activation of natural killer (NK) cell killing through TNF-related apoptosis-inducing ligand (TRAIL). This latter activity reverses the effect of melanoma cell-conditioned macrophages in blocking NK activation and synergizes with T cell-directed immunotherapy, such as antibodies to PD-1 or PD-L1, to enhance tumor killing. Our study thus reveals an approach to targeting the immunosuppressive tumor microenvironment with monoclonal antibodies to enhance NK cell activation and NK cell-mediated killing. This can complement existing T cell-directed immunotherapy, providing a promising approach to combinatorial immunotherapy for cancer.
Collapse
|
35
|
Marinkovic D, Marinkovic T. Putative role of marginal zone B cells in pathophysiological processes. Scand J Immunol 2020; 92:e12920. [PMID: 32594535 DOI: 10.1111/sji.12920] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/21/2020] [Indexed: 12/16/2022]
Abstract
The maintenance of inner integrity of an organism is founded on the proper performance of two immunity branches, innate and adaptive immune responses. Recently, it became apparent that subset of splenic B cells named marginal zone B cells (MZB cells) exhibits unique developmental and functional features that bridge these two immunity branches. Strategically positioned at the site where blood and lymph are filtered, MZB cells represent a population of sentinels that rapidly proliferate and differentiate into IgM plasmablast cells when encountered with blood-borne, thymus-independent (TI) Ags. Moreover, MZB cells have intrinsic capability to induce potent CD4+ helper T cell response and cytokine production upon stimulation with soluble antigens. Due to their ability to overcome a time gap prior the establishment of the full adaptive response towards pathogens, MZB cells connect and direct innate and adaptive immunity. An additional interesting characteristic of MZB cells is capacity to function as regulatory cells in autoimmune processes. MZB cells may also contribute to the control of autoimmunity via the induction of tolerance by apoptotic cells. Importantly, in the clear association with inflammation and autoimmunity, MZB cells may transform into MALT lymphoma, representing a concurrence point for the infection, immunity and malignancy. This paper presents an insight into the complex biology of marginal zone B cells and their role in intertwining and directing innate and adaptive immune processes at the physiological and pathological level.
Collapse
Affiliation(s)
- Dragan Marinkovic
- Faculty of Special Education and Rehabilitation, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
36
|
Liu C, Zhang P, Zhang W. Immunological mechanism of IgG4-related disease. J Transl Autoimmun 2020; 3:100047. [PMID: 32743528 PMCID: PMC7388377 DOI: 10.1016/j.jtauto.2020.100047] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 03/09/2020] [Indexed: 12/23/2022] Open
Abstract
IgG4-related disease (IgG4-RD) is an immune-mediated inflammatory disorder in multiple organs, characterized by abundant infiltration of IgG4-positive plasmacytes and fibrosis in the involved organs. The precise pathogenic mechanism of IgG4-RD still remains unclear. Aberrant innate and adaptive immunity are considered as the main pathogenesis of IgG4-RD. Recent studies have shown that abnormal adaptive immune responses mediated by T helper type 2 cells, regulatory T lymphocytes, CD4+ cytotoxic T lymphocytes, T follicular helper cells, T follicular regulatory cells, PD-1hiCXCR5-peripheral T helper cells and B cell subsets are involved in IgG4-RD. In addition to adaptive immune responses, innate immune responses play pathogenic roles in IgG4-RD. Macrophages, mast cells, basophils, complement, and plasmacytoid dendritic cells are activated to produce various kinds of cytokines in IgG4-RD. This review aims to summarize the most recent knowledge in the pathogenesis of IgG4-RD.
