1
|
Nouari W, Aribi M. Innate lymphoid cells, immune functional dynamics, epithelial parallels, and therapeutic frontiers in infections. Int Rev Immunol 2025:1-28. [PMID: 40242974 DOI: 10.1080/08830185.2025.2490233] [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: 04/03/2024] [Revised: 02/19/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Abstract
Innate lymphoid cells (ILCs) have emerged as pivotal players in the field of immunology, expanding our understanding of innate immunity beyond conventional paradigms. This comprehensive review delves into the multifaceted world of ILCs, beginning with their serendipitous discovery and traversing their ontogeny and heterogeneity. We explore the distinct subsets of ILCs unraveling their intriguing plasticity, which adds a layer of complexity to their functional repertoire. As we journey through the functional activities of ILCs, we address their role in immune responses against various infections, categorizing their interactions with helminthic parasites, bacterial pathogens, fungal infections, and viral invaders. Notably, this review offers a detailed examination of ILCs in the context of specific infections, such as Mycobacterium tuberculosis, Citrobacter rodentium, Clostridium difficile, Salmonella typhimurium, Helicobacter pylori, Listeria monocytogenes, Staphylococcus aureus, Pseudomonas aeruginosa, Influenza virus, Cytomegalovirus, Herpes simplex virus, and severe acute respiratory syndrome coronavirus 2. This selection aimed for a comprehensive exploration of ILCs in various infectious contexts, opting for microorganisms based on extensive research findings rather than considerations of virulence or emergence. Furthermore, we raise intriguing questions about the potential immune functional resemblances between ILCs and epithelial cells, shedding light on their interconnectedness within the mucosal microenvironment. The review culminates in a critical assessment of the therapeutic prospects of targeting ILCs during infection, emphasizing their promise as novel immunotherapeutic targets. Nevertheless, due to their recent discovery and evolving understanding, effectively manipulating ILCs is challenging. Ensuring specificity and safety while evaluating long-term effects in clinical settings will be crucial.
Collapse
Affiliation(s)
- Wafa Nouari
- Laboratory of Applied Molecular Biology and Immunology, University of Tlemcen, Tlemcen, Algeria
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, University of Tlemcen, Tlemcen, Algeria
| |
Collapse
|
2
|
Nejadebrahim S, Houserová J, Ječmen T, Kalousková B, Abreu C, Herynek Š, Skořepa O, Bláha J, Vaněk O. Multiple O- and an N-glycosylation of the stalk region of the NK cell activation receptor NKp46 mediates its interaction with the Candida glabrata epithelial adhesin 1. Int J Biol Macromol 2025; 310:143037. [PMID: 40216117 DOI: 10.1016/j.ijbiomac.2025.143037] [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: 11/22/2024] [Revised: 03/12/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
Natural killer (NK) cells are critical components of the innate immune system. Their primary role is to induce apoptosis in target cells, such as cancerous or virally infected cells. These targets are recognized through interactions between activating or inhibitory receptors on the NK cell surface. Among the activating receptors is the natural cytotoxicity receptor NKp46. Several ligands for this receptor have been identified, including the epithelial adhesin Epa1 from the yeast Candida glabrata. Invasive candidiasis caused by this yeast is a significant complication for patients with hematological diseases. The interaction between NKp46 and Epa1 is thought to depend specifically on an O-glycan at threonine 225 of NKp46. To elucidate the molecular details of this interaction, we optimized the recombinant production of soluble NKp46 and Epa1, generated glycosylation variants of multiple NKp46 mutants, and evaluated the role of NKp46 glycosylation in Epa1 binding using microscale thermophoresis and isothermal titration calorimetry. Additionally, for the first time, we provide a comprehensive glycosylation profile of NKp46, determined through mass spectrometry of intact glycopeptides obtained by O-glycoprotease and trypsin proteolysis. Our findings demonstrate that the NKp46 stalk is glycosylated at multiple sites, involving both an N-glycan and more than one O-glycan. These glycans are critical for the interaction with Epa1, providing NK cells with enhanced sensitivity to Candida glabrata epitopes.
Collapse
Affiliation(s)
- Shiva Nejadebrahim
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030/8, 12840 Prague, Czech Republic
| | - Jana Houserová
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030/8, 12840 Prague, Czech Republic
| | - Tomáš Ječmen
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030/8, 12840 Prague, Czech Republic
| | - Barbora Kalousková
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030/8, 12840 Prague, Czech Republic
| | - Celeste Abreu
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030/8, 12840 Prague, Czech Republic
| | - Štěpán Herynek
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030/8, 12840 Prague, Czech Republic
| | - Ondřej Skořepa
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030/8, 12840 Prague, Czech Republic
| | - Jan Bláha
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030/8, 12840 Prague, Czech Republic
| | - Ondřej Vaněk
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030/8, 12840 Prague, Czech Republic.
| |
Collapse
|
3
|
Mi M, Zheng Y, Fu X, Bao N, Pan L, Qin G, Zhao Y. Integrated omics profiling of individual variations in intestinal damage to the soybean allergen in piglets. Front Vet Sci 2025; 11:1521544. [PMID: 39881721 PMCID: PMC11774947 DOI: 10.3389/fvets.2024.1521544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction A small number of soybean allergens [including Glycinin (11S) and β-Conglycinin (7S)] in the commercially available corn-soybean meal diet can still cause allergy in some weaned piglets, which may be the result of the interaction of genetic, and nutrition, but the specific mechanism is still unclear. Methods In this study, 20 allergic piglets and 20 non-allergic piglets were selected from 92 weaned piglets by skin sensitization tests, which were used to examine the whole sequence genome. The indicators related to humoral and cellular immunity, transcriptomics, and metabolomics analysis were determined by randomly selecting 5 boars in the allergic group and non-allergic group and then performing a validation in vitro. Results The sensitization rate of soybean antigen in the corn-soybean meal diet was 21.74% and there was a gender difference with the sensitization rate of female pigs (31.34%) being higher than that of male pigs (13.23%). Moreover, the levels of inflammatory factors (IL-1β, IL-4, TNF-α) and antibodies (IgG, IgE, and specific IgG) in allergic piglets were significantly higher than those in non-allergic piglets (P < 0.05). Whole genome re-sequencing analysis revealed specific mutations in the exons and URT5 of TRAPPC2, PIR, CFP, and SOWAHD genes and showed significantly higher expression levels of related genes in the spleen of allergic piglets (P < 0.05). Transcriptome analysis identified IL17REL, CCL19, CD1E, CD1.1, etc. immune differential genes, metabolomics results showed that soybean antigen affected the utilization and metabolism of intestinal nutrients in piglets, mainly the digestion and absorption of protein and the synthesis and metabolism of amino acids. Transfection of CFP/TRAPPC2/CCL19 siRNA could partially alleviate the injury of RAW264.7 cells or IPEC-J2 cells induced by β-Conglycinin. Conclusion Therefore, the individual differences in intestinal damage induced by soybean antigen protein in the corn-soybean meal diet are closely related to PIR, CFP, TRAPPC2, SOWAHD, and CCL19 genes. Soybean antigens affect the intestinal nutrient utilization and metabolism of piglets, which provides a scientific reference for the study of soybean antigen sensitization mechanisms, precision nutrition, disease prevention, and control of piglets, and also lays a foundation for human foodborne diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuan Zhao
- Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| |
Collapse
|
4
|
Mariuzza RA, Singh P, Karade SS, Shahid S, Sharma VK. Recognition of Self and Viral Ligands by NK Cell Receptors. Immunol Rev 2025; 329:e13435. [PMID: 39748148 PMCID: PMC11695704 DOI: 10.1111/imr.13435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025]
Abstract
Natural killer (NK) cells are essential elements of the innate immune response against tumors and viral infections. NK cell activation is governed by NK cell receptors that recognize both cellular (self) and viral (non-self) ligands, including MHC, MHC-related, and non-MHC molecules. These diverse receptors belong to two distinct structural families, the C-type lectin superfamily and the immunoglobulin superfamily. NK receptors include Ly49s, KIRs, LILRs, and NKG2A/CD94, which bind MHC class I (MHC-I) molecules, and NKG2D, which binds MHC-I paralogs such MICA and ULBP. Other NK receptors recognize tumor-associated antigens (NKp30, NKp44, NKp46), cell-cell adhesion proteins (KLRG1, CD96), or genetically coupled C-type lectin-like ligands (NKp65, NKR-P1). Additionally, cytomegaloviruses have evolved various immunoevasins, such as m157, m12, and UL18, which bind NK receptors and act as decoys to enable virus-infected cells to escape NK cell-mediated lysis. We review the remarkable progress made in the past 25 years in determining structures of representatives of most known NK receptors bound to MHC, MHC-like, and non-MHC ligands. Together, these structures reveal the multiplicity of solutions NK receptors have developed to recognize these molecules, and thereby mediate crucial interactions for regulating NK cytolytic activity by self and viral ligands.
Collapse
Affiliation(s)
- Roy A. Mariuzza
- W. M. Keck Laboratory for Structural BiologyUniversity of Maryland Institute for Bioscience and Biotechnology ResearchRockvilleMarylandUSA
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkMarylandUSA
| | - Pragya Singh
- W. M. Keck Laboratory for Structural BiologyUniversity of Maryland Institute for Bioscience and Biotechnology ResearchRockvilleMarylandUSA
- College of Natural and Mathematical SciencesUniversity of MarylandCollege ParkMarylandUSA
| | - Sharanbasappa S. Karade
- W. M. Keck Laboratory for Structural BiologyUniversity of Maryland Institute for Bioscience and Biotechnology ResearchRockvilleMarylandUSA
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkMarylandUSA
| | - Salman Shahid
- W. M. Keck Laboratory for Structural BiologyUniversity of Maryland Institute for Bioscience and Biotechnology ResearchRockvilleMarylandUSA
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkMarylandUSA
| | - Vijay Kumar Sharma
- W. M. Keck Laboratory for Structural BiologyUniversity of Maryland Institute for Bioscience and Biotechnology ResearchRockvilleMarylandUSA
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkMarylandUSA
| |
Collapse
|
5
|
Seillet C, Xiong L. ILC1 as critical gatekeepers in autoimmune kidney damage. Immunol Cell Biol 2025; 103:9-11. [PMID: 39642908 DOI: 10.1111/imcb.12842] [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] [Indexed: 12/09/2024]
Abstract
A recent article has shown that blocking NKp46 signaling reduces injury, highlighting these cells as key drivers of organ damage and potential therapeutic targets in autoimmune diseases. In lupus nephritis, NKp46+ ILC1s orchestrate kidney inflammation by producing CSF2, driving the expansion of pro-inflammatory macrophages that infiltrate epithelial niches and exacerbate tissue damage.
Collapse
Affiliation(s)
- Cyril Seillet
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Le Xiong
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
6
|
Qian S, Zhou Y, Jin Z, Li X, Tian Y, Chen F, Zhang B, Yan Z. Advancements in the Study of the Immune Molecule NKp46 in Immune System-related Diseases. Clin Rev Allergy Immunol 2024; 67:96-110. [PMID: 39612130 PMCID: PMC11638288 DOI: 10.1007/s12016-024-09010-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2024] [Indexed: 11/30/2024]
Abstract
NKp46 is a natural killer cell activating receptor primarily expressed on NK cells and non-NK innate lymphoid cells. In the context of anti-infection, NKp46 activates NK cells by binding to ligands on pathogens or infected cells, enabling NK cells to kill the infected cells. In antitumor activities, NKp46 plays a pivotal role in combating tumor growth through mechanisms such as directly killing tumor cells, inhibiting tumor immune escape, and reducing tumor growth rate through immune editing. The expression levels of NKp46 are closely associated with the progression of immune-related diseases, viral infections, leukemia, tumors, and reproductive failure, affecting diagnosis and prognosis. However, the functionality and mechanistic actions of NKp46, as well as the identification of additional NKp46 ligands, require further investigation. This review provides a comprehensive understanding of NKp46, offering a theoretical foundation for the research and development of diagnostic and therapeutic approaches for related diseases.
Collapse
Affiliation(s)
- Siyi Qian
- The 2nd Department of Gynecology Oncology of Hunan Cancer Hospital the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, People's Republic of China
- Department of Histology and Embryology, Basic School of Medicine Sciences, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine Sciences, Central South University, Changsha, Hunan, 410078, People's Republic of China
| | - Zhongyuan Jin
- Department of Pathology, Xiangya Medical School, Central South University, Changsha, People's Republic of China
| | - Xiang Li
- Department of Pathology, Xiangya Medical School, Central South University, Changsha, People's Republic of China
| | - Yuxuan Tian
- Department of Histology and Embryology, Basic School of Medicine Sciences, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Fuxin Chen
- Department of Histology and Embryology, Basic School of Medicine Sciences, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Bin Zhang
- The 2nd Department of Gynecology Oncology of Hunan Cancer Hospital the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, People's Republic of China.
