1
|
Pan J, Fu B, Huang S, Jiang Y, Zhou X, Zhou M, Yu Z, Geng X, Zhu Y, Zheng H, Gong Y, Huang D, Guo L. Downregulated granzyme M expression: implications for the immune system and prognosis of thyroid cancer. Gene 2025; 961:149494. [PMID: 40228756 DOI: 10.1016/j.gene.2025.149494] [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: 11/26/2024] [Revised: 03/20/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025]
Abstract
Thyroid carcinoma (THCA), the most prevalent endocrine system cancer, is rising worldwide. Granzyme family member GZMM causes tumor cell inflammation and programmed cell death. However, the role of GZMM in THCA has not been investigated. Bioinformatics analysis and assays showed that THCA GZMM protein levels were down-regulated and associated with certain clinicopathological features. Additionally, univariate and multivariate Cox analysis and ROC curve analysis showed that low GZMM expression was related with poor overall survival and might be employed as a prognostic and diagnostic factor. We used functional tests to examine how GZMM affected angiogenesis, invasion, and migration in vitro. TIMER showed a link between GZMM expression, immune cell infiltration, and tumor purity. Overexpression of GZMM also greatly boosted CD8 + T cell-attracting chemokines. TCGA data analysis yielded a GZMM and epigenetic modification-related gene risk prediction model. Overall, GZMM inhibits tumor invasion, migration, and angiogenesis, and is closely related to the immune microenvironment, significantly associated with poor prognosis in THCA patients.
Collapse
Affiliation(s)
- Jingying Pan
- Department of Ultrasonography, Second Affiliated Hospital of Nanchang University, Nanchang, China; First College of Clinical Medicine, Nanchang University, China
| | - Bidong Fu
- Second College of Clinical Medicine, Nanchang University, China
| | - Shuhan Huang
- Second College of Clinical Medicine, Nanchang University, China
| | - Yike Jiang
- Second College of Clinical Medicine, Nanchang University, China
| | - Xuanrui Zhou
- Second College of Clinical Medicine, Nanchang University, China
| | - Minqin Zhou
- Second College of Clinical Medicine, Nanchang University, China
| | - Zichuan Yu
- Second College of Clinical Medicine, Nanchang University, China
| | - Xitong Geng
- Second College of Clinical Medicine, Nanchang University, China
| | | | - Hao Zheng
- Second College of Clinical Medicine, Nanchang University, China
| | - Yiyang Gong
- Second College of Clinical Medicine, Nanchang University, China
| | - Da Huang
- Department of Thyroid Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Liangyun Guo
- Department of Ultrasonography, Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
2
|
Kim NH, Sim SJ, Han HG, Yoon JH, Han YH. Immunosenescence and age-related immune cells: causes of age-related diseases. Arch Pharm Res 2025; 48:132-149. [PMID: 39725853 DOI: 10.1007/s12272-024-01529-7] [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: 06/24/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Immunosenescence is a weakening of the immune system due to aging, characterized by changes in immune cells and dysregulated immune function. Age-related immune cells are increasing with aging. They are associated with chronic prolonged inflammation, causing tissue dysfunction and age-related diseases. Here, we discuss increased pro-inflammatory activity of aged macrophages, accumulation of lymphocytes with an age-associated phenotype, and specific alterations in both functions and characteristics of these immune cells. These cellular changes are associated with development of age-related diseases. Additionally, we reviewed various therapeutic strategies targeting age-related immunosenescence, providing pathways to mitigate effects of age-related diseases.
Collapse
Affiliation(s)
- Nam-Hee Kim
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, 1, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - So-Jin Sim
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, 1, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Hong-Gyu Han
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, 1, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Jeong-Hyuk Yoon
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, 1, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, South Korea
| | - Yong-Hyun Han
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, 1, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, South Korea.
- Multidimentional Genomics Research Center, Kangwon National University, Chuncheon, 24341, South Korea.
| |
Collapse
|
3
|
Ni Y, Zhong L, Li Y, Zhang Z, Ming B, Qing Y, Zhang Q. Exploration of molecular biomarkers in ankylosing spondylitis transcriptomics. Front Immunol 2024; 15:1480492. [PMID: 39759509 PMCID: PMC11695275 DOI: 10.3389/fimmu.2024.1480492] [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: 08/14/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Background Inflammation of the spine and sacroiliac joints is a hallmark of the chronic, progressive inflammatory illness known as ankylosing spondylitis (AS). The insidious onset and non-specific early symptoms of AS often lead to delays in diagnosis and treatment, which may result in the onset of disability. It is therefore imperative to identify new biomarkers. Methods In this study, datasets GSE73754 and GSE25101 were derived from the Gene Expression Omnibus (GEO). Key genes were identified through differential expression analysis and weighted gene co-expression network analysis (WGCNA). A model was then established using LASSO regression, and then it was subjected to the receiver operating characteristic (ROC) curve analysis for evaluation of the diagnostic accuracy of the genes. Subsequently, immune infiltration analysis was conducted to demonstrate the immune infiltration status of the samples and the correlation between key genes and immune infiltration. Finally, the expression levels of key genes in peripheral blood mononuclear cells (PBMCs) and their correlation with clinical indicators were validated via RT-qPCR assay. Results Through WGCNA and differential expression analysis, 6 genes were identified. Ultimately, five key genes (ACSL1, SLC40A1, GZMM, TRIB1, XBP1) were determined using LASSO regression. The area under the ROC curve (AUC) for these genes was greater than 0.7, indicating favorable diagnostic performance. Immune infiltration analysis showed that AS was associated with infiltration levels of various immune cells. RT-qPCR validated that the expression of ACSL1, SLC40A1, GZMM, and XBP1 was consistent with the predictive model, whereas TRIB1 expression was contrary to the predictive model. Clinical correlation analysis of key genes revealed that ACSL1 was positively linked to hsCRP levels, GZMM was negatively linked to, hsCRP levels, and neutrophil absolute values, SLC40A1 was positively linked to ESR, hsCRP levels and neutrophil absolute values, and XBP1 was negatively linked to ESR, hsCRP levels, and neutrophil absolute values. Conclusion This study identified key genes that may reveal a potential association between AS and ferroptosis, demonstrating high diagnostic value. Furthermore, the expression levels of these genes in peripheral blood mononuclear cells (PBMCs) are strongly correlated with disease activity. These findings not only suggest potential biomarkers for the diagnosis of AS but also offer important references for exploring new therapeutic targets, highlighting their substantial clinical applicability.