Collapse
Affiliation(s)
- Changyan Liu
- Department of Rheumatology, The Second Hospital of Dalian Medical University, Dalian, 116023, China
- Department of Rheumatology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Panpan Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Wen Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, National Clinical Research Center for Dermatologic and Immunologic Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| |
Collapse
|
37
|
|
38
|
Li XC, Zhou J, Zhou JF, Wang Y, Ma H, Wang Y, Zhao S, Fang WH. SpBark Suppresses Bacterial Infection by Mediating Hemocyte Phagocytosis in an Invertebrate Model, Scylla paramamosain. Front Immunol 2019; 10:1992. [PMID: 31507600 PMCID: PMC6716108 DOI: 10.3389/fimmu.2019.01992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/06/2019] [Indexed: 11/13/2022] Open
Abstract
Scavenger receptors are cell surface membrane-bound receptors that typically bind multiple ligands and promote the removal of endogenous proteins and pathogens. In this study, we characterized a novel scavenger receptor-like protein, namely, SpBark. SpBark was upregulated in hemocytes after challenges with bacteria, suggesting that it might be involved in antibacterial defense. SpBark is a type I transmembrane protein with four extracellular domains, including three scavenger receptor cysteine-rich domains (SRCRDs) and a C-type lectin domain (CTLD). Western blot assay showed that SpBark CTLD possessed a much stronger binding activity to tested microbes than the three SRCRDs. It also exhibited apparent binding activities to lipopolysaccharide (LPS) and acetylated low-density lipoprotein (ac-LDL), whereas the other SRCRDs showed much lower or no binding activities to these components. Agglutination activities were observed in the presence of Ca2+ by incubating microorganisms with SpBark CTLD instead of SRCRDs. These results suggested that SpBark CTLD was the major binding site for ac-LDL and LPS. Coating Vibrio parahemolyticus with SpBark CTLD promoted bacterial clearance in vivo. This finding indicated that SpBark might participate in the immune defenses against Gram-negative bacteria through a certain mechanism. The promotion of bacterial clearance by SpBark was further determined using SpBark-silenced crabs injected with V. parahemolyticus. SpBark knockdown by injection of SpBark dsRNA remarkably suppressed the clearance of bacteria in hemolymph. Meanwhile, it also severely restrained the phagocytosis of bacteria. This finding suggested that SpBark could modulate the phagocytosis of bacteria, and the promotion of bacterial clearance by SpBark was closely related to SpBark-mediated phagocytosis activity. The likely mechanism of bacterial clearance mediated by SpBark was as follows: SpBark acted as a pattern recognition receptor, which could sense and bind to LPS on the surface of invading bacteria with its CTLD in hemolymph. The binding to LPS made the bacteria adhere to the surface of hemocytes. This process would facilitate phagocytosis of the bacteria, resulting in their removal. This study provided new insights into the hemocyte phagocytosis mechanisms of invertebrates and the multiple biological functions of Bark proteins.
Collapse
Affiliation(s)
- Xin-Cang Li
- Key Laboratory of East China Sea Fishery Resources Exploitation, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Jian Zhou
- Key Laboratory of East China Sea Fishery Resources Exploitation, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Jun-Fang Zhou
- Key Laboratory of East China Sea Fishery Resources Exploitation, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Yue Wang
- Key Laboratory of East China Sea Fishery Resources Exploitation, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China.,Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Hongyu Ma
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, China
| | - Yuan Wang
- Key Laboratory of East China Sea Fishery Resources Exploitation, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Shu Zhao
- Key Laboratory of East China Sea Fishery Resources Exploitation, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Wen-Hong Fang
- Key Laboratory of East China Sea Fishery Resources Exploitation, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| |
Collapse
|
39
|
Dominant role of splenic marginal zone lipid rafts in the classical complement pathway against S. pneumoniae. Cell Death Discov 2019; 5:133. [PMID: 31531231 PMCID: PMC6733876 DOI: 10.1038/s41420-019-0213-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/11/2019] [Accepted: 08/18/2019] [Indexed: 12/28/2022] Open
Abstract
Lipid rafts (LRs) play crucial roles in complex physiological processes, modulating innate and acquired immune responses to pathogens. The transmembrane C-type lectins human dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN) and its mouse homolog SIGN-R1 are distributed in LRs and expressed on splenic marginal zone (MZ) macrophages. The DC-SIGN-C1q or SIGN-R1-C1q complex could mediate the immunoglobulin (Ig)-independent classical complement pathway against Streptococcus pneumoniae. Precise roles of LRs during this complement pathway are unknown. Here we show that LRs are indispensable for accelerating the DC-SIGN- or SIGN-R1-mediated classical complement pathway against S. pneumoniae, thus facilitating rapid clearance of the pathogen. The trimolecular complex of SIGN-R1-C1q-C4 was exclusively enriched in LRs of splenic MZ macrophages and their localization was essential for activating C3 catabolism and enhancing pneumococcal clearance, which were abolished in SIGN-R1-knockout mice. However, DC-SIGN replacement on splenic MZ macrophage’s LRs of SIGN-R1-depleted mice reversed these defects. Disruption of LRs dramatically reduced pneumococcal uptake and decomposition. Additionally, DC- SIGN, C1q, C4, and C3 were obviously distributed in splenic LRs of cadavers. Therefore, LRs on splenic SIGN-R1+ or DC-SIGN+ macrophages could provide spatially confined and optimal bidirectional platforms, not only for usual intracellular events, for example recognition and phagocytosis of pathogens, but also an unusual extracellular event such as the complement system. These findings improve our understanding of the orchestrated roles of the spleen, unraveling a new innate immune system initiated from splenic MZ LRs, and yielding answers to several long-standing problems, including the need to understand the profound role of LRs in innate immunity, the need to identify how such a small portion of splenic SIGN-R1+ macrophages (<0.05% of splenic macrophages) effectively resist S. pneumoniae, and the need to understand how LRs can promote the protective function of DC-SIGN against S. pneumoniae in the human spleen.