- Department of Histology and Embryology, Basic School of Medicine Sciences, Central South University, Changsha, Hunan, 410013, People's Republic of China.
| | - Zhipeng Yan
- The 2nd Department of Gynecology Oncology of Hunan Cancer Hospital the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, People's Republic of China.
| |
Collapse
|
7
|
Brückner A, Brandtner A, Rieck S, Matthey M, Geisen C, Fels B, Stei M, Kusche-Vihrog K, Fleischmann BK, Wenzel D. Site-specific genetic and functional signatures of aortic endothelial cells at aneurysm predilection sites in healthy and AngII ApoE -/- mice. Angiogenesis 2024; 27:719-738. [PMID: 38965173 PMCID: PMC11564227 DOI: 10.1007/s10456-024-09933-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 06/16/2024] [Indexed: 07/06/2024]
Abstract
Aortic aneurysm is characterized by a pathological dilation at specific predilection sites of the vessel and potentially results in life-threatening vascular rupture. Herein, we established a modified "Häutchen method" for the local isolation of endothelial cells (ECs) from mouse aorta to analyze their spatial heterogeneity and potential role in site-specific disease development. When we compared ECs from aneurysm predilection sites of healthy mice with adjacent control segments we found regulation of genes related to extracellular matrix remodeling, angiogenesis and inflammation, all pathways playing a critical role in aneurysm development. We also detected enhanced cortical stiffness of the endothelium at these sites. Gene expression of ECs from aneurysms of the AngII ApoE-/- model when compared to sham animals mimicked expression patterns from predilection sites of healthy animals. Thus, this work highlights a striking genetic and functional regional heterogeneity in aortic ECs of healthy mice, which defines the location of aortic aneurysm formation in disease.
Collapse
Affiliation(s)
- Alexander Brückner
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Adrian Brandtner
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Sarah Rieck
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Michaela Matthey
- Department of Systems Physiology, Medical Faculty, Institute of Physiology, Ruhr University of Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Caroline Geisen
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Benedikt Fels
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner SiteHamburg/Luebeck/Kiel, Luebeck, Germany
| | - Marta Stei
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Kristina Kusche-Vihrog
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner SiteHamburg/Luebeck/Kiel, Luebeck, Germany
| | - Bernd K Fleischmann
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Daniela Wenzel
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany.
- Department of Systems Physiology, Medical Faculty, Institute of Physiology, Ruhr University of Bochum, Universitätsstr. 150, 44801, Bochum, Germany.
| |
Collapse
|
8
|
Liu G, He X, Zhao G, Lu Z. Complement regulation in tumor immune evasion. Semin Immunol 2024; 76:101912. [PMID: 39579520 DOI: 10.1016/j.smim.2024.101912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
The complement system plays crucial roles in both innate and adaptive immune responses, facilitating the elimination of pathogens such as microorganisms and damaged cells, including cancer cells. It is tightly regulated and integrated with cell-mediated immunity. In the tumor microenvironment, the complement system performs both immune and nonimmune functions in tumor and immune cells through pathways that depend on or are independent of complement activation, thereby promoting immune evasion and tumor progression.
Collapse
Affiliation(s)
- Guijun Liu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou, Zhejiang 310029, China; Zhejiang University Cancer Center, Hangzhou, Zhejiang 310029, China
| | - Xuxiao He
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou, Zhejiang 310029, China; Zhejiang University Cancer Center, Hangzhou, Zhejiang 310029, China
| | - Gaoxiang Zhao
- Department of Oncology, Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266061, China
| | - Zhimin Lu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou, Zhejiang 310029, China; Zhejiang University Cancer Center, Hangzhou, Zhejiang 310029, China.
| |
Collapse
|
9
|
Biniaris-Georgallis SI, Aschman T, Stergioula K, Schreiber F, Jafari V, Taranko A, Karmalkar T, Kasapi A, Lenac Rovis T, Jelencic V, Bejarano DA, Fabry L, Papacharalampous M, Mattiola I, Molgora M, Hou J, Hublitz KW, Heinrich F, Guerra GM, Durek P, Patone G, Lindberg EL, Maatz H, Hölsken O, Krönke G, Mortha A, Voll RE, Clarke AJ, Hauser AE, Colonna M, Thurley K, Schlitzer A, Schneider C, Stamatiades EG, Mashreghi MF, Jonjic S, Hübner N, Diefenbach A, Kanda M, Triantafyllopoulou A. Amplification of autoimmune organ damage by NKp46-activated ILC1s. Nature 2024; 634:952-960. [PMID: 39137897 DOI: 10.1038/s41586-024-07907-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
In systemic lupus erythematosus, loss of immune tolerance, autoantibody production and immune complex deposition are required but not sufficient for organ damage1. How inflammatory signals are initiated and amplified in the setting of autoimmunity remains elusive. Here we set out to dissect layers and hierarchies of autoimmune kidney inflammation to identify tissue-specific cellular hubs that amplify autoinflammatory responses. Using high-resolution single-cell profiling of kidney immune and parenchymal cells, in combination with antibody blockade and genetic deficiency, we show that tissue-resident NKp46+ innate lymphoid cells (ILCs) are crucial signal amplifiers of disease-associated macrophage expansion and epithelial cell injury in lupus nephritis, downstream of autoantibody production. NKp46 signalling in a distinct subset of group 1 ILCs (ILC1s) instructed an unconventional immune-regulatory transcriptional program, which included the expression of the myeloid cell growth factor CSF2. CSF2 production by NKp46+ ILCs promoted the population expansion of monocyte-derived macrophages. Blockade of the NKp46 receptor (using the antibody clone mNCR1.15; ref. 2) or genetic deficiency of NKp46 abrogated epithelial cell injury. The same cellular and molecular patterns were operative in human lupus nephritis. Our data provide support for the idea that NKp46+ ILC1s promote parenchymal cell injury by granting monocyte-derived macrophages access to epithelial cell niches. NKp46 activation in ILC1s therefore constitutes a previously unrecognized, crucial tissue rheostat that amplifies organ damage in autoimmune hosts, with broad implications for inflammatory pathologies and therapies.
Collapse
Affiliation(s)
- Stylianos-Iason Biniaris-Georgallis
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Free University of Berlin, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
| | - Tom Aschman
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center -University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neuropathology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
| | - Katerina Stergioula
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Free University of Berlin, Berlin, Germany
| | - Frauke Schreiber
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Vajiheh Jafari
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Free University of Berlin, Berlin, Germany
| | - Anna Taranko
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Free University of Berlin, Berlin, Germany
| | - Tejal Karmalkar
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Free University of Berlin, Berlin, Germany
| | - Ana Kasapi
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Tihana Lenac Rovis
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Vedrana Jelencic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - David A Bejarano
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Lea Fabry
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Michail Papacharalampous
- Department of Rheumatology and Clinical Immunology, Medical Center -University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Irene Mattiola
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Martina Molgora
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jinchao Hou
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Anesthesiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Karolin W Hublitz
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Frederik Heinrich
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | | | - Pawel Durek
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Giannino Patone
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Eric L Lindberg
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Henrike Maatz
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
| | - Oliver Hölsken
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Anesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Berlin Institute of Health at Charité Universitätsmedizin, Berlin, BIH Academy, Junior Clinician Scientist Program, Berlin, Germany
| | - Gerhard Krönke
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Medical Center -University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Anja E Hauser
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Kevin Thurley
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Biomathematics Division, Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Andreas Schlitzer
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | | | - Efstathios G Stamatiades
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Mir-Farzin Mashreghi
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- German Center for Child and Adolescent Health (DZKJ), Partner Site Berlin, Berlin, Germany
| | - Stipan Jonjic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Norbert Hübner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- Charité-Universitätsmedizin, Berlin, Germany
| | - Andreas Diefenbach
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany.
| | - Masatoshi Kanda
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- Department of Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan.
| | - Antigoni Triantafyllopoulou
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany.
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany.
| |
Collapse
|
10
|
Liu YT, Wu HL, Su YD, Wang Y, Li Y. Development in the Study of Natural Killer Cells for Malignant Peritoneal Mesothelioma Treatment. Cancer Biother Radiopharm 2024; 39:551-561. [PMID: 39093850 DOI: 10.1089/cbr.2024.0078] [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] [Indexed: 08/04/2024] Open
Abstract
Malignant peritoneal mesothelioma (MPeM) is a rare primary malignant tumor originating from peritoneal mesothelial cells. Insufficient specificity of the symptoms and their frequent reappearance following surgery make it challenging to diagnose, creating a need for more efficient treatment options. Natural killer cells (NK cells) are part of the innate immune system and are classified as lymphoid cells. Under the regulation of activating and inhibiting receptors, NK cells secrete various cytokines to exert cytotoxic effects and participate in antiforeign body, antiviral, and antitumor activities. This review provides a comprehensive summary of the specific alterations observed in NK cells following MPeM treatment, including changes in cell number, subpopulation distribution, active receptors, and cytotoxicity. In addition, we summarize the impact of various therapeutic interventions, such as chemotherapy, immunotherapy, and targeted therapy, on NK cell function post-MPeM treatment.
Collapse
Affiliation(s)
- Yi-Tong Liu
- Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - He-Liang Wu
- Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital, Peking University Ninth School of Clinical Medicine, Beijing, China
| | - Yan-Dong Su
- Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yi Wang
- Department of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yan Li
- Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of Surgical Oncology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| |
Collapse
|
11
|
Jiang P, Jing S, Sheng G, Jia F. The basic biology of NK cells and its application in tumor immunotherapy. Front Immunol 2024; 15:1420205. [PMID: 39221244 PMCID: PMC11361984 DOI: 10.3389/fimmu.2024.1420205] [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: 04/19/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Natural Killer (NK) cells play a crucial role as effector cells within the tumor immune microenvironment, capable of identifying and eliminating tumor cells through the expression of diverse activating and inhibitory receptors that recognize tumor-related ligands. Therefore, harnessing NK cells for therapeutic purposes represents a significant adjunct to T cell-based tumor immunotherapy strategies. Presently, NK cell-based tumor immunotherapy strategies encompass various approaches, including adoptive NK cell therapy, cytokine therapy, antibody-based NK cell therapy (enhancing ADCC mediated by NK cells, NK cell engagers, immune checkpoint blockade therapy) and the utilization of nanoparticles and small molecules to modulate NK cell anti-tumor functionality. This article presents a comprehensive overview of the latest advances in NK cell-based anti-tumor immunotherapy, with the aim of offering insights and methodologies for the clinical treatment of cancer patients.
Collapse
Affiliation(s)
- Pan Jiang
- Department of General Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Department of Infectious Diseases, Jingzhou First People’s Hospital, Jingzhou, China
| | - Shaoze Jing
- Department of Orthopedics, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fajing Jia
- Department of General Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
12
|
Yuan T, Zhou Q, Tian Y, Ou Y, Long Y, Tan Y. Innate lymphoid cells and infectious diseases. Innate Immun 2024; 30:120-135. [PMID: 39363687 PMCID: PMC11556573 DOI: 10.1177/17534259241287311] [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: 01/19/2024] [Revised: 08/19/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
Innate lymphoid cells (ILCs) are the main resident lymphocytes that mostly reside in tissues owing to the lack of adaptive antigen receptors. These cells are involved in early anti-infective immunity, antitumour immunity, regulation of tissue inflammation, and maintenance of homeostasis in the internal environment of tissues and have been referred to as the "first armies stationed in the human body". ILCs are widely distributed in the lungs, colon, lymph nodes, oral mucosa and even embryonic tissues. Due to the advantage of their distribution location, they are often among the first cells to come into contact with pathogens.Relevant studies have demonstrated that ILCs play an early role in the defence against a variety of pathogenic microorganisms, including bacteria, viruses, fungi and helminths, before they intervene in the adaptive immune system. ILCs can initiate a rapid, nonspecific response against pathogens prior to the initiation of an adaptive immune response and can generate a protective immune response against specific pathogens, secreting different effectors to play a role.There is growing evidence that ILCs play an important role in host control of infectious diseases. In this paper, we summarize and discuss the current known infectious diseases in which ILCs are involved and ILC contribution to the defence against infectious diseases. Further insights into the mechanisms of ILCs action in different infectious diseases will be useful in facilitating the development of therapeutic strategies for early control of infections.