Collapse
Affiliation(s)
- Yuanpiao Ni
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Linrui Zhong
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yanhui Li
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Zeng Zhang
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Geriatrics, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Bin Ming
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yufeng Qing
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Rheumatology and Immunology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Quanbo Zhang
- Research Center of Hyperuricemia and Gout, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- Department of Geriatrics, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
4
|
Li Z, Xie Q, Zhao F, Huo X, Ren D, Liu Z, Zhou X, Shen G, Zhao J. Exploring GZMK as a prognostic marker and predictor of immunotherapy response in breast cancer: unveiling novel insights into treatment outcomes. J Cancer Res Clin Oncol 2024; 150:286. [PMID: 38833021 PMCID: PMC11150209 DOI: 10.1007/s00432-024-05791-6] [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: 03/01/2024] [Accepted: 05/08/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Granzyme K (GZMK) is a crucial mediator released by immune cells to eliminate tumor cells, playing significant roles in inflammation and tumorigenesis. Despite its importance, the specific role of GZMK in breast cancer and its mechanisms are not well understood. METHODS We utilized data from the TCGA and GEO databases and employed a range of analytical methods including GO, KEGG, GSEA, ssGSEA, and PPI to investigate the impact of GZMK on breast cancer. In vitro studies, including RT-qPCR, CCK-8 assay, cell cycle experiments, apoptosis assays, Celigo scratch assays, Transwell assays, and immunohistochemical methods, were conducted to validate the effects of GZMK on breast cancer cells. Additionally, Cox regression analysis integrating TCGA and our clinical data was used to develop an overall survival (OS) prediction model. RESULTS Analysis of clinical pathological features revealed significant correlations between GZMK expression and lymph node staging, differentiation grade, and molecular breast cancer subtypes. High GZMK expression was associated with improved OS, progression-free survival (PFS), and recurrence-free survival (RFS), as confirmed by multifactorial Cox regression analysis. Functional and pathway enrichment analyses of genes positively correlated with GZMK highlighted involvement in lymphocyte differentiation, T cell differentiation, and T cell receptor signaling pathways. A robust association between GZMK expression and T cell presence was noted in the breast cancer tumor microenvironment (TME), with strong correlations with ESTIMATEScore (Cor = 0.743, P < 0.001), ImmuneScore (Cor = 0.802, P < 0.001), and StromalScore (Cor = 0.516, P < 0.001). GZMK also showed significant correlations with immune checkpoint molecules, including CTLA4 (Cor = 0.856, P < 0.001), PD-1 (Cor = 0.82, P < 0.001), PD-L1 (Cor = 0.56, P < 0.001), CD48 (Cor = 0.75, P < 0.001), and CCR7 (Cor = 0.856, P < 0.001). Studies indicated that high GZMK expression enhances patient responsiveness to immunotherapy, with higher levels observed in responsive patients compared to non-responsive ones. In vitro experiments confirmed that GZMK promotes cell proliferation, cell division, apoptosis, cell migration, and invasiveness (P < 0.05). CONCLUSION Our study provides insights into the differential expression of GZMK in breast cancer and its potential mechanisms in breast cancer pathogenesis. Elevated GZMK expression is associated with improved OS and RFS, suggesting its potential as a prognostic marker for breast cancer survival and as a predictor of the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Zitao Li
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining, 810000, China
- Breast Disease Diagnosis and Treatment Center of Qinghai University Affiliated Hospital & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Qiqi Xie
- Breast Disease Diagnosis and Treatment Center of Qinghai University Affiliated Hospital & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Fuxing Zhao
- Breast Disease Diagnosis and Treatment Center of Qinghai University Affiliated Hospital & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Xinfa Huo
- Breast Disease Diagnosis and Treatment Center of Qinghai University Affiliated Hospital & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Dengfeng Ren
- Breast Disease Diagnosis and Treatment Center of Qinghai University Affiliated Hospital & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Zhilin Liu
- Breast Disease Diagnosis and Treatment Center of Qinghai University Affiliated Hospital & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Xiaofeng Zhou
- Pathology Department, Affiliated Hospital of Qinghai University, Xining, 810000, China
| | - Guoshuang Shen
- Breast Disease Diagnosis and Treatment Center of Qinghai University Affiliated Hospital & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China.
| | - Jiuda Zhao
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining, 810000, China.
- Breast Disease Diagnosis and Treatment Center of Qinghai University Affiliated Hospital & Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China.
| |
Collapse
|
5
|
Jonsson AH. Granzyme K + CD8 T cells in autoimmunity. Best Pract Res Clin Rheumatol 2024; 38:101930. [PMID: 38307763 PMCID: PMC11291703 DOI: 10.1016/j.berh.2024.101930] [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: 01/03/2024] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/04/2024]
Abstract
CD8 T cells expressing granzyme K are enriched in synovial tissue from patients with rheumatoid arthritis and in tissues affected by several other autoimmune diseases. The roles these cells play in autoimmune disease is under active investigation, and several recent studies have begun to shed light on this question. Putting this cell type into functional perspective is especially important given their enrichment at the sites of disease. This review summarizes available evidence for the presence of CD8 T cells and other granzyme K-expressing cells in tissues in autoimmune diseases and discusses the effects these cells may have on the pathogenesis of autoimmune conditions.