Collapse
|
40
|
Leigh ND, Dunlap GS, Johnson K, Mariano R, Oshiro R, Wong AY, Bryant DM, Miller BM, Ratner A, Chen A, Ye WW, Haas BJ, Whited JL. Transcriptomic landscape of the blastema niche in regenerating adult axolotl limbs at single-cell resolution. Nat Commun 2018; 9:5153. [PMID: 30514844 PMCID: PMC6279788 DOI: 10.1038/s41467-018-07604-0] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 11/13/2018] [Indexed: 12/21/2022] Open
Abstract
Regeneration of complex multi-tissue structures, such as limbs, requires the coordinated effort of multiple cell types. In axolotl limb regeneration, the wound epidermis and blastema have been extensively studied via histology, grafting, and bulk-tissue RNA-sequencing. However, defining the contributions of these tissues is hindered due to limited information regarding the molecular identity of the cell types in regenerating limbs. Here we report unbiased single-cell RNA-sequencing on over 25,000 cells from axolotl limbs and identify a plethora of cellular diversity within epidermal, mesenchymal, and hematopoietic lineages in homeostatic and regenerating limbs. We identify regeneration-induced genes, develop putative trajectories for blastema cell differentiation, and propose the molecular identity of fibroblast-like blastema progenitor cells. This work will enable application of molecular techniques to assess the contribution of these populations to limb regeneration. Overall, these data allow for establishment of a putative framework for adult axolotl limb regeneration. Limb regeneration requires a blastema with progenitor cells, immune cells, and an overlying wound epidermis, but molecular identities of these populations are unclear. Here, the authors use single-cell RNA-sequencing to identify transcriptionally distinct cell populations in adult axolotl limb blastemas.
Collapse
Affiliation(s)
- Nicholas D Leigh
- Department of Orthopedic Surgery, Harvard Medical School, The Harvard Stem Cell Institute, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
| | - Garrett S Dunlap
- Department of Orthopedic Surgery, Harvard Medical School, The Harvard Stem Cell Institute, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
| | - Kimberly Johnson
- Department of Orthopedic Surgery, Harvard Medical School, The Harvard Stem Cell Institute, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
| | - Rachelle Mariano
- Department of Orthopedic Surgery, Harvard Medical School, The Harvard Stem Cell Institute, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Rachel Oshiro
- Department of Orthopedic Surgery, Harvard Medical School, The Harvard Stem Cell Institute, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Alan Y Wong
- Department of Orthopedic Surgery, Harvard Medical School, The Harvard Stem Cell Institute, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA, 02138, USA
| | - Donald M Bryant
- Department of Orthopedic Surgery, Harvard Medical School, The Harvard Stem Cell Institute, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Bess M Miller
- Department of Orthopedic Surgery, Harvard Medical School, The Harvard Stem Cell Institute, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA.,Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
| | - Alex Ratner
- ICCB-L Single Cell Core, Harvard Medical School, 200 Longwood Avenue, Boston, MA, 02115, USA
| | - Andy Chen
- Department of Orthopedic Surgery, Harvard Medical School, The Harvard Stem Cell Institute, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - William W Ye
- Department of Orthopedic Surgery, Harvard Medical School, The Harvard Stem Cell Institute, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Brian J Haas
- Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA
| | - Jessica L Whited
- Department of Orthopedic Surgery, Harvard Medical School, The Harvard Stem Cell Institute, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA. .,Broad Institute of MIT and Harvard, 7 Cambridge Center, Cambridge, MA, 02142, USA. .,Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA, 02138, USA.