Collapse
Affiliation(s)
- Ting Yuan
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Qianhui Zhou
- Department of Respiratory and Critical Care Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Yuqiu Tian
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Yangjing Ou
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - YunZhu Long
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - YingZheng Tan
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| |
Collapse
|
13
|
Lee MJ, de los Rios Kobara I, Barnard TR, Vales Torres X, Tobin NH, Ferbas KG, Rimoin AW, Yang OO, Aldrovandi GM, Wilk AJ, Fulcher JA, Blish CA. NK Cell-Monocyte Cross-talk Underlies NK Cell Activation in Severe COVID-19. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1693-1705. [PMID: 38578283 PMCID: PMC11102029 DOI: 10.4049/jimmunol.2300731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/13/2024] [Indexed: 04/06/2024]
Abstract
NK cells in the peripheral blood of severe COVID-19 patients exhibit a unique profile characterized by activation and dysfunction. Previous studies have identified soluble factors, including type I IFN and TGF-β, that underlie this dysregulation. However, the role of cell-cell interactions in modulating NK cell function during COVID-19 remains unclear. To address this question, we combined cell-cell communication analysis on existing single-cell RNA sequencing data with in vitro primary cell coculture experiments to dissect the mechanisms underlying NK cell dysfunction in COVID-19. We found that NK cells are predicted to interact most strongly with monocytes and that this occurs via both soluble factors and direct interactions. To validate these findings, we performed in vitro cocultures in which NK cells from healthy human donors were incubated with monocytes from COVID-19+ or healthy donors. Coculture of healthy NK cells with monocytes from COVID-19 patients recapitulated aspects of the NK cell phenotype observed in severe COVID-19, including decreased expression of NKG2D, increased expression of activation markers, and increased proliferation. When these experiments were performed in a Transwell setting, we found that only CD56bright CD16- NK cells were activated in the presence of severe COVID-19 patient monocytes. O-link analysis of supernatants from Transwell cocultures revealed that cultures containing severe COVID-19 patient monocytes had significantly elevated levels of proinflammatory cytokines and chemokines, as well as TGF-β. Collectively, these results demonstrate that interactions between NK cells and monocytes in the peripheral blood of COVID-19 patients contribute to NK cell activation and dysfunction in severe COVID-19.
Collapse
Affiliation(s)
- Madeline J. Lee
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
- Stanford Immunology Program, Stanford University School of Medicine, Palo Alto, CA
| | - Izumi de los Rios Kobara
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
- Stanford Immunology Program, Stanford University School of Medicine, Palo Alto, CA
| | - Trisha R. Barnard
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
| | - Xariana Vales Torres
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
- Stanford Immunology Program, Stanford University School of Medicine, Palo Alto, CA
| | - Nicole H. Tobin
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Kathie G. Ferbas
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Anne W. Rimoin
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA
| | - Otto O. Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Grace M. Aldrovandi
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Aaron J. Wilk
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Palo Alto, CA
| | - Jennifer A. Fulcher
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Catherine A. Blish
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
- Chan Zuckerberg Biohub, San Francisco, CA
| |
Collapse
|
14
|
Aguilar OA, Fong LK, Lanier LL. ITAM-based receptors in natural killer cells. Immunol Rev 2024; 323:40-53. [PMID: 38411263 PMCID: PMC11102329 DOI: 10.1111/imr.13313] [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: 10/31/2023] [Accepted: 02/15/2024] [Indexed: 02/28/2024]
Abstract
The ability of cells of the immune system to acquire features such as increased longevity and enhanced secondary responses was long thought to be restricted to cells of the adaptive immune system. Natural killer (NK) cells have challenged this notion by demonstrating that they can also gain adaptive features. This has been observed in both humans and mice during infection with cytomegalovirus (CMV). The generation of adaptive NK cells requires antigen-specific recognition of virally infected cells through stimulatory NK receptors. These receptors lack the ability to signal on their own and rather rely on adaptor molecules that contain ITAMs for driving signals. Here, we highlight our understanding of how these receptors influence the production of adaptive NK cells and propose areas in the field that merit further investigation.
Collapse
Affiliation(s)
- Oscar A. Aguilar
- Dept. of Microbiology and Immunology, University of California - San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California - San Francisco, San Francisco, CA, USA
| | - Lam-Kiu Fong
- Dept. of Pharmaceutical Chemistry, University of California – San Francisco, San Francisco, CA
| | - Lewis L. Lanier
- Dept. of Microbiology and Immunology, University of California - San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California - San Francisco, San Francisco, CA, USA
| |
Collapse
|
15
|
Lu M, Lee Z, Hu WS. Multi-omics kinetic analysis of recombinant adeno-associated virus production by plasmid transfection of HEK293 cells. Biotechnol Prog 2024; 40:e3428. [PMID: 38289617 DOI: 10.1002/btpr.3428] [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: 09/01/2023] [Revised: 12/06/2023] [Accepted: 12/27/2023] [Indexed: 04/19/2024]
Abstract
Recombinant adeno-associated virus (rAAV) is among the most commonly used vectors for gene therapy. It is commonly produced by transfection of HEK293 cells with three plasmids each containing the vector genome including gene of interest (GOI), helper functions, and rep and cap genes for genome replication and capsid formation. To meet the potential clinical needs, the productivity of the production system needs to be enhanced. A better process characterization of the production system will further advance our insights into ways to enhance productivity. Here, we employed transcriptomic analysis to quantify the dynamics of different isoforms of viral transcripts and to assess the shift of cellular physiology, and deployed targeted proteomic analysis for absolute quantification of viral proteins and tandem mass tags (TMTs) for assessing cellular responses at the protein level. Functional analysis at transcriptome and proteome levels identified defense and immune response, unfolded protein response, p53 signaling as enriched. The small molecule additive intervention study based on functional analysis showed the potential of such omics-guided productivity enhancement. Together, multi-omics analysis advanced understanding of rAAV production and provided insight into enhancing rAAV production by plasmid transfection.
Collapse
Affiliation(s)
- Min Lu
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Zion Lee
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Wei-Shou Hu
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
16
|
Mastellos DC, Hajishengallis G, Lambris JD. A guide to complement biology, pathology and therapeutic opportunity. Nat Rev Immunol 2024; 24:118-141. [PMID: 37670180 DOI: 10.1038/s41577-023-00926-1] [Citation(s) in RCA: 89] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2023] [Indexed: 09/07/2023]
Abstract
Complement has long been considered a key innate immune effector system that mediates host defence and tissue homeostasis. Yet, growing evidence has illuminated a broader involvement of complement in fundamental biological processes extending far beyond its traditional realm in innate immunity. Complement engages in intricate crosstalk with multiple pattern-recognition and signalling pathways both in the extracellular and intracellular space. Besides modulating host-pathogen interactions, this crosstalk guides early developmental processes and distinct cell trajectories, shaping tissue immunometabolic and regenerative programmes in different physiological systems. This Review provides a guide to the system-wide functions of complement. It highlights illustrative paradigm shifts that have reshaped our understanding of complement pathobiology, drawing examples from evolution, development of the central nervous system, tissue regeneration and cancer immunity. Despite its tight spatiotemporal regulation, complement activation can be derailed, fuelling inflammatory tissue pathology. The pervasive contribution of complement to disease pathophysiology has inspired a resurgence of complement therapeutics with major clinical developments, some of which have challenged long-held dogmas. We thus highlight major therapeutic concepts and milestones in clinical complement intervention.
Collapse
Affiliation(s)
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Liu PY, Li HQ, Dong MQ, Gu XY, Xu SY, Xia SN, Bao XY, Xu Y, Cao X. Infiltrating myeloid cell-derived properdin markedly promotes microglia-mediated neuroinflammation after ischemic stroke. J Neuroinflammation 2023; 20:260. [PMID: 37951917 PMCID: PMC10640761 DOI: 10.1186/s12974-023-02946-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 10/31/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Emerging evidence has shown that myeloid cells that infiltrate into the peri-infarct region may influence the progression of ischemic stroke by interacting with microglia. Properdin, which is typically secreted by immune cells such as neutrophils, monocytes, and T cells, has been found to possess damage-associated molecular patterns (DAMPs) properties and can perform functions unrelated to the complement pathway. However, the role of properdin in modulating microglia-mediated post-stroke neuroinflammation remains unclear. METHODS Global and conditional (myeloid-specific) properdin-knockout mice were subjected to transient middle cerebral artery occlusion (tMCAO). Histopathological and behavioral tests were performed to assess ischemic brain injury in mice. Single-cell RNA sequencing and immunofluorescence staining were applied to explore the source and the expression level of properdin. The transcriptomic profile of properdin-activated primary microglia was depicted by transcriptome sequencing. Lentivirus was used for macrophage-inducible C-type lectin (Mincle) silencing in microglia. Conditioned medium from primary microglia was administered to primary cortex neurons to determine the neurotoxicity of microglia. A series of cellular and molecular biological techniques were used to evaluate the proinflammatory response, neuronal death, protein-protein interactions, and related signaling pathways, etc. RESULTS: The level of properdin was significantly increased, and brain-infiltrating neutrophils and macrophages were the main sources of properdin in the ischemic brain. Global and conditional myeloid knockout of properdin attenuated microglial overactivation and inflammatory responses at the acute stage of tMCAO in mice. Accordingly, treatment with recombinant properdin enhanced the production of proinflammatory cytokines and augmented microglia-potentiated neuronal death in primary culture. Mechanistically, recombinant properdin served as a novel ligand that activated Mincle receptors on microglia and downstream pathways to drive primary microglia-induced inflammatory responses. Intriguingly, properdin can directly bind to the microglial Mincle receptor to exert the above effects, while Mincle knockdown limits properdin-mediated microglial inflammation. CONCLUSION Properdin is a new medium by which infiltrating peripheral myeloid cells communicate with microglia, further activate microglia, and exacerbate brain injury in the ischemic brain, suggesting that targeted disruption of the interaction between properdin and Mincle on microglia or inhibition of their downstream signaling may improve the prognosis of ischemic stroke.
Collapse
Affiliation(s)
- Pin-Yi Liu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, People's Republic of China
| | - Hui-Qin Li
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, People's Republic of China
| | - Meng-Qi Dong
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, People's Republic of China
| | - Xin-Ya Gu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, People's Republic of China
| | - Si-Yi Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, People's Republic of China
| | - Sheng-Nan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, People's Republic of China
| | - Xin-Yu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, People's Republic of China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, People's Republic of China.
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu, 210008, People's Republic of China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, People's Republic of China.
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, Jiangsu, 210008, People's Republic of China.
- Nanjing Neurology Medical Center, Nanjing, Jiangsu, 210008, People's Republic of China.
| | - Xiang Cao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, People's Republic of China.
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu, 210008, People's Republic of China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, 210008, People's Republic of China.
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, Jiangsu, 210008, People's Republic of China.
- Nanjing Neurology Medical Center, Nanjing, Jiangsu, 210008, People's Republic of China.
| |
Collapse
|
18
|
Corvino D, Rommel D, Schneppenheim F, Bald T. Stressed out: NKp46 binds ecto-calreticulin. Immunol Cell Biol 2023; 101:684-686. [PMID: 37226854 DOI: 10.1111/imcb.12659] [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] [Indexed: 05/26/2023]
Abstract
In a recent article, Sen Santara et al. demonstrated that the activating natural killer (NK) cell receptor NKp46 binds to externalized calreticulin (ecto-CRT), leading to NK cell degranulation and target cell killing. They show that endoplasmic reticulum stress-induced ecto-CRT serves as a danger-associated molecular pattern, helping NK cells identify and eliminate infected, malignant, stressed or senescent cells.
Collapse
Affiliation(s)
- Dillon Corvino
- Institute for Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Denise Rommel
- Institute for Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | | | - Tobias Bald
- Institute for Experimental Oncology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
19
|
Molfetta R, Petillo S, Cippitelli M, Paolini R. SUMOylation and related post-translational modifications in natural killer cell anti-cancer responses. Front Cell Dev Biol 2023; 11:1213114. [PMID: 37313439 PMCID: PMC10258607 DOI: 10.3389/fcell.2023.1213114] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/17/2023] [Indexed: 06/15/2023] Open
Abstract
SUMOylation is a reversible modification that involves the covalent attachment of small ubiquitin-like modifier (SUMO) to target proteins, leading to changes in their localization, function, stability, and interactor profile. SUMOylation and additional related post-translational modifications have emerged as important modulators of various biological processes, including regulation of genomic stability and immune responses. Natural killer (NK) cells are innate immune cells that play a critical role in host defense against viral infections and tumors. NK cells can recognize and kill infected or transformed cells without prior sensitization, and their activity is tightly regulated by a balance of activating and inhibitory receptors. Expression of NK cell receptors as well as of their specific ligands on target cells is finely regulated during malignant transformation through the integration of different mechanisms including ubiquitin- and ubiquitin-like post-translational modifications. Our review summarizes the role of SUMOylation and other related pathways in the biology of NK cells with a special emphasis on the regulation of their response against cancer. The development of novel selective inhibitors as useful tools to potentiate NK-cell mediated killing of tumor cells is also briefly discussed.