Collapse
Affiliation(s)
- Anna Helena Jonsson
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA.
| |
Collapse
|
6
|
Cigalotto L, Martinvalet D. Granzymes in health and diseases: the good, the bad and the ugly. Front Immunol 2024; 15:1371743. [PMID: 38646541 PMCID: PMC11026543 DOI: 10.3389/fimmu.2024.1371743] [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: 01/16/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Granzymes are a family of serine proteases, composed of five human members: GA, B, H, M and K. They were first discovered in the 1980s within cytotoxic granules released during NK cell- and T cell-mediated killing. Through their various proteolytic activities, granzymes can trigger different pathways within cells, all of which ultimately lead to the same result, cell death. Over the years, the initial consideration of granzymes as mere cytotoxic mediators has changed due to surprising findings demonstrating their expression in cells other than immune effectors as well as new intracellular and extracellular activities. Additional roles have been identified in the extracellular milieu, following granzyme escape from the immunological synapse or their release by specific cell types. Outside the cell, granzyme activities mediate extracellular matrix alteration via the degradation of matrix proteins or surface receptors. In certain contexts, these processes are essential for tissue homeostasis; in others, excessive matrix degradation and extensive cell death contribute to the onset of chronic diseases, inflammation, and autoimmunity. Here, we provide an overview of both the physiological and pathological roles of granzymes, highlighting their utility while also recognizing how their unregulated presence can trigger the development and/or worsening of diseases.
Collapse
Affiliation(s)
- Lavinia Cigalotto
- Laboratory of Reactive Oxygen Species and Cytotoxic Immunity, Department Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute Of Molecular Medicine (VIMM), Padova, Italy
| | - Denis Martinvalet
- Laboratory of Reactive Oxygen Species and Cytotoxic Immunity, Department Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute Of Molecular Medicine (VIMM), Padova, Italy
| |
Collapse
|
7
|
Ijaz A, Broere F, Rutten VPMG, Jansen CA, Veldhuizen EJA. Perforin and granzyme A release as novel tool to measure NK cell activation in chickens. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 149:105047. [PMID: 37625470 DOI: 10.1016/j.dci.2023.105047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/22/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
Natural killer (NK) cells are cytotoxic lymphocytes that are present in the circulation but also in many organs including spleen and gut, where they play an important role in the defense against infections. Interaction of NK cells with target cells leads to degranulation, which results in the release of perforin and granzymes in the direct vicinity of the target cell. Chicken NK cells have many characteristics similar to their mammalian counterparts and based on similarities with studies on human NK cells, surface expression of CD107 was always presumed to correlate with granule release. However, proof of this degranulation or in fact the actual presence of perforin (PFN) and granzyme A (GrA) in chicken NK cells and their release upon activation is lacking. Therefore, the purpose of the present study was to determine the presence of perforin and granzyme A in primary chicken NK cells and to measure their release upon degranulation, as an additional tool to study the function of chicken NK cells. Using human specific antibodies against PFN and GrA in fluorescent and confocal microscopy resulted in staining in chicken NK cells. The presence of PFN and GrA was also confirmed by Western blot analyses and its gene expression by PCR. Stimulation of NK cells with the pectin SPE6 followed by flow cytometry resulted in reduced levels of intracellular PFN and GrA, suggesting release of PFN and GrA. Expression of PFN and GrA reversely correlated with increased surface expression of the lysosomal marker CD107. Finally it was shown that the supernatant of activated NK cells, containing the NK cell granule content including PFN and GrA, was able to kill Escherichia coli. This study correlates PFN and GrA release to activation of chicken NK cells and establishes an additional tool to study activity of cytotoxic lymphocytes in chickens.
Collapse
Affiliation(s)
- Adil Ijaz
- Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Femke Broere
- Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Victor P M G Rutten
- Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Christine A Jansen
- Cell Biology and Immunology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, the Netherlands
| | - Edwin J A Veldhuizen
- Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
8
|
Duquette D, Harmon C, Zaborowski A, Michelet X, O'Farrelly C, Winter D, Koay HF, Lynch L. Human Granzyme K Is a Feature of Innate T Cells in Blood, Tissues, and Tumors, Responding to Cytokines Rather than TCR Stimulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:633-647. [PMID: 37449888 DOI: 10.4049/jimmunol.2300083] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/02/2023] [Indexed: 07/18/2023]
Abstract
NK cells and CD8 T cells use cytotoxic molecules to kill virally infected and tumor cell targets. While perforin and granzyme B (GzmB) are the most commonly studied lytic molecules, less is known about granzyme K (GzmK). However, this granzyme has been recently associated with improved prognosis in solid tumors. In this study, we show that, in humans, GzmK is predominantly expressed by innate-like lymphocytes, as well as a newly identified population of GzmK+CD8+ non- mucosal-associated invariant T cells with innate-like characteristics. We found that GzmK+ T cells are KLRG1+EOMES+IL-7R+CD62L-Tcf7int, suggesting that they are central memory T and effector memory T cells. Furthermore, GzmK+ cells are absent/low in cord blood, suggesting that GzmK is upregulated with immune experience. Surprisingly, GzmK+ cells respond to cytokine stimuli alone, whereas TCR stimulation downregulates GzmK expression, coinciding with GzmB upregulation. GzmK+ cells have reduced IFN-γ production compared with GzmB+ cells in each T cell lineage. Collectively, this suggests that GzmK+ cells are not naive, and they may be an intermediate memory-like or preterminally differentiated population. GzmK+ cells are enriched in nonlymphoid tissues such as the liver and adipose. In colorectal cancer, GzmK+ cells are enriched in the tumor and can produce IFN-γ, but GzmK+ expression is mutually exclusive with IL-17a production. Thus, in humans, GzmK+ cells are innate memory-like cells that respond to cytokine stimulation alone and may be important effector cells in the tumor.