| |
Collapse
|
41
|
Nakayama H, Nagafuku M, Suzuki A, Iwabuchi K, Inokuchi JI. The regulatory roles of glycosphingolipid-enriched lipid rafts in immune systems. FEBS Lett 2018; 592:3921-3942. [PMID: 30320884 DOI: 10.1002/1873-3468.13275] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 01/04/2023]
Abstract
Lipid rafts formed by glycosphingolipids (GSLs) on cellular membranes play important roles in innate and adaptive immunity. Lactosylceramide (LacCer) forms lipid rafts on plasma and granular membranes of human neutrophils. These LacCer-enriched lipid rafts bind directly to pathogenic components, such as pathogenic fungi-derived β-glucan and Mycobacteria-derived lipoarabinomannan via carbohydrate-carbohydrate interactions, and mediate innate immune responses to these pathogens. In contrast, a-series and o-series gangliosides form distinct rafts on CD4+ and CD8+ T cell subsets, respectively, contributing to the respective functions of these cells and stimulating adaptive immune responses through T cell receptors. These findings suggest that gangliosides play indispensable roles in T cell selection and activation. This Review introduces the involvement of GSL-enriched lipid rafts in innate and adaptive immunity.
Collapse
Affiliation(s)
- Hitoshi Nakayama
- Laboratory of Biochemistry, Juntendo University Faculty of Health Care and Nursing, Urayasu, Japan.,Institute for Environmental and Gender-specific Medicine, Juntendo University Graduate School of Medicine, Urayasu, Japan
| | - Masakazu Nagafuku
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Akemi Suzuki
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kazuhisa Iwabuchi
- Laboratory of Biochemistry, Juntendo University Faculty of Health Care and Nursing, Urayasu, Japan.,Institute for Environmental and Gender-specific Medicine, Juntendo University Graduate School of Medicine, Urayasu, Japan.,Infection Control Nursing, Juntendo University Graduate School of Health Care and Nursing, Urayasu, Japan
| | - Jin-Ichi Inokuchi
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
42
|
Abstract
Infiltration of macrophages in solid tumours is associated with poor prognosis and correlates with chemotherapy resistance in most cancers. In mouse models of cancer, macrophages promote cancer initiation and malignant progression by stimulating angiogenesis, increasing tumour cell migration, invasion and intravasation and suppressing antitumour immunity. At metastatic sites, macrophages promote tumour cell extravasation, survival and subsequent growth. Each of these pro-tumoural activities is promoted by a subpopulation of macrophages that express canonical markers but have unique transcriptional profiles, which makes tumour-associated macrophages (TAMs) good targets for anticancer therapy in humans through either their ablation or their re-differentiation away from pro-tumoural towards antitumoural states. In this Review, we evaluate the state of the art of TAM-targeting strategies, focusing on the limitations and potential side effects of the different therapies such as toxicity, rebound effects and compensatory mechanisms. We provide an extensive overview of the different types of therapy used in the clinic and their limitations in light of known macrophage biology and propose new strategies for targeting TAMs.
Collapse
|
43
|
Pombinho R, Sousa S, Cabanes D. Scavenger Receptors: Promiscuous Players during Microbial Pathogenesis. Crit Rev Microbiol 2018; 44:685-700. [PMID: 30318962 DOI: 10.1080/1040841x.2018.1493716] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Innate immunity is the most broadly effective host defense, being essential to clear the majority of microbial infections. Scavenger Receptors comprise a family of sensors expressed in a multitude of host cells, whose dual role during microbial pathogenesis gained importance over recent years. SRs regulate the recruitment of immune cells and control both host inflammatory response and bacterial load. In turn, pathogens have evolved different strategies to overcome immune response, avoid recognition by SRs and exploit them to favor infection. Here, we discuss the most relevant findings regarding the interplay between SRs and pathogens, discussing how these multifunctional proteins recognize a panoply of ligands and act as bacterial phagocytic receptors.