Collapse
|
20
|
Roles of natural killer cells in immunity to cancer, and applications to immunotherapy. Nat Rev Immunol 2023; 23:90-105. [PMID: 35637393 DOI: 10.1038/s41577-022-00732-1] [Citation(s) in RCA: 251] [Impact Index Per Article: 125.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 02/04/2023]
Abstract
Great strides have been made in recent years towards understanding the roles of natural killer (NK) cells in immunity to tumours and viruses. NK cells are cytotoxic innate lymphoid cells that produce inflammatory cytokines and chemokines. By lysing transformed or infected cells, they limit tumour growth and viral infections. Whereas T cells recognize peptides presented by MHC molecules, NK cells display receptors that recognize stress-induced autologous proteins on cancer cells. At the same time, their functional activity is inhibited by MHC molecules displayed on such cells. The enormous potential of NK cells for immunotherapy for cancer is illustrated by their broad recognition of stressed cells regardless of neoantigen presentation, and enhanced activity against tumours that have lost expression of MHC class I owing to acquired resistance mechanisms. As a result, many efforts are under way to mobilize endogenous NK cells with therapeutics, or to provide populations of ex vivo-expanded NK cells as a cellular therapy, in some cases by equipping the NK cells with chimeric antigen receptors. Here we consider the key features that underlie why NK cells are emerging as important new additions to the cancer therapeutic arsenal.
Collapse
|
21
|
Morimoto T, Nakazawa T, Maeoka R, Nakagawa I, Tsujimura T, Matsuda R. Natural Killer Cell-Based Immunotherapy against Glioblastoma. Int J Mol Sci 2023; 24:ijms24032111. [PMID: 36768432 PMCID: PMC9916747 DOI: 10.3390/ijms24032111] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive and malignant primary brain tumor in adults. Despite multimodality treatment involving surgical resection, radiation therapy, chemotherapy, and tumor-treating fields, the median overall survival (OS) after diagnosis is approximately 2 years and the 5-year OS is poor. Considering the poor prognosis, novel treatment strategies are needed, such as immunotherapies, which include chimeric antigen receptor T-cell therapy, immune checkpoint inhibitors, vaccine therapy, and oncolytic virus therapy. However, these therapies have not achieved satisfactory outcomes. One reason for this is that these therapies are mainly based on activating T cells and controlling GBM progression. Natural killer (NK) cell-based immunotherapy involves the new feature of recognizing GBM via differing mechanisms from that of T cell-based immunotherapy. In this review, we focused on NK cell-based immunotherapy as a novel GBM treatment strategy.
Collapse
Affiliation(s)
- Takayuki Morimoto
- Department of Neurosurgery, Nara Medical University, Kashihara 634-8521, Japan
- Department of Neurosurgery, Nara City Hospital, Nara 630-8305, Japan
- Correspondence: (T.M.); (T.N.); Tel.: +81-744-22-3051 (T.M.); +81-745-84-9335 (T.N.)
| | - Tsutomu Nakazawa
- Department of Neurosurgery, Nara Medical University, Kashihara 634-8521, Japan
- Grandsoul Research Institute for Immunology, Inc., Uda 633-2221, Japan
- Clinic Grandsoul Nara, Uda 633-2221, Japan
- Correspondence: (T.M.); (T.N.); Tel.: +81-744-22-3051 (T.M.); +81-745-84-9335 (T.N.)
| | - Ryosuke Maeoka
- Department of Neurosurgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Ichiro Nakagawa
- Department of Neurosurgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Takahiro Tsujimura
- Grandsoul Research Institute for Immunology, Inc., Uda 633-2221, Japan
- Clinic Grandsoul Nara, Uda 633-2221, Japan
| | - Ryosuke Matsuda
- Department of Neurosurgery, Nara Medical University, Kashihara 634-8521, Japan
| |
Collapse
|
22
|
Moore SR, Menon SS, Galwankar NS, Khuder SA, Pangburn MK, Ferreira VP. A novel assay that characterizes properdin function shows neutrophil-derived properdin has a distinct oligomeric distribution. Front Immunol 2023; 13:918856. [PMID: 36713423 PMCID: PMC9880526 DOI: 10.3389/fimmu.2022.918856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 12/21/2022] [Indexed: 01/15/2023] Open
Abstract
Properdin acts as an essential positive regulator of the alternative pathway of complement by stabilizing enzymatic convertases. Identical properdin monomers form head-to-tail associations of oligomers in a reported 20:54:26 ratio (most often described as an approximate 1:2:1 ratio) of tetramers (P4), trimers (P3), and dimers (P2), in blood, under normal physiological conditions. Oligomeric size is proportional to properdin function with tetramers being more active, followed by trimers and dimers. Neutrophils are the most abundant granulocyte, are recruited to inflammatory microenvironments, and are a significant source of properdin, yet the ratio of properdin oligomers released from neutrophils is unknown. The oligomer ratio of neutrophil-derived properdin could have functional consequences in local microenvironments where neutrophils are abundant and complement drives inflammation. We investigated the oligomer properties of neutrophil-derived properdin, as compared to that of normal human sera, using a novel ELISA-based method that detects function of properdin in a way that was proportional to the oligomeric size of properdin (i.e., the larger the oligomer, the higher the detected function). Unexpectedly, neutrophil-derived properdin had 5-fold lower function than donor-matched serum-derived properdin. The lower function was due to a lower percentage of tetramers/trimers and more dimers, indicating a significantly different P4:P3:P2 ratio in neutrophil-derived properdin (18:34:48) as compared to donor-matched serum (29:43:29). Release of lower-order oligomers by neutrophils may constitute a novel regulatory mechanism to control the rate of complement activation in cellular microenvironments. Further studies to determine the factors that affect properdin oligomerization and whether, or how, the predominant dimers in neutrophil-derived properdin, assimilate to the ~1:2:1 ratio found in serum are warranted.
Collapse
Affiliation(s)
- Sara R. Moore
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Smrithi S. Menon
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Neeti S. Galwankar
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Sadik A. Khuder
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Michael K. Pangburn
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, TX, United States
| | - Viviana P. Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States,*Correspondence: Viviana P. Ferreira,
| |
Collapse
|
23
|
Pedersen DV, Lorentzen J, Andersen GR. Structural studies offer a framework for understanding the role of properdin in the alternative pathway and beyond. Immunol Rev 2023; 313:46-59. [PMID: 36097870 PMCID: PMC10087229 DOI: 10.1111/imr.13129] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Structures of alternative pathway proteins have offered a comprehensive structural basis for understanding the molecular mechanisms governing activation and regulation of the amplification pathway of the complement cascade. Although properdin (FP) is required in vivo to sustain a functional alternative pathway, structural studies have been lagging behind due to the extended structure and polydisperse nature of FP. We review recent progress with respect to structure determination of FP and its proconvertase/convertase complexes. These structures identify in detail regions in C3b, factor B and FP involved in their mutual interactions. Structures of FP oligomers obtained by integrative studies have shed light on how FP activity depends on its oligomerization state. The accumulated structural knowledge allows us to rationalize the effect of point mutations causing FP deficiency. The structural basis for FP inhibition by the tick CirpA proteins is reviewed and the potential of alphafold2 predictions for understanding the interaction of FP with other tick proteins and the NKp46 receptor on host immune cells is discussed. The accumulated structural knowledge forms a comprehensive basis for understanding molecular interactions involving FP, pathological conditions arising from low levels of FP, and the molecular strategies used by ticks to suppress the alternative pathway.
Collapse
Affiliation(s)
| | - Josefine Lorentzen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Gregers Rom Andersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
24
|
Zafarani A, Taghavi-Farahabadi M, Razizadeh MH, Amirzargar MR, Mansouri M, Mahmoudi M. The Role of NK Cells and Their Exosomes in Graft Versus Host Disease and Graft Versus Leukemia. Stem Cell Rev Rep 2023; 19:26-45. [PMID: 35994137 DOI: 10.1007/s12015-022-10449-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2022] [Indexed: 02/07/2023]
Abstract
Natural killer (NK) cells are one of the innate immune cells that play an important role in preventing and controlling tumors and viral diseases, but their role in hematopoietic stem cell transplantation (HCT) is not yet fully understood. However, according to some research, these cells can prevent infections and tumor relapse without causing graft versus host disease (GVHD). In addition to NK cells, several studies are about the anti-leukemia effects of NK cell-derived exosomes that can highlight their roles in graft-versus-leukemia (GVL). In this paper, we intend to investigate the results of various articles on the role of NK cells in allogeneic hematopoietic cell transplantation and also their exosomes in GVL. Also, we have discussed the antiviral effects of these cells in post-HCT cytomegalovirus infection.
Collapse
Affiliation(s)
- Alireza Zafarani
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Taghavi-Farahabadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Reza Amirzargar
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mansoure Mansouri
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahmoudi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Peipp M, Klausz K, Boje AS, Zeller T, Zielonka S, Kellner C. Immunotherapeutic targeting of activating natural killer cell receptors and their ligands in cancer. Clin Exp Immunol 2022; 209:22-32. [PMID: 35325068 PMCID: PMC9307233 DOI: 10.1093/cei/uxac028] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/15/2022] [Accepted: 03/22/2022] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells exert an important role in cancer immune surveillance. Recognition of malignant cells and controlled activation of effector functions are facilitated by the expression of activating and inhibitory receptors, which is a complex interplay that allows NK cells to discriminate malignant cells from healthy tissues. Due to their unique profile of effector functions, the recruitment of NK cells is attractive in cancer treatment and a key function of NK cells in antibody therapy is widely appreciated. In recent years, besides the low-affinity fragment crystallizable receptor for immunoglobulin G (FcγRIIIA), the activating natural killer receptors p30 (NKp30) and p46 (NKp46), as well as natural killer group 2 member D (NKG2D), have gained increasing attention as potential targets for bispecific antibody-derivatives to redirect NK cell cytotoxicity against tumors. Beyond modulation of the receptor activity on NK cells, therapeutic targeting of the respective ligands represents an attractive approach. Here, novel therapeutic approaches to unleash NK cells by engagement of activating NK-cell receptors and alternative strategies targeting their tumor-expressed ligands in cancer therapy are summarized.
Collapse
Affiliation(s)
- Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ammelie Svea Boje
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Tobias Zeller
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Stefan Zielonka
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Christian Kellner
- Correspondence: Christian Kellner, Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
26
|
Papak I, Chruściel E, Dziubek K, Kurkowiak M, Urban-Wójciuk Z, Marjański T, Rzyman W, Marek-Trzonkowska N. What Inhibits Natural Killers’ Performance in Tumour. Int J Mol Sci 2022; 23:ijms23137030. [PMID: 35806034 PMCID: PMC9266640 DOI: 10.3390/ijms23137030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/16/2022] [Accepted: 06/22/2022] [Indexed: 12/21/2022] Open
Abstract
Natural killer cells are innate lymphocytes with the ability to lyse tumour cells depending on the balance of their activating and inhibiting receptors. Growing numbers of clinical trials show promising results of NK cell-based immunotherapies. Unlike T cells, NK cells can lyse tumour cells independent of antigen presentation, based simply on their activation and inhibition receptors. Various strategies to improve NK cell-based therapies are being developed, all with one goal: to shift the balance to activation. In this review, we discuss the current understanding of ways NK cells can lyse tumour cells and all the inhibitory signals stopping their cytotoxic potential.
Collapse
Affiliation(s)
- Ines Papak
- International Centre for Cancer Vaccine Science, University of Gdansk, Ul. Kładki 24, 80-822 Gdansk, Poland; (I.P.); (E.C.); (K.D.); (M.K.); (Z.U.-W.)
| | - Elżbieta Chruściel
- International Centre for Cancer Vaccine Science, University of Gdansk, Ul. Kładki 24, 80-822 Gdansk, Poland; (I.P.); (E.C.); (K.D.); (M.K.); (Z.U.-W.)
| | - Katarzyna Dziubek
- International Centre for Cancer Vaccine Science, University of Gdansk, Ul. Kładki 24, 80-822 Gdansk, Poland; (I.P.); (E.C.); (K.D.); (M.K.); (Z.U.-W.)
| | - Małgorzata Kurkowiak
- International Centre for Cancer Vaccine Science, University of Gdansk, Ul. Kładki 24, 80-822 Gdansk, Poland; (I.P.); (E.C.); (K.D.); (M.K.); (Z.U.-W.)
| | - Zuzanna Urban-Wójciuk
- International Centre for Cancer Vaccine Science, University of Gdansk, Ul. Kładki 24, 80-822 Gdansk, Poland; (I.P.); (E.C.); (K.D.); (M.K.); (Z.U.-W.)
| | - Tomasz Marjański
- Department of Thoracic Surgery, Medical University of Gdansk, 80-210 Gdansk, Poland; (T.M.); (W.R.)
| | - Witold Rzyman
- Department of Thoracic Surgery, Medical University of Gdansk, 80-210 Gdansk, Poland; (T.M.); (W.R.)
| | - Natalia Marek-Trzonkowska
- International Centre for Cancer Vaccine Science, University of Gdansk, Ul. Kładki 24, 80-822 Gdansk, Poland; (I.P.); (E.C.); (K.D.); (M.K.); (Z.U.-W.)