Collapse
Affiliation(s)
- Danielle Duquette
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Cathal Harmon
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | | | - Xavier Michelet
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Des Winter
- St. Vincent's University Hospital, Dublin, Ireland
| | - Hui-Fern Koay
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Austria
| | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- St. Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
9
|
Zheng Y, Zhao J, Shan Y, Guo S, Schrodi SJ, He D. Role of the granzyme family in rheumatoid arthritis: Current Insights and future perspectives. Front Immunol 2023; 14:1137918. [PMID: 36875082 PMCID: PMC9977805 DOI: 10.3389/fimmu.2023.1137918] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/03/2023] [Indexed: 02/18/2023] Open
Abstract
Rheumatoid arthritis (RA) is a complex autoimmune disease characterized by chronic inflammation that affects synovial tissues of multiple joints. Granzymes (Gzms) are serine proteases that are released into the immune synapse between cytotoxic lymphocytes and target cells. They enter target cells with the help of perforin to induce programmed cell death in inflammatory and tumor cells. Gzms may have a connection with RA. First, increased levels of Gzms have been found in the serum (GzmB), plasma (GzmA, GzmB), synovial fluid (GzmB, GzmM), and synovial tissue (GzmK) of patients with RA. Moreover, Gzms may contribute to inflammation by degrading the extracellular matrix and promoting cytokine release. They are thought to be involved in RA pathogenesis and have the potential to be used as biomarkers for RA diagnosis, although their exact role is yet to be fully elucidated. The purpose of this review was to summarize the current knowledge regarding the possible role of the granzyme family in RA, with the aim of providing a reference for future research on the mechanisms of RA and the development of new therapies.
Collapse
Affiliation(s)
- Yixin Zheng
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Jianan Zhao
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yu Shan
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Shicheng Guo
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Steven J. Schrodi
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Dongyi He
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| |
Collapse
|
10
|
Jonsson AH, Zhang F, Dunlap G, Gomez-Rivas E, Watts GFM, Faust HJ, Rupani KV, Mears JR, Meednu N, Wang R, Keras G, Coblyn JS, Massarotti EM, Todd DJ, Anolik JH, McDavid A, Wei K, Rao DA, Raychaudhuri S, Brenner MB. Granzyme K + CD8 T cells form a core population in inflamed human tissue. Sci Transl Med 2022; 14:eabo0686. [PMID: 35704599 PMCID: PMC9972878 DOI: 10.1126/scitranslmed.abo0686] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
T cell-derived pro-inflammatory cytokines are a major driver of rheumatoid arthritis (RA) pathogenesis. Although these cytokines have traditionally been attributed to CD4 T cells, we have found that CD8 T cells are notably abundant in synovium and make more interferon (IFN)-γ and nearly as much tumor necrosis factor (TNF) as their CD4 T cell counterparts. Furthermore, using unbiased high-dimensional single-cell RNA-seq and flow cytometric data, we found that the vast majority of synovial tissue and synovial fluid CD8 T cells belong to an effector CD8 T cell population characterized by high expression of granzyme K (GzmK) and low expression of granzyme B (GzmB) and perforin. Functional experiments demonstrate that these GzmK+ GzmB+ CD8 T cells are major cytokine producers with low cytotoxic potential. Using T cell receptor repertoire data, we found that CD8 GzmK+ GzmB+ T cells are clonally expanded in synovial tissues and maintain their granzyme expression and overall cell state in blood, suggesting that they are enriched in tissue but also circulate. Using GzmK and GzmB signatures, we found that GzmK-expressing CD8 T cells were also the major CD8 T cell population in the gut, kidney, and coronavirus disease 2019 (COVID-19) bronchoalveolar lavage fluid, suggesting that they form a core population of tissue-associated T cells across diseases and human tissues. We term this population tissue-enriched expressing GzmK or TteK CD8 cells. Armed to produce cytokines in response to both antigen-dependent and antigen-independent stimuli, CD8 TteK cells have the potential to drive inflammation.
Collapse
Affiliation(s)
- A. Helena Jonsson
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Fan Zhang
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
- Center for Data Sciences, Brigham and Women’s Hospital; Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Boston, MA 02115, USA
- Department of Biomedical Informatics, Harvard Medical School; Boston, MA 02115, USA
- Broad Institute of MIT and Harvard; Cambridge, MA 02142, USA
- Division of Rheumatology and the Center for Health Artificial Intelligence, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Garrett Dunlap
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Emma Gomez-Rivas
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Gerald F. M. Watts
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Heather J. Faust
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Karishma Vijay Rupani
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Joseph R. Mears
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
- Center for Data Sciences, Brigham and Women’s Hospital; Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Boston, MA 02115, USA
- Department of Biomedical Informatics, Harvard Medical School; Boston, MA 02115, USA
- Broad Institute of MIT and Harvard; Cambridge, MA 02142, USA
| | - Nida Meednu
- Division of Rheumatology and the Center for Health Artificial Intelligence, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Runci Wang
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Gregory Keras
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Jonathan S. Coblyn
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Elena M. Massarotti
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Derrick J. Todd
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Jennifer H. Anolik
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center; Rochester, NY 14642, USA
| | - Andrew McDavid
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry; Rochester, NY 14642, USA
| | | | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Deepak A. Rao
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Soumya Raychaudhuri
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
- Center for Data Sciences, Brigham and Women’s Hospital; Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Boston, MA 02115, USA
- Department of Biomedical Informatics, Harvard Medical School; Boston, MA 02115, USA
- Broad Institute of MIT and Harvard; Cambridge, MA 02142, USA
- Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, The University of Manchester; Manchester M13 9PT, UK
| | - Michael B. Brenner
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| |
Collapse
|
11
|
Pączek S, Łukaszewicz-Zając M, Mroczko B. Granzymes-Their Role in Colorectal Cancer. Int J Mol Sci 2022; 23:ijms23095277. [PMID: 35563668 PMCID: PMC9104098 DOI: 10.3390/ijms23095277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is among the most common malignancies worldwide. CRC is considered a heterogeneous disease due to various clinical symptoms, biological behaviours, and a variety of mutations. A number of studies demonstrate that as many as 50% of CRC patients have distant metastases at the time of diagnosis. However, despite the fact that social and medical awareness of CRC has increased in recent years and screening programmes have expanded, there is still an urgent need to find new diagnostic tools for early detection of CRC. The effectiveness of the currently used classical tumour markers in CRC diagnostics is very limited. Therefore, new proteins that play an important role in the formation and progression of CRC are being sought. A number of recent studies show the potential significance of granzymes (GZMs) in carcinogenesis. These proteins are released by cytotoxic lymphocytes, which protect the body against viral infection as well specific signalling pathways that ultimately lead to cell death. Some studies suggest a link between GZMs, particularly the expression of Granzyme A, and inflammation. This paper summarises the role of GZMs in CRC pathogenesis through their involvement in the inflammatory process. Therefore, it seems that GZMs could become the focus of research into new CRC biomarkers.