Collapse
Affiliation(s)
- Rita Pombinho
- a Instituto de Investigação e Inovação em Saúde (i3S), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal.,b Instituto de Biologia Molecular e Celular (IBMC), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal
| | - Sandra Sousa
- a Instituto de Investigação e Inovação em Saúde (i3S), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal.,b Instituto de Biologia Molecular e Celular (IBMC), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal
| | - Didier Cabanes
- a Instituto de Investigação e Inovação em Saúde (i3S), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal.,b Instituto de Biologia Molecular e Celular (IBMC), Group of Molecular Microbiology , Universidade do Porto , Porto , Portugal
| |
Collapse
|
44
|
T'Jonck W, Guilliams M, Bonnardel J. Niche signals and transcription factors involved in tissue-resident macrophage development. Cell Immunol 2018; 330:43-53. [PMID: 29463401 PMCID: PMC6108424 DOI: 10.1016/j.cellimm.2018.02.005] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/07/2018] [Accepted: 02/10/2018] [Indexed: 12/25/2022]
Abstract
Tissue-resident macrophages form an essential part of the first line of defense in all tissues of the body. Next to their immunological role, they play an important role in maintaining tissue homeostasis. Recently, it was shown that they are primarily of embryonic origin. During embryogenesis, precursors originating in the yolk sac and fetal liver colonize the embryonal tissues where they develop into mature tissue-resident macrophages. Their development is governed by two distinct sets of transcription factors. First, in the pre-macrophage stage, a core macrophage program is established by lineage-determining transcription factors. Under the influence of tissue-specific signals, this core program is refined by signal-dependent transcription factors. This nurturing by the niche allows the macrophages to perform tissue-specific functions. In the last 15 years, some of these niche signals and transcription factors have been identified. However, detailed insight in the exact mechanism of development is still lacking.
Collapse
Affiliation(s)
- Wouter T'Jonck
- Laboratory of Myeloid Cell Ontogeny and Functional Specialization, VIB-UGent Center for Inflammation Research, Technologiepark 927, 9052 Gent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Gent, Belgium.
| | - Martin Guilliams
- Laboratory of Myeloid Cell Ontogeny and Functional Specialization, VIB-UGent Center for Inflammation Research, Technologiepark 927, 9052 Gent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Gent, Belgium
| | - Johnny Bonnardel
- Laboratory of Myeloid Cell Ontogeny and Functional Specialization, VIB-UGent Center for Inflammation Research, Technologiepark 927, 9052 Gent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Gent, Belgium.
| |
Collapse
|
45
|
Entry of Herpes Simplex Virus 1 into Epidermis and Dermal Fibroblasts Is Independent of the Scavenger Receptor MARCO. J Virol 2018; 92:JVI.00490-18. [PMID: 29769337 DOI: 10.1128/jvi.00490-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/10/2018] [Indexed: 11/20/2022] Open
Abstract
To enter host cells, herpes simplex virus 1 (HSV-1) initially attaches to cell surface glycosaminoglycans, followed by the requisite binding to one of several cellular receptors, leading to viral internalization. Although virus-receptor interactions have been studied in various cell lines, the contributions of individual receptors to uptake into target tissues such as mucosa, skin, and cornea are not well understood. We demonstrated that nectin-1 acts as a major receptor for HSV-1 entry into murine epidermis, while herpesvirus entry mediator (HVEM) can serve as an alternative receptor. Recently, the macrophage receptor with collagenous structure (MARCO) has been described to mediate adsorption of HSV-1 to epithelial cells. Here, we investigated the impact of MARCO on the entry process of HSV-1 into the two major cell types of skin, keratinocytes in the epidermis and fibroblasts in the underlying dermis. Using ex vivo infection of murine epidermis, we showed that HSV-1 entered basal keratinocytes of MARCO-/- epidermis as efficiently as those of control epidermis. In addition, entry into dermal fibroblasts was not impaired in the absence of MARCO. When we treated epidermis, primary keratinocytes, or fibroblasts with poly(I), a ligand for class A scavenger receptors, HSV-1 entry was strongly reduced. As we also observed reducing effects of poly(I) in the absence of both MARCO and scavenger receptor A1, we concluded that the inhibitory effects of poly(I) on HSV-1 infection are not directly linked to class A scavenger receptors. Overall, our results support that HSV-1 entry into skin cells is independent of MARCO.IMPORTANCE During entry into its host cells, the human pathogen herpes simplex virus (HSV) interacts with various cellular receptors. Initially, receptor interaction can mediate cellular adsorption, followed by receptor binding that triggers viral internalization. The intriguing question is which receptors are responsible for the various steps during entry into the natural target tissues of HSV? Previously, we demonstrated the role of nectin-1 as a major receptor and that of HVEM as an alternative receptor for HSV-1 to invade murine epidermis. As MARCO has been described to promote infection in skin, we explored the predicted role of MARCO as a receptor that mediates adsorption to epithelial cells. Our infection studies of murine skin cells indicate that the absence of MARCO does not interfere with the efficiency of HSV-1 entry and that the inhibitory effect on viral adsorption by poly(I), a ligand of MARCO, is independent of MARCO.