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdansk, 80-210 Gdansk, Poland
- Correspondence:
| |
Collapse
|
27
|
Cortes C, Desler C, Mazzoli A, Chen JY, Ferreira VP. The role of properdin and Factor H in disease. Adv Immunol 2022; 153:1-90. [PMID: 35469595 DOI: 10.1016/bs.ai.2021.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The complement system consists of three pathways (alternative, classical, and lectin) that play a fundamental role in immunity and homeostasis. The multifunctional role of the complement system includes direct lysis of pathogens, tagging pathogens for phagocytosis, promotion of inflammatory responses to control infection, regulation of adaptive cellular immune responses, and removal of apoptotic/dead cells and immune complexes from circulation. A tight regulation of the complement system is essential to avoid unwanted complement-mediated damage to the host. This regulation is ensured by a set of proteins called complement regulatory proteins. Deficiencies or malfunction of these regulatory proteins may lead to pro-thrombotic hematological diseases, renal and ocular diseases, and autoimmune diseases, among others. This review focuses on the importance of two complement regulatory proteins of the alternative pathway, Factor H and properdin, and their role in human diseases with an emphasis on: (a) characterizing the main mechanism of action of Factor H and properdin in regulating the complement system and protecting the host from complement-mediated attack, (b) describing the dysregulation of the alternative pathway as a result of deficiencies, or mutations, in Factor H and properdin, (c) outlining the clinical findings, management and treatment of diseases associated with mutations and deficiencies in Factor H, and (d) defining the unwanted and inadequate functioning of properdin in disease, through a discussion of various experimental research findings utilizing in vitro, mouse and human models.
Collapse
Affiliation(s)
- Claudio Cortes
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, United States.
| | - Caroline Desler
- Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| | - Amanda Mazzoli
- Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| | - Jin Y Chen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States.
| |
Collapse
|
28
|
Essen MF, Schlagwein N, den Hoven EM, Gijlswijk‐Janssen DJ, Lubbers R, den Bos RM, den Born J, Ruben JM, Trouw LA, Kooten C. Initial properdin binding contributes to alternative pathway activation at the surface of viable and necrotic cells. Eur J Immunol 2022; 52:597-608. [PMID: 35092629 PMCID: PMC9303752 DOI: 10.1002/eji.202149259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 12/01/2021] [Accepted: 01/10/2022] [Indexed: 11/16/2022]
Abstract
Properdin, the only known positive regulator of the complement system, stabilizes the C3 convertase, thereby increasing its half‐life. In contrast to most other complement factors, properdin is mainly produced extrahepatically by myeloid cells. Recent data suggest a role for properdin as a pattern recognition molecule. Here, we confirmed previous findings of properdin binding to different necrotic cells including Jurkat T cells. Binding can occur independent of C3, as demonstrated by HAP‐1 C3 KO cells, excluding a role for endogenous C3. In view of the cellular source of properdin, interaction with myeloid cells was examined. Properdin bound to the surface of viable monocyte‐derived pro‐ and anti‐inflammatory macrophages, but not to DCs. Binding was demonstrated for purified properdin as well as fractionated P2, P3, and P4 properdin oligomers. Binding contributed to local complement activation as determined by C3 and C5b‐9 deposition on the cell surfaces and seems a prerequisite for alternative pathway activation. Interaction of properdin with cell surfaces could be inhibited with the tick protein Salp20 and by different polysaccharides, depending on sulfation and chain length. These data identify properdin as a factor interacting with different cell surfaces, being either dead or alive, contributing to the local stimulation of complement activation.
Collapse
Affiliation(s)
- Mieke F. Essen
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Nicole Schlagwein
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Elisa M.P. den Hoven
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Daniëlle J. Gijlswijk‐Janssen
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Rosalie Lubbers
- Department of Rheumatology Leiden University Medical Center Leiden The Netherlands
| | - Ramon M. den Bos
- Crystal and Structural Chemistry Bijvoet Center for Biomolecular Research Department of Chemistry Faculty of Science Utrecht University Utrecht The Netherlands
| | - Jacob den Born
- Department of Nephrology University Medical Center Groningen Groningen The Netherlands
| | - Jurjen M. Ruben
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | - Leendert A. Trouw
- Department of Rheumatology Leiden University Medical Center Leiden The Netherlands
- Department of Immunology Leiden University Medical Center Leiden The Netherlands
| | - Cees Kooten
- Div. of Nephrology and Transplant Medicine Dept. of Medicine Leiden University Medical Center Leiden The Netherlands
| | | |
Collapse
|
29
|
Bergantini L, d’Alessandro M, Cameli P, Cavallaro D, Gangi S, Cekorja B, Sestini P, Bargagli E. NK and T Cell Immunological Signatures in Hospitalized Patients with COVID-19. Cells 2021; 10:3182. [PMID: 34831404 PMCID: PMC8618013 DOI: 10.3390/cells10113182] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023] Open
Abstract
Severe acute respiratory syndrome caused by coronavirus 2 emerged in Wuhan (China) in December 2019 and has severely challenged the human population. NK and T cells are involved in the progression of COVID-19 infection through the ability of NK cells to modulate T-cell responses, and by the stimulation of cytokine release. No detailed investigation of the NK cell landscape in clinical SARS-CoV-2 infection has yet been reported. A total of 35 COVID-19 hospitalised patients were stratified for clinical severity and 17 healthy subjects were enrolled. NK cell subsets and T cell subsets were analysed with flow cytometry. Serum cytokines were detected with a bead-based multiplex assay. Fewer CD56dimCD16brightNKG2A+NK cells and a parallel increase in the CD56+CD69+NK, CD56+PD-1+NK, CD56+NKp44+NK subset were reported in COVID-19 than HC. A significantly higher adaptive/memory-like NK cell frequency in patients with severe disease than in those with mild and moderate phenotypes were reported. Moreover, adaptive/memory-like NK cell frequencies were significantly higher in patients who died than in survivors. Severe COVID-19 patients showed higher serum concentrations of IL-6 than mild and control groups. Direct correlation emerged for IL-6 and adaptive/memory-like NK. All these findings provide new insights into the immune response of patients with COVID-19. In particular, they demonstrate activation of NK through overexpression of CD69 and CD25 and show that PD-1 inhibitory signalling maintains an exhausted phenotype in NK cells. These results suggest that adaptive/memory-like NK cells could be the basis of promising targeted therapy for future viral infections.
Collapse
|
30
|
Sahu SK, Kulkarni DH, Ozanturk AN, Ma L, Kulkarni HS. Emerging roles of the complement system in host-pathogen interactions. Trends Microbiol 2021; 30:390-402. [PMID: 34600784 DOI: 10.1016/j.tim.2021.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/01/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022]
Abstract
The complement system has historically been entertained as a fluid-phase, hepatically derived system which protects the intravascular space from encapsulated bacteria. However, there has been an increasing appreciation for its role in protection against non-encapsulated pathogens. Specifically, we have an improved understanding of how pathogens are recognized by specific complement proteins, as well as how they trigger and evade them. Additionally, we have an improved understanding of locally derived complement proteins, many of which promote host defense. Moreover, intracellular complement proteins have been identified that facilitate local protection and barrier function despite pathogen invasion. Our review aims to summarize these advances in the field as well as provide an insight into the pathophysiological changes occurring when the system is dysregulated in infection.
Collapse
Affiliation(s)
- Sanjaya K Sahu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Devesha H Kulkarni
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ayse N Ozanturk
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lina Ma
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Hrishikesh S Kulkarni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
31
|
Natural Killer Cells and Type 1 Innate Lymphoid Cells in Hepatocellular Carcinoma: Current Knowledge and Future Perspectives. Int J Mol Sci 2021; 22:ijms22169044. [PMID: 34445750 PMCID: PMC8396475 DOI: 10.3390/ijms22169044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/10/2021] [Accepted: 08/18/2021] [Indexed: 02/07/2023] Open
Abstract
Natural killer (NK) cells and type 1 innate lymphoid cells (ILC1) are specific innate lymphoid cell subsets that are key for the detection and elimination of pathogens and cancer cells. In liver, while they share a number of characteristics, they differ in many features. These include their developmental pathways, tissue distribution, phenotype and functions. NK cells and ILC1 contribute to organ homeostasis through the production of key cytokines and chemokines and the elimination of potential harmful bacteria and viruses. In addition, they are equipped with a wide range of receptors, allowing them to detect “stressed cells’ such as cancer cells. Our understanding of the role of innate lymphoid cells in hepatocellular carcinoma (HCC) is growing owing to the development of mouse models, the progress in immunotherapeutic treatment and the recent use of scRNA sequencing analyses. In this review, we summarize the current understanding of NK cells and ILC1 in hepatocellular carcinoma and discuss future strategies to take advantage of these innate immune cells in anti-tumor immunity. Immunotherapies hold great promise in HCC, and a better understanding of the role and function of NK cells and ILC1 in liver cancer could pave the way for new NK cell and/or ILC1-targeted treatment.
Collapse
|
32
|
Khalil M, Wang D, Hashemi E, Terhune SS, Malarkannan S. Implications of a 'Third Signal' in NK Cells. Cells 2021; 10:cells10081955. [PMID: 34440725 PMCID: PMC8393955 DOI: 10.3390/cells10081955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/17/2022] Open
Abstract
Innate and adaptive immune systems are evolutionarily divergent. Primary signaling in T and B cells depends on somatically rearranged clonotypic receptors. In contrast, NK cells use germline-encoded non-clonotypic receptors such as NCRs, NKG2D, and Ly49H. Proliferation and effector functions of T and B cells are dictated by unique peptide epitopes presented on MHC or soluble humoral antigens. However, in NK cells, the primary signals are mediated by self or viral proteins. Secondary signaling mediated by various cytokines is involved in metabolic reprogramming, proliferation, terminal maturation, or memory formation in both innate and adaptive lymphocytes. The family of common gamma (γc) cytokine receptors, including IL-2Rα/β/γ, IL-7Rα/γ, IL-15Rα/β/γ, and IL-21Rα/γ are the prime examples of these secondary signals. A distinct set of cytokine receptors mediate a ‘third’ set of signaling. These include IL-12Rβ1/β2, IL-18Rα/β, IL-23R, IL-27R (WSX-1/gp130), IL-35R (IL-12Rβ2/gp130), and IL-39R (IL-23Rα/gp130) that can prime, activate, and mediate effector functions in lymphocytes. The existence of the ‘third’ signal is known in both innate and adaptive lymphocytes. However, the necessity, context, and functional relevance of this ‘third signal’ in NK cells are elusive. Here, we define the current paradigm of the ‘third’ signal in NK cells and enumerate its clinical implications.
Collapse
Affiliation(s)
- Mohamed Khalil
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; (M.K.); (D.W.); (E.H.)
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Dandan Wang
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; (M.K.); (D.W.); (E.H.)
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Elaheh Hashemi
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; (M.K.); (D.W.); (E.H.)
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Scott S. Terhune
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Correspondence: (S.S.T.); (S.M.)
| | - Subramaniam Malarkannan
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Versiti, Milwaukee, WI 53226, USA; (M.K.); (D.W.); (E.H.)
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Correspondence: (S.S.T.); (S.M.)
| |
Collapse
|
33
|
Sportoletti P, De Falco F, Del Papa B, Baldoni S, Guarente V, Marra A, Dorillo E, Rompietti C, Adamo FM, Ruggeri L, Di Ianni M, Rosati E. NK Cells in Chronic Lymphocytic Leukemia and Their Therapeutic Implications. Int J Mol Sci 2021; 22:ijms22136665. [PMID: 34206399 PMCID: PMC8268440 DOI: 10.3390/ijms22136665] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022] Open
Abstract
Key features of chronic lymphocytic leukemia (CLL) are defects in the immune system and the ability of leukemic cells to evade immune defenses and induce immunosuppression, resulting in increased susceptibility to infections and disease progression. Several immune effectors are impaired in CLL, including T and natural killer (NK) cells. The role of T cells in defense against CLL and in CLL progression and immunotherapy has been extensively studied. Less is known about the role of NK cells in this leukemia, and data on NK cell alterations in CLL are contrasting. Besides studies showing that NK cells have intrinsic defects in CLL, there is a large body of evidence indicating that NK cell dysfunctions in CLL mainly depend on the escape mechanisms employed by leukemic cells. In keeping, it has been shown that NK cell functions, including antibody-dependent cellular cytotoxicity (ADCC), can be retained and/or restored after adequate stimulation. Therefore, due to their preserved ADCC function and the reversibility of CLL-related dysfunctions, NK cells are an attractive source for novel immunotherapeutic strategies in this disease, including chimeric antigen receptor (CAR) therapy. Recently, satisfying clinical responses have been obtained in CLL patients using cord blood-derived CAR-NK cells, opening new possibilities for further exploring NK cells in the immunotherapy of CLL. However, notwithstanding the promising results of this clinical trial, more evidence is needed to fully understand whether and in which CLL cases NK cell-based immunotherapy may represent a valid, alternative/additional therapeutic option for this leukemia. In this review, we provide an overview of the current knowledge about phenotypic and functional alterations of NK cells in CLL and the mechanisms by which CLL cells circumvent NK cell-mediated immunosurveillance. Additionally, we discuss the potential relevance of using NK cells in CLL immunotherapy.