Collapse
Affiliation(s)
- Sara Pączek
- Department of Biochemical Diagnostics, Medical University in Bialystok, 15-269 Bialystok, Poland; (M.Ł.-Z.); (B.M.)
- Correspondence: ; Tel.: +48-85-831-8587
| | - Marta Łukaszewicz-Zając
- Department of Biochemical Diagnostics, Medical University in Bialystok, 15-269 Bialystok, Poland; (M.Ł.-Z.); (B.M.)
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, Medical University in Bialystok, 15-269 Bialystok, Poland; (M.Ł.-Z.); (B.M.)
- Department of Neurodegeneration Diagnostics, Medical University in Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
12
|
van Aalderen MC, van Lier RAW, Hombrink P. How to Reliably Define Human CD8 + T-Cell Subsets: Markers Playing Tricks. Cold Spring Harb Perspect Biol 2021; 13:a037747. [PMID: 33782028 PMCID: PMC8559543 DOI: 10.1101/cshperspect.a037747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In recent years, our understanding about the functional complexity of CD8+ T-cell populations has increased tremendously. The immunology field is now facing challenges to translate these insights into phenotypic definitions that correlate reliably with distinct functional traits. This is key to adequately monitor and understand compound immune responses including vaccination and immunotherapy regimens. Here we will summarize our understanding of the current state in the human CD8+ T-cell subset characterization field. We will address how reliably the currently used cell surface markers are connected to differentiation status and function of particular subsets. By restricting ourselves to CD8+ αβ T cells, we will focus mostly on major histocompatibility complex (MHC) class I-restricted virus- and tumor-specific T cells. This comes with a major advantage as fluorescently labeled peptide-loaded MHC class I multimers have been widely used to identify and characterize these cells.
Collapse
Affiliation(s)
- Michiel C van Aalderen
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Centre (AUMC), Amsterdam 1105 AZ, The Netherlands
| | - Rene A W van Lier
- Adaptive Immunity Laboratory and Landsteiner Laboratory of the AUMC at Sanquin Blood Supply Foundation, Amsterdam 1066 CX, The Netherlands
| | - Pleun Hombrink
- Adaptive Immunity Laboratory and Landsteiner Laboratory of the AUMC at Sanquin Blood Supply Foundation, Amsterdam 1066 CX, The Netherlands
| |
Collapse
|
13
|
Lavergne M, Hernández-Castañeda MA, Mantel PY, Martinvalet D, Walch M. Oxidative and Non-Oxidative Antimicrobial Activities of the Granzymes. Front Immunol 2021; 12:750512. [PMID: 34707614 PMCID: PMC8542974 DOI: 10.3389/fimmu.2021.750512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/23/2021] [Indexed: 01/11/2023] Open
Abstract
Cell-mediated cytotoxicity is an essential immune defense mechanism to fight against viral, bacterial or parasitic infections. Upon recognition of an infected target cell, killer lymphocytes form an immunological synapse to release the content of their cytotoxic granules. Cytotoxic granules of humans contain two membrane-disrupting proteins, perforin and granulysin, as well as a homologous family of five death-inducing serine proteases, the granzymes. The granzymes, after delivery into infected host cells by the membrane disrupting proteins, may contribute to the clearance of microbial pathogens through different mechanisms. The granzymes can induce host cell apoptosis, which deprives intracellular pathogens of their protective niche, therefore limiting their replication. However, many obligate intracellular pathogens have evolved mechanisms to inhibit programed cells death. To overcome these limitations, the granzymes can exert non-cytolytic antimicrobial activities by directly degrading microbial substrates or hijacked host proteins crucial for the replication or survival of the pathogens. The granzymes may also attack factors that mediate microbial virulence, therefore directly affecting their pathogenicity. Many mechanisms applied by the granzymes to eliminate infected cells and microbial pathogens rely on the induction of reactive oxygen species. These reactive oxygen species may be directly cytotoxic or enhance death programs triggered by the granzymes. Here, in the light of the latest advances, we review the antimicrobial activities of the granzymes in regards to their cytolytic and non-cytolytic activities to inhibit pathogen replication and invasion. We also discuss how reactive oxygen species contribute to the various antimicrobial mechanisms exerted by the granzymes.
Collapse
Affiliation(s)
- Marilyne Lavergne
- Department of Oncology, Microbiology and Immunology, Anatomy Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Maria Andrea Hernández-Castañeda
- Division Infectious Disease and International Medicine, Department of Medicine, Center for Immunology, Minneapolis, MN, United States
| | - Pierre-Yves Mantel
- Department of Oncology, Microbiology and Immunology, Anatomy Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Denis Martinvalet
- Department of Biomedical Sciences, Venetian Institute of Molecular Medicine, Padova, Italy.,Department of Biomedical Sciences, University of Padua, Padova, Italy
| | - Michael Walch
- Department of Oncology, Microbiology and Immunology, Anatomy Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
14
|
Cazzetta V, Bruni E, Terzoli S, Carenza C, Franzese S, Piazza R, Marzano P, Donadon M, Torzilli G, Cimino M, Simonelli M, Bello L, Villa A, Tan L, Ravens S, Prinz I, Supino D, Colombo FS, Lugli E, Marcenaro E, Vivier E, Della Bella S, Mikulak J, Mavilio D. NKG2A expression identifies a subset of human Vδ2 T cells exerting the highest antitumor effector functions. Cell Rep 2021; 37:109871. [PMID: 34686325 DOI: 10.1016/j.celrep.2021.109871] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/13/2021] [Accepted: 09/29/2021] [Indexed: 01/13/2023] Open
Abstract
Human Vδ2 cells are innate-like γδ T effectors performing potent immune surveillance against tumors. The constitutive expression of NKG2A identifies a subset of Vδ2 T cells licensed with an intrinsic hyper-responsiveness against cancer. Indeed, the transcriptomic profiles of NKG2A+ and NKG2A- cells characterize two distinct "intralineages" of Vδ2 T lymphocytes that appear early during development, keep their phenotypes, and show self-renewal capabilities in adult life. The hyper-responsiveness of NKG2A+ Vδ2 T cells is counterbalanced by the inhibitory signaling delivered by human leukocyte antigen E (HLA-E) expressed on malignant cells as a tumor-escape mechanism. However, either masking or knocking out NKG2A restores the capacity of Vδ2 T cells to exert the highest effector functions even against HLA-E+ tumors. This is highly relevant in the clinic, as the different degrees of engagement of the NKG2A-HLA-E checkpoint in hepatocellular carcinoma, glioblastoma, and non-small cell lung cancer directly impact patients' overall survival. These findings open avenues for developing combined cellular and immunologic anticancer therapies.