Collapse
|
46
|
Lai JD, Cartier D, Hartholt RB, Swystun LL, van Velzen AS, den Haan JMM, Hough C, Voorberg J, Lillicrap D. Early cellular interactions and immune transcriptome profiles in human factor VIII-exposed hemophilia A mice. J Thromb Haemost 2018; 16:533-545. [PMID: 29285874 DOI: 10.1111/jth.13936] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Indexed: 12/16/2022]
Abstract
Essentials Initial immune cell interactions leading to factor (F) VIII immunity are not well characterized. We assessed cellular interactions and expression profiles in hemophilia A mice. MARCO+, followed by SIGLEC1+ and SIGNR1+ macrophages co-localize most with human FVIII. The splenic transcriptome highlights potential therapeutic targets to prevent inhibitors. SUMMARY Background Developing factor VIII (FVIII) inhibitory antibodies is the most serious complication in hemophilia A treatment, representing a significant health and economic burden. A better understanding of the early events in an immune response leading to this outcome may provide insight into inhibitor development. Objective To identify early mediators of FVIII immunity and to detail immune expression profiles in the spleen and liver. Methods C57Bl/6 F8 E16 knockout mice were infused with 5-20 μg (2000-8000 IU kg-1 ) of recombinant FVIII. Spleens were frozen at various time-points post-infusion and stained for FVIII and cellular markers. Splenic and liver RNA expression analysis was performed 3 h post-infusion of 0.6 μg (240 IU kg-1 ) FVIII by nCounter technology using a 561-gene immunology panel. Results FVIII localization in the spleen did not change over 2.5 h. We observed significantly higher co-localization of FVIII with MARCO+ cells compared with SIGLEC1+ and SIGNR1+ in the splenic marginal zone. FVIII exhibited little co-localization with CD11c+ dendritic cells and the macrophage mannose receptor, CD206. Following FVIII infusion, the splenic mRNA profiling identified genes such as Tnfaip6 and Il23r, which are implicated in chemotaxis and a proinflammatory Th17 response, respectively. In contrast, an upregulation of Gfi1 in the liver suggests an anti-inflammatory environment. Conclusions FVIII co-localizes predominantly with marginal zone macrophages (MARCO+ ) in the murine spleen following intravenous infusion. Targeting pathways that are implicated in the early FVIII innate immune response in the spleen may lead to therapeutic interventions to prevent inhibitor formation.
Collapse
Affiliation(s)
- J D Lai
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - D Cartier
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - R B Hartholt
- Department of Plasma Proteins, Sanquin-Academic Medical Center Landsteiner Laboratory, Amsterdam, the Netherlands
| | - L L Swystun
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - A S van Velzen
- Pediatrics, Hematology, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - J M M den Haan
- Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, the Netherlands
| | - C Hough
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - J Voorberg
- Department of Plasma Proteins, Sanquin-Academic Medical Center Landsteiner Laboratory, Amsterdam, the Netherlands
| | - D Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
47
|
Zhang L, Nie L, Cai SY, Chen J, Chen J. Role of a macrophage receptor with collagenous structure (MARCO) in regulating monocyte/macrophage functions in ayu, Plecoglossus altivelis. FISH & SHELLFISH IMMUNOLOGY 2018; 74:141-151. [PMID: 29305330 DOI: 10.1016/j.fsi.2017.12.059] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/23/2017] [Accepted: 12/28/2017] [Indexed: 06/07/2023]
Abstract
Macrophage receptor with collagenous structure (MARCO) plays essential roles in phagocytic cell-mediated innate immune responses. However, studies regarding MARCO, especially its functions, are limited in teleost species. In this study, we identified a MARCO molecule (PaMARCO) from ayu (Plecoglossus altivelis). PaMARCO shared conserved functional domains with its mammalian counterparts. Sequence analysis showed that PaMARCO was most closely related to its rainbow trout (Oncorhynchus mykiss) counterpart. PaMARCO expression was upregulated in all tested immune tissues and monocytes/macrophages (MO/MΦ) upon Vibrio anguillarum infection, and blocking its function significantly decreased the immune responses of MO/MΦ during infection. PaMARCO could bind to the tested gram-positive and -negative bacteria in a Ca2+-dependent manner in vitro. Furthermore, the phagocytosis and bacterial killing activities of MO/MΦ were significantly decreased upon PaMARCO blockade using anti-PaMARCO IgG. PaMARCO was also involved in the polarization processes of ayu MO/MΦ. The upregulated expression of representative cytokines in LPS-induced M1 type (TNF-α, IL-1β) or cAMP-induced M2 type (TGF-β, IL-10) were inhibited in the anti-PaMARCO IgG-treated group, indicating that PaMARCO may be involved in the regulation of both inflammation priming and inflammation resolution of MO/MΦ. In conclusion, our results implicate that PaMARCO has essential regulatory roles for bacterial binding, clearance, and the polarization processes of ayu MO/MΦ.