Collapse
MESH Headings
- Biomarkers
- Cell Communication
- Disease Management
- Disease Susceptibility
- Humans
- Immune System/immunology
- Immune System/metabolism
- Immunotherapy/adverse effects
- Immunotherapy/methods
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Ligands
- Protein Binding
- Receptors, Natural Killer Cell/genetics
- Receptors, Natural Killer Cell/metabolism
- Treatment Outcome
- Tumor Escape/genetics
- Tumor Escape/immunology
Collapse
Affiliation(s)
- Paolo Sportoletti
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Filomena De Falco
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Beatrice Del Papa
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Stefano Baldoni
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
- Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Valerio Guarente
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Andrea Marra
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Erica Dorillo
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Chiara Rompietti
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Francesco Maria Adamo
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Loredana Ruggeri
- Centro di Ricerca Emato-Oncologica (CREO), Department of Medicine and Surgery, Institute of Hematology, University of Perugia, 06129 Perugia, Italy; (P.S.); (F.D.F.); (B.D.P.); (S.B.); (V.G.); (A.M.); (E.D.); (C.R.); (F.M.A.); (L.R.)
| | - Mauro Di Ianni
- Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Department of Oncology and Hematology, Ospedale Civile “Santo Spirito”, ASL Pescara, 65124 Pescara, Italy
| | - Emanuela Rosati
- Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy
- Correspondence:
| |
Collapse
|
34
|
Demaria O, Gauthier L, Debroas G, Vivier E. Natural killer cell engagers in cancer immunotherapy: Next generation of immuno-oncology treatments. Eur J Immunol 2021; 51:1934-1942. [PMID: 34145579 DOI: 10.1002/eji.202048953] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/01/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022]
Abstract
Immuno-oncology is revolutionizing the treatment of cancers, by inducing the recognition and elimination of tumor cells by the immune system. Recent advances have focused on generating or unleashing tumor antigen-specific T-cell responses, leading to alternative treatment paradigms for many cancers. Despite these successes, the clinical benefit has been limited to a subset of patients and certain tumor types, highlighting the need for alternative strategies. One innovative approach is to broaden and amplify antitumoral immune responses by targeting innate immunity. Particularly, the aim has been to develop new antibody formats capable of stimulating the antitumor activity of innate immune cells, boosting not only their direct role in tumor elimination, but also their function in eliciting multicellular immune responses ultimately resulting in long-lasting tumor control by adaptive immunity. This review covers the development of a new class of synthetic molecules, natural killer cell engagers (NKCEs), which are built from fragments of monoclonal antibodies (mAbs) and are designed to harness the immune functions of NK cells in cancer. As currently shown in preclinical studies and clinical trials, NKCEs are promising candidates for the next generation of tumor immunotherapies.
Collapse
Affiliation(s)
| | | | | | - Eric Vivier
- Innate Pharma, Marseille, France.,Aix Marseille University, CNRS, INSERM, CIML, Marseille, France.,APHM, Hôpital de la Timone, Marseille-Immunopôle, Marseille, France
| |
Collapse
|
35
|
Murugaiah V, Varghese PM, Beirag N, DeCordova S, Sim RB, Kishore U. Complement Proteins as Soluble Pattern Recognition Receptors for Pathogenic Viruses. Viruses 2021; 13:v13050824. [PMID: 34063241 PMCID: PMC8147407 DOI: 10.3390/v13050824] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/28/2021] [Indexed: 12/11/2022] Open
Abstract
The complement system represents a crucial part of innate immunity. It contains a diverse range of soluble activators, membrane-bound receptors, and regulators. Its principal function is to eliminate pathogens via activation of three distinct pathways: classical, alternative, and lectin. In the case of viruses, the complement activation results in effector functions such as virion opsonisation by complement components, phagocytosis induction, virolysis by the membrane attack complex, and promotion of immune responses through anaphylatoxins and chemotactic factors. Recent studies have shown that the addition of individual complement components can neutralise viruses without requiring the activation of the complement cascade. While the complement-mediated effector functions can neutralise a diverse range of viruses, numerous viruses have evolved mechanisms to subvert complement recognition/activation by encoding several proteins that inhibit the complement system, contributing to viral survival and pathogenesis. This review focuses on these complement-dependent and -independent interactions of complement components (especially C1q, C4b-binding protein, properdin, factor H, Mannose-binding lectin, and Ficolins) with several viruses and their consequences.
Collapse
Affiliation(s)
- Valarmathy Murugaiah
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (V.M.); (P.M.V.); (N.B.); (S.D.)
| | - Praveen M. Varghese
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (V.M.); (P.M.V.); (N.B.); (S.D.)
| | - Nazar Beirag
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (V.M.); (P.M.V.); (N.B.); (S.D.)
| | - Syreeta DeCordova
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (V.M.); (P.M.V.); (N.B.); (S.D.)
| | - Robert B. Sim
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK;
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (V.M.); (P.M.V.); (N.B.); (S.D.)
- Correspondence: or
| |
Collapse
|
36
|
Zhong G. Chlamydia overcomes multiple gastrointestinal barriers to achieve long-lasting colonization. Trends Microbiol 2021; 29:1004-1012. [PMID: 33865675 DOI: 10.1016/j.tim.2021.03.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/19/2022]
Abstract
Chlamydia trachomatis (CT) is frequently detected in the human gastrointestinal (GI) tract despite its leading role in sexually transmitted bacterial infections in the genital tract. Chlamydia muridarum (CM), a model pathogen for investigating CT pathogenesis in the genital tract, can also colonize the mouse GI tract for long periods. Genital-tract mutants of CM no longer colonize the GI tract. The mutants lacking plasmid functions are more defective in colonizing the upper GI tract while certain chromosomal gene-deficient mutants are more defective in the lower GI tract, suggesting that Chlamydia may use the plasmid for promoting its spread to the large intestine while using the chromosome-encoded factors for maintaining its colonization in the large intestine. The plasmid-encoded Pgp3 is critical for Chlamydia to resist the acid barrier in the stomach and to overcome a CD4+ T cell barrier in the small intestine. On reaching the large intestine, Pgp3 is no longer required. Instead, the chromosome-encoded open reading frames TC0237/TC0668 become essential for Chlamydia to evade the group 3-like innate lymphoid cell-secreted interferon (IFN)γ in the large intestine. These findings are important for exploring the medical significance of chlamydial colonization in the gut and for understanding the mechanisms of chlamydial pathogenicity in the genital tract.
Collapse
Affiliation(s)
- Guangming Zhong
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
37
|
Cui G, Geng L, Zhu L, Lin Z, Liu X, Miao Z, Jiang J, Feng X, Wei F. CFP is a prognostic biomarker and correlated with immune infiltrates in Gastric Cancer and Lung Cancer. J Cancer 2021; 12:3378-3390. [PMID: 33976747 PMCID: PMC8100816 DOI: 10.7150/jca.50832] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 03/21/2021] [Indexed: 01/06/2023] Open
Abstract
Complement factor properdin (CFP), encodes plasma glycoprotein, is a critical gene that regulates the complement pathway of the innate immune system. However, correlations of CFP in cancers remain unclear. In this study, the expression pattern and prognostic value of CFP in pan-cancer were analyzed via the Oncomine, PrognoScan, GEPIA and Kaplan-Meier plotters. In addition, we used immunohistochemical staining to validate CFP expression in clinical tissue samples. Finally, we evaluated the correlations between CFP and cancer immune infiltrates particularly in stomach adenocarcinoma (STAD) and lung adenocarcinoma (LUAD) by using GEPIA and TIMER databases. The results of database analysis and immunohistochemistry showed that the expression level of CFP in STAD and LUAD was lower than that in normal tissues. Low expression level of CFP was associated with poorer overall survival (OS), first progression (FP), post progression survival (PPS) and was detrimental to the prognosis of STAD and LUAD, specifically in stage 3, stage T3, stage N2 and N3 of STAD (P<0.05). Moreover, expression of CFP had significant positive correlations with the infiltration levels of CD8+ T cells, CD4+ T cells, macrophages, neutrophils and dendritic cells (DCs) in STAD and LUAD. Furthermore, gene markers of infiltrating immune cells exhibited different CFP-related immune infiltration patterns such as tumor-associated-macrophages (TAMs). These results suggest that CFP can serve as a prognostic biomarker for determining prognosis and immune infiltration in STAD and LUAD.
Collapse
Affiliation(s)
- Guoliang Cui
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.,Institute of Integrated Chinese and Western Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China.,The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210017, Jiangsu, China
| | - Le Geng
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.,Institute of Integrated Chinese and Western Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Li Zhu
- The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210017, Jiangsu, China
| | - Zhenyan Lin
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.,Institute of Integrated Chinese and Western Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Xuan Liu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.,Institute of Integrated Chinese and Western Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Zhengyue Miao
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.,Institute of Integrated Chinese and Western Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Jintao Jiang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.,Institute of Integrated Chinese and Western Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Xiaoke Feng
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.,Institute of Integrated Chinese and Western Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Fei Wei
- Department of Physiology, School of medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| |
Collapse
|
38
|
van de Pavert SA. Lymphoid Tissue inducer (LTi) cell ontogeny and functioning in embryo and adult. Biomed J 2021; 44:123-132. [PMID: 33849806 PMCID: PMC8178546 DOI: 10.1016/j.bj.2020.12.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/05/2020] [Accepted: 12/09/2020] [Indexed: 12/19/2022] Open
Abstract
Innate Lymphoid Cells (ILC) are involved in homeostasis and immunity. Their dynamic differentiation and characterization depend on their tissue of residency and is adapted to their role within these tissues. Lymphoid Tissue inducer (LTi) cells are an ILC member and essential for embryonic lymph node (LN) formation. LNs are formed at pre-defined and strategic positions throughout the body and how LTi cells are initially attracted towards these areas is under debate. Besides their role in LN formation, LTi-like and the closely related ILC type 3 (ILC3) cells have been observed within the embryonic gut. New studies have now shown more information on their origin and differentiation within the embryo. This review will evaluate the embryonic LTi cell origin from a specific embryonic hemogenic wave, which has recently been described in mouse. Moreover, I will discuss their differentiation and similarities with the closely related ILC3 cells in embryo and adult.
Collapse
Affiliation(s)
- Serge A van de Pavert
- Aix-Marseille University, Centre National de la Recherche Scientifique (CNRS), National Institute for Health and Medical Research (INSERM), Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France.
| |
Collapse
|
39
|
Pedersen DV, Pedersen MN, Mazarakis SM, Wang Y, Lindorff-Larsen K, Arleth L, Andersen GR. Properdin oligomers adopt rigid extended conformations supporting function. eLife 2021; 10:63356. [PMID: 33480354 PMCID: PMC7857727 DOI: 10.7554/elife.63356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
Properdin stabilizes convertases formed upon activation of the complement cascade within the immune system. The biological activity of properdin depends on the oligomerization state, but whether properdin oligomers are rigid and how their structure links to function remains unknown. We show by combining electron microscopy and solution scattering, that properdin oligomers adopt extended rigid and well-defined conformations which are well approximated by single models of apparent n-fold rotational symmetry with dimensions of 230–360 Å. Properdin monomers are pretzel-shaped molecules with limited flexibility. In solution, properdin dimers are curved molecules, whereas trimers and tetramers are close to being planar molecules. Structural analysis indicates that simultaneous binding through all binding sites to surface-linked convertases is unlikely for properdin trimer and tetramers. We show that multivalency alone is insufficient for full activity in a cell lysis assay. Hence, the observed rigid extended oligomer structure is an integral component of properdin function.