Collapse
Affiliation(s)
- Valentina Cazzetta
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Elena Bruni
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Sara Terzoli
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Claudia Carenza
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Sara Franzese
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Paolo Marzano
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Matteo Donadon
- Department of Biomedical Science, Humanitas University, 20090 Pieve Emanuele, Milan, Italy; Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Guido Torzilli
- Department of Biomedical Science, Humanitas University, 20090 Pieve Emanuele, Milan, Italy; Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Matteo Cimino
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Matteo Simonelli
- Department of Biomedical Science, Humanitas University, 20090 Pieve Emanuele, Milan, Italy; Department of Medical Oncology and Hematology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Lorenzo Bello
- U.O. Neurochirurgia Oncologica, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Anna Villa
- Division of Regenerative, Medicine, Stem Cells and Gene Therapy, San Raffaele Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Milan, Italy; Institute of Genetic and Biomedical Research, UOS Milan, National Research Council, Rozzano, Milan, Italy
| | - Likai Tan
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Sarina Ravens
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Domenico Supino
- Department of Biomedical Science of Clinical and Experimental Immunology, Humanitas University, 20090 Pieve Emanuele, Milan, Italy
| | - Federico S Colombo
- Humanitas Flow Cytometry Core, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Enrico Lugli
- Humanitas Flow Cytometry Core, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy; Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy
| | - Emanuela Marcenaro
- Department of Experimental Medicine, Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Eric Vivier
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France; Research Laboratories, Innate Pharma, Marseille, France; Service d'Immunologie, Hôpital de la Timone, APHM, Marseille-Immunopôle, Marseille, France
| | - Silvia Della Bella
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Joanna Mikulak
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Domenico Mavilio
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy.
| |
Collapse
|
15
|
de Jong LC, Crnko S, ten Broeke T, Bovenschen N. Noncytotoxic functions of killer cell granzymes in viral infections. PLoS Pathog 2021; 17:e1009818. [PMID: 34529743 PMCID: PMC8445437 DOI: 10.1371/journal.ppat.1009818] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cytotoxic lymphocytes produce granules armed with a set of 5 serine proteases (granzymes (Gzms)), which, together with the pore-forming protein (perforin), serve as a major defense against viral infections in humans. This granule-exocytosis pathway subsumes a well-established mechanism in which target cell death is induced upon perforin-mediated entry of Gzms and subsequent activation of various (apoptosis) pathways. In the past decade, however, a growing body of evidence demonstrated that Gzms also inhibit viral replication and potential reactivation in cell death–independent manners. For example, Gzms can induce proteolysis of viral or host cell proteins necessary for the viral entry, release, or intracellular trafficking, as well as augment pro-inflammatory antiviral cytokine response. In this review, we summarize current evidence for the noncytotoxic mechanisms and roles by which killer cells can use Gzms to combat viral infections, and we discuss the potential thereof for the development of novel therapies.
Collapse
Affiliation(s)
- Lisanne C. de Jong
- Radboud University, Nijmegen, the Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sandra Crnko
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Toine ten Broeke
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
- * E-mail:
| |
Collapse
|
16
|
The role of CD8 + Granzyme B + T cells in the pathogenesis of Takayasu's arteritis. Clin Rheumatol 2021; 41:167-176. [PMID: 34494213 DOI: 10.1007/s10067-021-05903-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/11/2021] [Accepted: 09/01/2021] [Indexed: 01/26/2023]
Abstract
OBJECTIVE T cell-mediated immune response plays a key role in Takayasu arteritis (TAK). Although previous studies have showed the roles of CD4+T cell and its subsets in TAK, the change of CD8+ T cell subsets remains unclear. This study investigated the role of CD8+ T cell subsets in TAK. METHODS The study consisted of 56 TA patients and 51 healthy controls. The percentages of CD8+T cells, CD8+GranzymeB+ T cells, CD8+Perforin+ T cells, and CD8+IFN-γ+ T cells in blood samples were analyzed by flow cytometry. RESULTS We found that the percentages of CD8+GranzymeB+ T cells (P = 0.030), CD8+Perforin+ T cells (P = 0.000), and CD8+IFN-γ+ T cells (P = 0.002) in CD8+T cells were higher in TAK patients compared to control group. After 6 months of treatment, the proportion of CD8+T cells in lymphocytes were significantly lower in TAK patients than the baseline assessment (P = 0.033). A lower ratio of CD8+GranzymeB+ T cells/CD8+ T cells were showed in TAK patents after treatment compared with TAK patients before treatments (P = 0.011). The change of CD8+GranzymeB+ T cells/CD8+ T cells ratio was positively correlated with the change of ITAS (r = 0.721, P = 0.002) and ITAS-A (r = 0.637, P = 0.008). Finally, the immunofluorescence staining showed the infiltration of CD8+ Granzyme B + cells in the aortic tissue of TAK patients. CONCLUSION Our results disclose that the CD8+ T lymphocytes may play a role in TAK pathogenesis. Targeting CD8+GranzymeB+ T lymphocytes or Granzyme B inhibitors could be a potential therapeutic approach for the treatment of TAK. Key Points • Our study investigated role the of CD8+ T cell subsets in TAK. • We found the percentages of CD8+GranzymeB+ T cells, CD8+Perforin+ T cells, and CD8+IFN-γ+ T cells in CD3+CD8+T cells were higher in TAK patients. • The proportion of CD8+T cells in lymphocytes and the ratio of CD8+GranzymeB+ T cells/CD8+ T cells were significantly lower in TAK patients after treatment.