Collapse
Affiliation(s)
- Le Zhang
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Li Nie
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Shi-Yu Cai
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Jie Chen
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, China
| | - Jiong Chen
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
48
|
Li SS, Ogbomo H, Mansour MK, Xiang RF, Szabo L, Munro F, Mukherjee P, Mariuzza RA, Amrein M, Vyas JM, Robbins SM, Mody CH. Identification of the fungal ligand triggering cytotoxic PRR-mediated NK cell killing of Cryptococcus and Candida. Nat Commun 2018; 9:751. [PMID: 29467448 PMCID: PMC5821813 DOI: 10.1038/s41467-018-03014-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 01/11/2018] [Indexed: 01/08/2023] Open
Abstract
Natural killer (NK) cells use the activating receptor NKp30 as a microbial pattern-recognition receptor to recognize, activate cytolytic pathways, and directly kill the fungi Cryptococcus neoformans and Candida albicans. However, the fungal pathogen-associated molecular pattern (PAMP) that triggers NKp30-mediated killing remains to be identified. Here we show that β-1,3-glucan, a component of the fungal cell wall, binds to NKp30. We further demonstrate that β-1,3-glucan stimulates granule convergence and polarization, as shown by live cell imaging. Through Src Family Kinase signaling, β-1,3-glucan increases expression and clustering of NKp30 at the microbial and NK cell synapse to induce perforin release for fungal cytotoxicity. Rather than blocking the interaction between fungi and NK cells, soluble β-1,3-glucan enhances fungal killing and restores defective cryptococcal killing by NK cells from HIV-positive individuals, implicating β-1,3-glucan to be both an activating ligand and a soluble PAMP that shapes NK cell host immunity. Natural killer (NK) cells has been show to mediate fungi killing via the activating receptor NKp30, but the fungal target for NKp30 is still unclear. Here the authors show, using atomic force microscopy and live cell imaging, that β-1,3-glucan is expressed by Cryptococcus neoformans and Candida albicans and responsible for NKp30-mediated NK killing.
Collapse
Affiliation(s)
- Shu Shun Li
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, T2N 4N1, Canada.,The Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, T2N 4N1, Canada
| | - Henry Ogbomo
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, T2N 4N1, Canada.,The Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, T2N 4N1, Canada
| | - Michael K Mansour
- Department of Medicine Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Richard F Xiang
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, T2N 4N1, Canada.,The Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, T2N 4N1, Canada
| | - Lian Szabo
- Department of Medicine, University of Calgary, Calgary, T2N 4N1, Canada
| | - Fay Munro
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, T2N 4N1, Canada
| | - Priyanka Mukherjee
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, T2N 4N1, Canada
| | - Roy A Mariuzza
- Department of Cell Biology & Molecular Genetics, University of Maryland, College Park, MD, 20742, USA
| | - Matthias Amrein
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, T2N 4N1, Canada
| | - Jatin M Vyas
- Department of Medicine Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Stephen M Robbins
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, T2N 4N1, Canada.,Southern Alberta Cancer Research Institute, University of Calgary, Calgary, T2N 4N1, Canada
| | - Christopher H Mody
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, T2N 4N1, Canada. .,The Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, T2N 4N1, Canada. .,Department of Medicine, University of Calgary, Calgary, T2N 4N1, Canada.