Collapse
Affiliation(s)
- Dennis V Pedersen
- Department of Molecular Biology and Genetics, Center for Structural Biology, Aarhus University, Aarhus, Denmark
| | - Martin Nors Pedersen
- Structural Biophysics, X-ray and Neutron Science, the Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
| | - Sofia Mm Mazarakis
- Department of Molecular Biology and Genetics, Center for Structural Biology, Aarhus University, Aarhus, Denmark
| | - Yong Wang
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kresten Lindorff-Larsen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Lise Arleth
- Structural Biophysics, X-ray and Neutron Science, the Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
| | - Gregers R Andersen
- Department of Molecular Biology and Genetics, Center for Structural Biology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
40
|
Influenza A Virus Hemagglutinin and Other Pathogen Glycoprotein Interactions with NK Cell Natural Cytotoxicity Receptors NKp46, NKp44, and NKp30. Viruses 2021; 13:v13020156. [PMID: 33494528 PMCID: PMC7911750 DOI: 10.3390/v13020156] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Natural killer (NK) cells are part of the innate immunity repertoire, and function in the recognition and destruction of tumorigenic and pathogen-infected cells. Engagement of NK cell activating receptors can lead to functional activation of NK cells, resulting in lysis of target cells. NK cell activating receptors specific for non-major histocompatibility complex ligands are NKp46, NKp44, NKp30, NKG2D, and CD16 (also known as FcγRIII). The natural cytotoxicity receptors (NCRs), NKp46, NKp44, and NKp30, have been implicated in functional activation of NK cells following influenza virus infection via binding with influenza virus hemagglutinin (HA). In this review we describe NK cell and influenza A virus biology, and the interactions of influenza A virus HA and other pathogen lectins with NK cell natural cytotoxicity receptors (NCRs). We review concepts which intersect viral immunology, traditional virology and glycobiology to provide insights into the interactions between influenza virus HA and the NCRs. Furthermore, we provide expert opinion on future directions that would provide insights into currently unanswered questions.
Collapse
|
41
|
Mangogna A, Varghese PM, Agostinis C, Alrokayan SH, Khan HA, Stover CM, Belmonte B, Martorana A, Ricci G, Bulla R, Kishore U. Prognostic Value of Complement Properdin in Cancer. Front Immunol 2021; 11:614980. [PMID: 33542722 PMCID: PMC7851055 DOI: 10.3389/fimmu.2020.614980] [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: 10/07/2020] [Accepted: 12/08/2020] [Indexed: 02/05/2023] Open
Abstract
The complement system is readily triggered by the presence of damage-associated molecular patterns on the surface of tumor cells. The complement alternative pathway provides rapid amplification of the molecular stress signal, leading to complement cascade activation to deal with pathogens or malignant cells. Properdin is the only known positive regulator of the alternative pathway. In addition, properdin promotes the phagocytic uptake of apoptotic T cells by macrophages and dendritic cells without activating the complement system, thus, establishing its ability to recognize "altered-self". Dysregulation of properdin has been implicated in substantial tissue damage in the host, and in some cases, chronic unresolved inflammation. A corollary of this may be the development of cancer. Hence, to establish a correlation between properdin presence/levels in normal and cancer tissues, we performed bioinformatics analysis, using Oncomine and UALCAN. Survival analyses were performed using UALCAN and PROGgeneV2 to assess if properdin can serve as a potential prognostic marker for human lung adenocarcinoma (LUAD), liver hepatocellular carcinoma (LIHC), cervical squamous cell carcinoma (CESC), and pancreatic adenocarcinoma (PAAD). We also analyzed levels of tumor-infiltrating immune cells using TIMER, a tool for characterizing immune cell composition in cancers. We found that in LUAD and LIHC, there was a lower expression of properdin in the tumors compared to normal tissues, while no significant difference was observed in CESC and PAAD. Survival analysis demonstrated a positive association between properdin mRNA expression and overall survival in all 4 types of cancers. TIMER analysis revealed that properdin expression correlated negatively with tumor purity and positively with levels of infiltrating B cells, cytotoxic CD8+ T cells, CD4+ helper T cells, macrophages, neutrophils and dendritic cells in LUAD, CESC and PAAD, and with levels of B cells, CD8+ T cells and dendritic cells in LIHC. Immunohistochemical analysis revealed that infiltrating immune cells were the most likely source of properdin in the tumor microenvironment. Thus, complement protein properdin shows promise as a prognostic marker in cancer and warrants further study.
Collapse
Affiliation(s)
- Alessandro Mangogna
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Burlo Garofolo, Trieste, Italy
| | - Praveen M. Varghese
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Chiara Agostinis
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Burlo Garofolo, Trieste, Italy
| | - Salman H. Alrokayan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Haseeb A. Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Cordula M. Stover
- School of Biological Sciences, University of Leicester, Leicester, United Kingdom
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Anna Martorana
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Burlo Garofolo, Trieste, Italy
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| |
Collapse
|
42
|
Duev-Cohen A, Isaacson B, Berhani O, Charpak-Amikam Y, Friedman N, Drori Y, Mandelboim M, Mandelboim O. Altered NKp46 Recognition and Elimination of Influenza B Viruses. Viruses 2020; 13:v13010034. [PMID: 33375516 PMCID: PMC7824211 DOI: 10.3390/v13010034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/17/2020] [Accepted: 12/23/2020] [Indexed: 11/23/2022] Open
Abstract
Every year, millions of people worldwide are infected with influenza, causing enormous health and economic problems. The most common type of influenza is influenza A. It is known that Natural Killer (NK) cells play an important role in controlling influenza A infection, mostly through the recognition of the viral protein hemagglutinin (HA) by the activating receptor, NKp46. In contrast, little is known regarding NK cell recognition of influenza B viruses, even though they are responsible for a third of all pediatric influenza deaths and are therefore included in the seasonal vaccine each year. Here we show that NKp46 also recognizes influenza B viruses. We show that NKp46 binds the HA protein of influenza B in a sialic acid-dependent manner, and identified the glycosylated residue in NKp46, which is critical for this interaction. We discovered that this interaction has a binding affinity approximately seven times lower than NKp46 binding of influenza A’s HA. Finally, we demonstrated, using mice deficient for the mouse orthologue of NKp46, named NCR1, that NKp46 is not important for influenza B elimination. These findings enable us to better understand the interactions between the different influenza viruses and NK cells that are known to be crucial for viral elimination.
Collapse
Affiliation(s)
- Alexandra Duev-Cohen
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, The Hebrew University Hadassah Medical School, Jerusalem 9112001, Israel; (A.D.-C.); (B.I.); (O.B.); (Y.C.-A.)
| | - Batya Isaacson
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, The Hebrew University Hadassah Medical School, Jerusalem 9112001, Israel; (A.D.-C.); (B.I.); (O.B.); (Y.C.-A.)
| | - Orit Berhani
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, The Hebrew University Hadassah Medical School, Jerusalem 9112001, Israel; (A.D.-C.); (B.I.); (O.B.); (Y.C.-A.)
| | - Yoav Charpak-Amikam
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, The Hebrew University Hadassah Medical School, Jerusalem 9112001, Israel; (A.D.-C.); (B.I.); (O.B.); (Y.C.-A.)
| | - Nehemya Friedman
- Central Virology Laboratory, Ministry of Health, Public Health Services, Chaim Sheba Medical Center, Tel Hashomer, Ramat-Gan 5265601, Israel; (N.F.); (Y.D.); (M.M.)
| | - Yaron Drori
- Central Virology Laboratory, Ministry of Health, Public Health Services, Chaim Sheba Medical Center, Tel Hashomer, Ramat-Gan 5265601, Israel; (N.F.); (Y.D.); (M.M.)
| | - Michal Mandelboim
- Central Virology Laboratory, Ministry of Health, Public Health Services, Chaim Sheba Medical Center, Tel Hashomer, Ramat-Gan 5265601, Israel; (N.F.); (Y.D.); (M.M.)
| | - Ofer Mandelboim
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, The Hebrew University Hadassah Medical School, Jerusalem 9112001, Israel; (A.D.-C.); (B.I.); (O.B.); (Y.C.-A.)
- Correspondence:
| |
Collapse
|
43
|
Ferretti E, Carlomagno S, Pesce S, Muccio L, Obino V, Greppi M, Solari A, Setti C, Marcenaro E, Della Chiesa M, Sivori S. Role of the Main Non HLA-Specific Activating NK Receptors in Pancreatic, Colorectal and Gastric Tumors Surveillance. Cancers (Basel) 2020; 12:E3705. [PMID: 33321719 PMCID: PMC7763095 DOI: 10.3390/cancers12123705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022] Open
Abstract
Human NK cells can control tumor growth and metastatic spread thanks to their powerful cytolytic activity which relies on the expression of an array of activating receptors. Natural cytotoxicity receptors (NCRs) NKG2D and DNAM-1 are those non-HLA-specific activating NK receptors that are mainly involved in sensing tumor transformation by the recognition of different ligands, often stress-induced molecules, on the surface of cancer cells. Tumors display several mechanisms aimed at dampening/evading NK-mediated responses, a relevant fraction of which is based on the downregulation of the expression of activating receptors and/or their ligands. In this review, we summarize the role of the main non-HLA-specific activating NK receptors, NCRs, NKG2D and DNAM-1, in controlling tumor growth and metastatic spread in solid malignancies affecting the gastrointestinal tract with high incidence in the world population, i.e., pancreatic ductal adenocarcinoma (PDAC), colorectal cancer (CRC), and gastric cancer (GC), also describing the phenotypic and functional alterations induced on NK cells by their tumor microenvironment.
Collapse
Affiliation(s)
- Elisa Ferretti
- Centro di Eccellenza per la Ricerca Biomedica, University of Genoa, 16132 Genoa, Italy;
| | - Simona Carlomagno
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Silvia Pesce
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Letizia Muccio
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Valentina Obino
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Marco Greppi
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Agnese Solari
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Chiara Setti
- Dipartimento di Medicina Sperimentale (DIMES), University of Genoa, 16132 Genoa, Italy; (S.C.); (S.P.); (L.M.); (V.O.); (M.G.); (A.S.); (C.S.)
| | - Emanuela Marcenaro
- Dipartimento di Medicina Sperimentale (DIMES) and Centro di Eccellenza per la Ricerca Biomedica, University of Genoa, 16132 Genoa, Italy;
| | - Mariella Della Chiesa
- Dipartimento di Medicina Sperimentale (DIMES) and Centro di Eccellenza per la Ricerca Biomedica, University of Genoa, 16132 Genoa, Italy;
| | - Simona Sivori
- Dipartimento di Medicina Sperimentale (DIMES) and Centro di Eccellenza per la Ricerca Biomedica, University of Genoa, 16132 Genoa, Italy;
| |
Collapse
|
44
|
Cantoni C, Wurzer H, Thomas C, Vitale M. Escape of tumor cells from the NK cell cytotoxic activity. J Leukoc Biol 2020; 108:1339-1360. [PMID: 32930468 DOI: 10.1002/jlb.2mr0820-652r] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022] Open
Abstract
In recent years, NK cells, initially identified as potent cytotoxic effector cells, have revealed an unexpected complexity, both at phenotypic and functional levels. The discovery of different NK cell subsets, characterized by distinct gene expression and phenotypes, was combined with the characterization of the diverse functions NK cells can exert, not only as circulating cells, but also as cells localized or recruited in lymphoid organs and in multiple tissues. Besides the elimination of tumor and virus-infected cells, these functions include the production of cytokines and chemokines, the regulation of innate and adaptive immune cells, the influence on tissue homeostasis. In addition, NK cells display a remarkable functional plasticity, being able to adapt to the environment and to develop a kind of memory. Nevertheless, the powerful cytotoxic activity of NK cells remains one of their most relevant properties, particularly in the antitumor response. In this review, the process of tumor cell recognition and killing mediated by NK cells, starting from the generation of cytolytic granules and recognition of target cell, to the establishment of the NK cell immunological synapse, the release of cytotoxic molecules, and consequent tumor cell death is described. Next, the review focuses on the heterogeneous mechanisms, either intrinsic to tumors or induced by the tumor microenvironment, by which cancer cells can escape the NK cell-mediated attack.