Collapse
|
17
|
Abstract
Inflammaging is associated with chronic diseases, but tissue-specific changes in the immune system remain unknown. In this issue of Immunity, Mogilenko et al. use single-cell RNA sequencing, flow cytometry, and mass cytometry to describe age-related differences, including the accumulation of age-associated T cells that contribute to inflammaging.
Collapse
|
18
|
Li S, van Dijk CGM, Meeldijk J, Kok HM, Blommestein I, Verbakel ALF, Kotte M, Broekhuizen R, Laclé MM, Goldschmeding R, Cheng C, Bovenschen N. Extracellular Granzyme K Modulates Angiogenesis by Regulating Soluble VEGFR1 Release From Endothelial Cells. Front Oncol 2021; 11:681967. [PMID: 34178673 PMCID: PMC8220216 DOI: 10.3389/fonc.2021.681967] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/19/2021] [Indexed: 01/02/2023] Open
Abstract
Angiogenesis is crucial for normal development and homeostasis, but also plays a role in many diseases including cardiovascular diseases, autoimmune diseases, and cancer. Granzymes are serine proteases stored in the granules of cytotoxic cells, and have predominantly been studied for their pro-apoptotic role upon delivery in target cells. A growing body of evidence is emerging that granzymes also display extracellular functions, which largely remain unknown. In the present study, we show that extracellular granzyme K (GrK) inhibits angiogenesis and triggers endothelial cells to release soluble VEGFR1 (sVEGFR1), a decoy receptor that inhibits angiogenesis by sequestering VEGF-A. GrK does not cleave off membrane-bound VEGFR1 from the cell surface, does not release potential sVEGFR1 storage pools from endothelial cells, and does not trigger sVEGFR1 release via protease activating receptor-1 (PAR-1) activation. GrK induces de novo sVEGFR1 mRNA and protein expression and subsequent release of sVEGFR1 from endothelial cells. GrK protein is detectable in human colorectal tumor tissue and its levels positively correlate with sVEGFR1 protein levels and negatively correlate with T4 intratumoral angiogenesis and tumor size. In conclusion, extracellular GrK can inhibit angiogenesis via secretion of sVEGFR1 from endothelial cells, thereby sequestering VEGF-A and impairing VEGFR signaling. Our observation that GrK positively correlates with sVEGFR1 and negatively correlates with angiogenesis in colorectal cancer, suggest that the GrK-sVEGFR1-angiogenesis axis may be a valid target for development of novel anti-angiogenic therapies in cancer.
Collapse
Affiliation(s)
- Shuang Li
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Christian G M van Dijk
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jan Meeldijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Helena M Kok
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Isabelle Blommestein
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Annick L F Verbakel
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marit Kotte
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Roel Broekhuizen
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Miangela M Laclé
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Caroline Cheng
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
19
|
García-Laorden MI, Hoogendijk AJ, Wiewel MA, van Vught LA, Schultz MJ, Bovenschen N, de Vos AF, van der Poll T. Intracellular expression of granzymes A, B, K and M in blood lymphocyte subsets of critically ill patients with or without sepsis. Clin Exp Immunol 2021; 205:222-231. [PMID: 33866542 PMCID: PMC8274148 DOI: 10.1111/cei.13601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 11/29/2022] Open
Abstract
Sepsis is a complex syndrome related to an infection-induced exaggerated inflammatory response, which is associated with a high mortality. Granzymes (Gzm) are proteases mainly found in cytotoxic lymphocytes that not only have a role in target cell death, but also as mediators of infection and inflammation. In this study we sought to analyse the intracellular expression of GzmA, B, M and K by flow cytometry in diverse blood lymphocyte populations from 22 sepsis patients, 12 non-infected intensive care unit (ICU) patients and 32 healthy controls. Additionally, we measured GzmA and B plasma levels. Both groups of patients presented decreased percentage of natural killer (NK) cells expressing GzmA, B and M relative to healthy controls, while sepsis patients showed an increased proportion of CD8+ T cells expressing GzmB compared to controls. Expression of GzmK remained relatively unaltered between groups. Extracellular levels of GzmB were increased in non-infected ICU patients relative to sepsis patients and healthy controls. Our results show differential alterations in intracellular expression of Gzm in sepsis patients and non-infected critically ill patients compared to healthy individuals depending on the lymphocyte population and on the Gzm.
Collapse
Affiliation(s)
- M Isabel García-Laorden
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.,CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas de Gran Canaria, Spain
| | - Arie J Hoogendijk
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Maryse A Wiewel
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Lonneke A van Vught
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Marcus J Schultz
- Department of Intensive Care Medicine, and Laboratory of Experimental Intensive Care and Anesthesiology (LEICA), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.,Mahidol-Oxford Tropical Medicine Research Unit (MORU), Mahidol University, Bangkok, Thailand.,Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Niels Bovenschen
- Department of Pathology and Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Alex F de Vos
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.,Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
20
|
Bouwman AC, van Daalen KR, Crnko S, Ten Broeke T, Bovenschen N. Intracellular and Extracellular Roles of Granzyme K. Front Immunol 2021; 12:677707. [PMID: 34017346 PMCID: PMC8129556 DOI: 10.3389/fimmu.2021.677707] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/21/2021] [Indexed: 12/30/2022] Open
Abstract
Granzymes are a family of serine proteases stored in granules inside cytotoxic cells of the immune system. Granzyme K (GrK) has been only limitedly characterized and knowledge on its molecular functions is emerging. Traditionally GrK is described as a granule-secreted, pro-apoptotic serine protease. However, accumulating evidence is redefining the functions of GrK by the discovery of novel intracellular (e.g. cytotoxicity, inhibition of viral replication) and extracellular roles (e.g. endothelial activation and modulation of a pro-inflammatory immune cytokine response). Moreover, elevated GrK levels are associated with disease, including viral and bacterial infections, airway inflammation and thermal injury. This review aims to summarize and discuss the current knowledge of i) intracellular and extracellular GrK activity, ii) cytotoxic and non-cytotoxic GrK functioning, iii) the role of GrK in disease, and iv) GrK as a potential therapeutic target.