| |
Collapse
|
49
|
Nakagaki BN, Vieira AT, Rezende RM, David BA, Menezes GB. Tissue macrophages as mediators of a healthy relationship with gut commensal microbiota. Cell Immunol 2018; 330:16-26. [PMID: 29422270 DOI: 10.1016/j.cellimm.2018.01.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/26/2018] [Accepted: 01/31/2018] [Indexed: 12/12/2022]
Abstract
Mammals and microorganisms have evolved a complex and tightly controlled mutual relationship. This interaction grants protection and energy source for the microorganisms, and on the other hand, provides several immunologic, metabolic and physiological advantages for the host. The gastrointestinal tract (GI) harbors the largest bacteria diversity within the body and complex mechanisms control microbiota community under homeostasis. However, once disrupted, microbiota imbalance can lead to overt growth of resident and invasive populations, with potential risk for lethal diseases. In these cases, bacteria might also escape from the intestines and reach different organs through the blood and lymphatic circulation. To control these unwanted conditions, all body tissues are populated with resident macrophages that have the ability to capture and eliminate pathogens, avoiding their dissemination. Here we discuss the different routes for bacterial translocation from the intestinal tract, and how macrophages act in the removal of these microorganisms to prevent systemic infections and restore the homeostasis.
Collapse
Affiliation(s)
- Brenda Naemi Nakagaki
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | - Angélica Thomaz Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | - Rafael Machado Rezende
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bruna Araujo David
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, University of Calgary. Calgary, Alberta, Canada.
| | - Gustavo Batista Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil.
| |
Collapse
|
50
|
Iwabuchi K. Gangliosides in the Immune System: Role of Glycosphingolipids and Glycosphingolipid-Enriched Lipid Rafts in Immunological Functions. Methods Mol Biol 2018; 1804:83-95. [PMID: 29926405 DOI: 10.1007/978-1-4939-8552-4_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Although individuals are constantly exposed to infectious agents, these agents are generally resisted by the innate and acquired immune systems. Both the innate and acquired immune systems protect against invading organisms, but they differ functionally in several ways. The innate immune system is the body's inborn defense mechanism and the first line of defense against invading organisms, such as bacteria, fungi, and viruses. Glycosphingolipids (GSLs), which are expressed on the outer leaflet of plasma membranes (Murate et al., J Cell Sci 128(8):1627-1638, 2015), are involved in both innate and acquired immunity (Inokuchi et al., Biochim Biophys Acta 1851(1):98-106, 2015; Nakayama et al., Arch Immunol Ther Exp (Warsz) 61(3):217-228, 2013; Rueda, Br J Nutr 98(Suppl 1):S68-73, 2007; Popa and Portoukalian, Pathol Biol (Paris) 51(5):253-255, 2003).Recent studies have indicated that innate immunity is not a "nonspecific" immune system. Large numbers of viruses, bacteria, and bacterial toxins have been reported to bind to host surface carbohydrates, a number of which are components of GSLs (Schengrund, Biochem Pharmacol 65(5):699-707, 2003). Binding studies have also demonstrated that some glycolipids function as receptors for microorganisms and bacterial toxins (Yates and Rampersaud, Ann N Y Acad Sci 845:57-71, 1998). These findings clearly indicate that GSLs are involved in host-pathogen interactions.GSLs are composed of hydrophobic ceramide and hydrophilic sugar moieties (Hakomori, Annu Rev Biochem 50:733-764, 1980). The ceramide moiety of sphingolipids and the cholesterol sterol-ring system are thought to interact via hydrogen bonds and hydrophobic van der Waal's forces (Mukherjee and Maxfield, Annu Rev Cell Dev Biol 20:839-866, 2004). Additional hydrophilic cis interactions among GSL headgroups have been found to promote their lateral associations with surrounding lipid and protein membrane components. These interactions result in the separation in cell membranes of lipid rafts, which are lipid domains rich in GSLs, cholesterol, glycosylphosphatidylinositol (GPI)-anchored proteins and membrane-anchored signaling molecules (Pike, J Lipid Res 47(7):1597-1598, 2006). These GSL-enriched lipid rafts play important roles in immunological functions (Inokuchi et al., Biochim Biophys Acta 1851(1):98-106, 2015; Iwabuchi et al., Mediators Inflamm 2015:120748, 2015; Anderson and Roche, Biochim Biophys Acta 1853(4):775-780, 2015; Zuidscherwoude et al., J Leukoc Biol 95(2):251-263, 2014; Dykstra et al., Annu Rev Immunol 21:457-481, 2003). This introductory chapter describes the roles of GSLs and their lipid rafts in the immune system.
Collapse
Affiliation(s)
- Kazuhisa Iwabuchi
- Infection Control Nursing, Graduate School of Health Care and Nursing, Juntendo University, Chiba, Japan.
- Institute for Environmental and Gender Specific Medicine, Graduate school of Medicine, Juntendo University, Chiba, Japan.
| |
Collapse
|