Collapse
Affiliation(s)
- Claudia Cantoni
- Department of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.,Laboratory of Clinical and Experimental Immunology, Integrated Department of Services and Laboratories, IRCCS Istituto G. Gaslini, Genoa, Italy
| | - Hannah Wurzer
- Cytoskeleton and Cancer Progression, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg.,Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Clément Thomas
- Cytoskeleton and Cancer Progression, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Massimo Vitale
- UO Immunologia, IRCCS Ospedale Policlinico San Martino Genova, Genoa, Italy
| |
Collapse
|
45
|
Sivori S, Pende D, Quatrini L, Pietra G, Della Chiesa M, Vacca P, Tumino N, Moretta F, Mingari MC, Locatelli F, Moretta L. NK cells and ILCs in tumor immunotherapy. Mol Aspects Med 2020; 80:100870. [PMID: 32800530 DOI: 10.1016/j.mam.2020.100870] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/05/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023]
Abstract
Cells of the innate immunity play an important role in tumor immunotherapy. Thus, NK cells can control tumor growth and metastatic spread. Thanks to their strong cytolytic activity against tumors, different approaches have been developed for exploiting/harnessing their function in patients with leukemia or solid tumors. Pioneering trials were based on the adoptive transfer of autologous NK cell-enriched cell populations that were expanded in vitro and co-infused with IL-2. Although relevant results were obtained in patients with advanced melanoma, the effect was mostly limited to certain metastatic localizations, particularly to the lung. In addition, the severe IL-2-related toxicity and the preferential IL-2-induced expansion of Treg limited this type of approach. This limitation may be overcome by the use of IL-15, particularly of modified IL-15 molecules to improve its half-life and optimize the biological effects. Other approaches to harness NK cell function include stimulation via TLR, the use of bi- and tri-specific NK cell engagers (BiKE and TriKE) linking activating NK receptors (e.g. CD16) to tumor-associated antigens and even incorporating an IL-15 moiety (TriKE). As recently shown, in tumor patients, NK cells may also express inhibitory checkpoints, primarily PD-1. Accordingly, the therapeutic use of checkpoint inhibitors may unleash NK cells against PD-L1+ tumors. This effect may be predominant and crucial in tumors that have lost HLA cl-I expression, thus resulting "invisible" to T lymphocytes. Additional approaches in which NK cells may represent an important tool for cancer therapy, are to exploit the unique properties of the "adaptive" NK cells. These CD57+ NKG2C+ cells, despite their mature stage and a potent cytolytic activity, maintain a strong proliferating capacity. This property revealed to be crucial in hematopoietic stem cell transplantation (HSCT), particularly in the haplo-HSCT setting, to cure high-risk leukemias. T depleted haplo-HSCT (e.g. from one of the parents) allowed to save the life of thousands of patients lacking a HLA-compatible donor. In this setting, NK cells have been shown to play an essential role against leukemia cells and infections. Another major advance is represented by chimeric antigen receptor (CAR)-engineered NK cells. CAR-NK, different from CAR-T cells, may be obtained from allogeneic donors since they do not cause GvHD. Accordingly, they may represent "off-the-shelf" products to promptly treat tumor patients, with affordable costs. Different from NK cells, helper ILC (ILC1, ILC2 and ILC3), the innate counterpart of T helper cell subsets, remain rather ambiguous with respect to their anti-tumor activity. A possible exception is represented by a subset of ILC3: their frequency in peri-tumoral tissues in patients with NSCLC directly correlates with a better prognosis, possibly reflecting their ability to contribute to the organization of tertiary lymphoid structures, an important site of T cell-mediated anti-tumor responses. It is conceivable that innate immunity may significantly contribute to the major advances that immunotherapy has ensured and will continue to ensure to the cure of cancer.
Collapse
Affiliation(s)
- Simona Sivori
- Department of Experimental Medicine, University of Genoa, Italy; Centre of Excellence for Biomedical Research, University of Genoa, Italy
| | - Daniela Pende
- UO Immunologia, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Linda Quatrini
- Department of Immunology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Gabriella Pietra
- Department of Experimental Medicine, University of Genoa, Italy; UO Immunologia, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Mariella Della Chiesa
- Department of Experimental Medicine, University of Genoa, Italy; Centre of Excellence for Biomedical Research, University of Genoa, Italy
| | - Paola Vacca
- Department of Immunology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Nicola Tumino
- Department of Immunology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Francesca Moretta
- Department of Laboratory Medicine, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy
| | - Maria Cristina Mingari
- Department of Experimental Medicine, University of Genoa, Italy; UO Immunologia, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Franco Locatelli
- Department of Hematology/Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy; Department of Gynecology/Obstetrics and Pediatrics, Sapienza University, Rome, Italy
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy.
| |
Collapse
|
46
|
Patrizi O, Rampinelli F, Coltrini D, Pesce S, Carlomagno S, Sivori S, Pascale A, Marcenaro E, Parolini S, Tabellini G. Natural killer cell impairment in ovarian clear cell carcinoma. J Leukoc Biol 2020; 108:1425-1434. [PMID: 32794325 DOI: 10.1002/jlb.5ma0720-295r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022] Open
Abstract
In the present study, we report the analysis of NK cells derived from patients suffering from a rare ovarian cancer histotype of clear cell carcinoma (OCCC) resistant to conventional chemotherapies. We analyzed the phenotype of NK cells derived from peripheral blood (PB) and peritoneal fluid (PF) and evaluated cytotoxic interactions between NK cells and autologous tumor cells (ATC) derived from patients. We provided evidence of impaired degranulation capacity of NK cells derived from patients' PF in the presence of ATC. Analyzing tumor cell ligands recognized by NK cell receptors, we found that ATC are characterized by an HLA class I+ phenotype (although the level of HLA-I expression varies among all patients) and by a heterogeneous expression of ligands for activating NK receptors (from normal to decreased expression of some markers). Furthermore, we observed a down-regulation of crucial NK cell activating receptors, primarily DNAX Accessory Molecule-1 (DNAM-1), on tumor-associated NK cells. Based on these results, we propose that this severe lysis defect may be due to both negative interactions between HLA-I-specific inhibitory NK cell receptors/HLA-I molecules and to defective interactions between activating NK receptors and cognate ligands. In conclusion, for the first time, the phenotypic and functional properties of tumor-associated NK cells and their ATC derived from PF of patients with advanced stage of OCCC were characterized. Taken together results indicate altered interactions between NK cells and ATC and shed light on the aggressive mechanisms of this cancer histotype. Further studies on this rare tumor will be helpful to improve and define more effective therapies.
Collapse
Affiliation(s)
- Ornella Patrizi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Fabio Rampinelli
- Department of Obstetrics and Gynecology, Spedali Civili di Brescia, Brescia, Italy
| | - Daniela Coltrini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Silvia Pesce
- Department of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Simona Carlomagno
- Department of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Simona Sivori
- Department of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | | | - Emanuela Marcenaro
- Department of Experimental Medicine and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Silvia Parolini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giovanna Tabellini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
47
|
Wu SY, Fu T, Jiang YZ, Shao ZM. Natural killer cells in cancer biology and therapy. Mol Cancer 2020; 19:120. [PMID: 32762681 PMCID: PMC7409673 DOI: 10.1186/s12943-020-01238-x] [Citation(s) in RCA: 477] [Impact Index Per Article: 95.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment is highly complex, and immune escape is currently considered an important hallmark of cancer, largely contributing to tumor progression and metastasis. Named for their capability of killing target cells autonomously, natural killer (NK) cells serve as the main effector cells toward cancer in innate immunity and are highly heterogeneous in the microenvironment. Most current treatment options harnessing the tumor microenvironment focus on T cell-immunity, either by promoting activating signals or suppressing inhibitory ones. The limited success achieved by T cell immunotherapy highlights the importance of developing new-generation immunotherapeutics, for example utilizing previously ignored NK cells. Although tumors also evolve to resist NK cell-induced cytotoxicity, cytokine supplement, blockade of suppressive molecules and genetic engineering of NK cells may overcome such resistance with great promise in both solid and hematological malignancies. In this review, we summarized the fundamental characteristics and recent advances of NK cells within tumor immunometabolic microenvironment, and discussed potential application and limitations of emerging NK cell-based therapeutic strategies in the era of presicion medicine.
Collapse
Affiliation(s)
- Song-Yang Wu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Tong Fu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi-Zhou Jiang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
48
|
Cantoni C, Granata S, Bruschi M, Spaggiari GM, Candiano G, Zaza G. Recent Advances in the Role of Natural Killer Cells in Acute Kidney Injury. Front Immunol 2020; 11:1484. [PMID: 32903887 PMCID: PMC7438947 DOI: 10.3389/fimmu.2020.01484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/08/2020] [Indexed: 01/18/2023] Open
Abstract
Growing evidence is revealing a central role for natural killer (NK) cells, cytotoxic cells belonging to the broad family of innate lymphoid cells (ILCs), in acute and chronic forms of renal disease. NK cell effector functions include both the recognition and elimination of virus-infected and tumor cells and the capability of sensing pathogens through Toll-like receptor (TLR) engagement. Notably, they also display immune regulatory properties, exerted thanks to their ability to secrete cytokines/chemokines and to establish interactions with different innate and adaptive immune cells. Therefore, because of their multiple functions, NK cells may have a major pathogenic role in acute kidney injury (AKI), and a better understanding of the molecular mechanisms driving NK cell activation in AKI and their downstream interactions with intrinsic renal cells and infiltrating immune cells could help to identify new potential biomarkers and to select clinically valuable novel therapeutic targets. In this review, we discuss the current literature regarding the potential involvement of NK cells in AKI.
Collapse
Affiliation(s)
- Claudia Cantoni
- Laboratory of Clinical and Experimental Immunology, Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, Genoa, Italy.,Department of Experimental Medicine (DIMES) and Center of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Simona Granata
- Renal Unit, Department of Medicine, University-Hospital of Verona, Verona, Italy
| | - Maurizio Bruschi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Grazia Maria Spaggiari
- Department of Experimental Medicine (DIMES) and Center of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona, Verona, Italy
| |
Collapse
|
49
|
Delauzun V, Amigues B, Gaubert A, Leone P, Grange M, Gauthier L, Roussel A. Extracellular vesicles as a platform to study cell-surface membrane proteins. Methods 2020; 180:35-44. [PMID: 32156657 DOI: 10.1016/j.ymeth.2020.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 01/08/2023] Open
Abstract
Producing intact recombinant membrane proteins for structural studies is an inherently challenging task due to their requirement for a cell-lipid environment. Most of the procedures developed involve isolating the protein by solubilization with detergent and further reconstitutions into artificial membranes. These procedures are highly time consuming and suffer from further drawbacks, including low yields and high cost. We describe here an alternative method for rapidly obtaining recombinant cell-surface membrane proteins displayed on extracellular vesicles (EVs) derived from cells in culture. Interaction between these membrane proteins and ligands can be analyzed directly on EVs. Moreover, EVs can also be used for protein structure determination or immunization purposes.
Collapse
Affiliation(s)
- Vincent Delauzun
- Architecture et Fonction des Macromolécules Biologiques (AFMB), CNRS, Aix-Marseille Université, UMR 7257, 163 Avenue de Luminy, Case 932, 13009 Marseille, France
| | - Beatrice Amigues
- Architecture et Fonction des Macromolécules Biologiques (AFMB), CNRS, Aix-Marseille Université, UMR 7257, 163 Avenue de Luminy, Case 932, 13009 Marseille, France
| | - Anais Gaubert
- Architecture et Fonction des Macromolécules Biologiques (AFMB), CNRS, Aix-Marseille Université, UMR 7257, 163 Avenue de Luminy, Case 932, 13009 Marseille, France
| | - Philippe Leone
- Architecture et Fonction des Macromolécules Biologiques (AFMB), CNRS, Aix-Marseille Université, UMR 7257, 163 Avenue de Luminy, Case 932, 13009 Marseille, France
| | - Magali Grange
- Architecture et Fonction des Macromolécules Biologiques (AFMB), CNRS, Aix-Marseille Université, UMR 7257, 163 Avenue de Luminy, Case 932, 13009 Marseille, France
| | | | - Alain Roussel
- Architecture et Fonction des Macromolécules Biologiques (AFMB), CNRS, Aix-Marseille Université, UMR 7257, 163 Avenue de Luminy, Case 932, 13009 Marseille, France.
| |
Collapse
|
50
|
Mellors J, Tipton T, Longet S, Carroll M. Viral Evasion of the Complement System and Its Importance for Vaccines and Therapeutics. Front Immunol 2020; 11:1450. [PMID: 32733480 PMCID: PMC7363932 DOI: 10.3389/fimmu.2020.01450] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/04/2020] [Indexed: 12/17/2022] Open
Abstract
The complement system is a key component of innate immunity which readily responds to invading microorganisms. Activation of the complement system typically occurs via three main pathways and can induce various antimicrobial effects, including: neutralization of pathogens, regulation of inflammatory responses, promotion of chemotaxis, and enhancement of the adaptive immune response. These can be vital host responses to protect against acute, chronic, and recurrent viral infections. Consequently, many viruses (including dengue virus, West Nile virus and Nipah virus) have evolved mechanisms for evasion or dysregulation of the complement system to enhance viral infectivity and even exacerbate disease symptoms. The complement system has multifaceted roles in both innate and adaptive immunity, with both intracellular and extracellular functions, that can be relevant to all stages of viral infection. A better understanding of this virus-host interplay and its contribution to pathogenesis has previously led to: the identification of genetic factors which influence viral infection and disease outcome, the development of novel antivirals, and the production of safer, more effective vaccines. This review will discuss the antiviral effects of the complement system against numerous viruses, the mechanisms employed by these viruses to then evade or manipulate this system, and how these interactions have informed vaccine/therapeutic development. Where relevant, conflicting findings and current research gaps are highlighted to aid future developments in virology and immunology, with potential applications to the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Jack Mellors
- Public Health England, National Infection Service, Salisbury, United Kingdom.,Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Tom Tipton
- Public Health England, National Infection Service, Salisbury, United Kingdom
| | - Stephanie Longet
- Public Health England, National Infection Service, Salisbury, United Kingdom
| | - Miles Carroll
- Public Health England, National Infection Service, Salisbury, United Kingdom
| |
Collapse
|