Collapse
Affiliation(s)
- Annemieke C Bouwman
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Kim R van Daalen
- Cardiovascular Epidemiology Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Sandra Crnko
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Toine Ten Broeke
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands.,Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
21
|
Santiago L, Castro M, Sanz-Pamplona R, Garzón M, Ramirez-Labrada A, Tapia E, Moreno V, Layunta E, Gil-Gómez G, Garrido M, Peña R, Lanuza PM, Comas L, Jaime-Sanchez P, Uranga-Murillo I, Del Campo R, Pelegrín P, Camerer E, Martínez-Lostao L, Muñoz G, Uranga JA, Alcalde A, Galvez EM, Ferrandez A, Bird PI, Metkar S, Arias MA, Pardo J. Extracellular Granzyme A Promotes Colorectal Cancer Development by Enhancing Gut Inflammation. Cell Rep 2021; 32:107847. [PMID: 32640217 DOI: 10.1016/j.celrep.2020.107847] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/11/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023] Open
Abstract
If not properly regulated, the inflammatory immune response can promote carcinogenesis, as evident in colorectal cancer (CRC). Aiming to gain mechanistic insight into the link between inflammation and CRC, we perform transcriptomics analysis of human CRC, identifying a strong correlation between expression of the serine protease granzyme A (GzmA) and inflammation. In a dextran sodium sulfate and azoxymethane (DSS/AOM) mouse model, deficiency and pharmacological inhibition of extracellular GzmA both attenuate gut inflammation and prevent CRC development, including the initial steps of cell transformation and epithelial-to-mesenchymal transition. Mechanistically, extracellular GzmA induces NF-κB-dependent IL-6 production in macrophages, which in turn promotes STAT3 activation in cultured CRC cells. Accordingly, colon tissues from DSS/AOM-treated, GzmA-deficient animals present reduced levels of pSTAT3. By identifying GzmA as a proinflammatory protease that promotes CRC development, these findings provide information on mechanisms that link immune cell infiltration to cancer progression and present GzmA as a therapeutic target for CRC.
Collapse
Affiliation(s)
- Llipsy Santiago
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Marta Castro
- Department of Pharmacology and Physiology, Faculty of Health and Sports Sciences, University of Zaragoza, 22002 Huesca, Spain
| | - Rebeca Sanz-Pamplona
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERESP, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Marcela Garzón
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Ariel Ramirez-Labrada
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Elena Tapia
- Animal Unit, University of Zaragoza, 50009 Zaragoza, Spain
| | - Víctor Moreno
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERESP, L'Hospitalet de Llobregat, Barcelona, Spain; Department of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Elena Layunta
- Department of Pharmacology and Physiology, Faculty of Veterinary, University of Zaragoza, 50013 Zaragoza, Spain
| | - Gabriel Gil-Gómez
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), 08003 Barcelona
| | - Marta Garrido
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), 08003 Barcelona
| | - Raúl Peña
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), 08003 Barcelona
| | - Pilar M Lanuza
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Laura Comas
- Instituto de Carboquímica ICB-CSIC, 50018 Zaragoza, Spain
| | - Paula Jaime-Sanchez
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Iratxe Uranga-Murillo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Rosa Del Campo
- Department of Microbiology, University Hospital Ramón y Cajal & Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Pablo Pelegrín
- Unidad de Inflamación Molecular y Cirugía Experimental, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Eric Camerer
- Université de Paris, Paris Cardiovascular Research Center, INSERM U970, 75015 Paris, France
| | - Luis Martínez-Lostao
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain; Department of Immunology, University Clinic Hospital Lozano Blesa, 50009, Zaragoza, Spain and Department of Pathology, University Clinic Hospital Lozano Blesa, University of Zaragoza, IIS Aragón, CIBEREHD, 50009 Zaragoza, Spain; Nanoscience Institute of Aragon (INA), University of Zaragoza, 50018 Zaragoza, Spain; Department Biochemistry and Molecular and Cell Biology and Department Microbiology, Preventive Medicine and Public Health, University of Zaragoza, 50009 Zaragoza, Spain
| | - Guillermo Muñoz
- Department of Immunology, University Clinic Hospital Lozano Blesa, 50009, Zaragoza, Spain and Department of Pathology, University Clinic Hospital Lozano Blesa, University of Zaragoza, IIS Aragón, CIBEREHD, 50009 Zaragoza, Spain
| | - José A Uranga
- Department of Basis Health Sciences, Faculty of Health Sciences, Rey Juan Carlos University, 28922 Madrid, Spain
| | - Anabel Alcalde
- Department of Pharmacology and Physiology, Faculty of Veterinary, University of Zaragoza, 50013 Zaragoza, Spain
| | - Eva M Galvez
- Instituto de Carboquímica ICB-CSIC, 50018 Zaragoza, Spain
| | - Angel Ferrandez
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, University of Zaragoza, IIS Aragón, CIBEREHD, Zaragoza, Spain
| | - Phillip I Bird
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University 3800 Melbourne, Australia
| | | | - Maykel A Arias
- Instituto de Carboquímica ICB-CSIC, 50018 Zaragoza, Spain.
| | - Julian Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain; Aragon I+D Foundation (ARAID), Zaragoza, Spain; Nanoscience Institute of Aragon (INA), University of Zaragoza, 50018 Zaragoza, Spain; Department Biochemistry and Molecular and Cell Biology and Department Microbiology, Preventive Medicine and Public Health, University of Zaragoza, 50009 Zaragoza, Spain; CIBER-BBN, Madrid, Spain.
| |
Collapse
|
22
|
Saccon E, Mikaeloff F, Figueras Ivern P, Végvári Á, Sönnerborg A, Neogi U, van Domselaar R. Cytotoxic Lymphocytes Target HIV-1 Gag Through Granzyme M-Mediated Cleavage. Front Immunol 2021; 12:669347. [PMID: 33953729 PMCID: PMC8089382 DOI: 10.3389/fimmu.2021.669347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/31/2021] [Indexed: 11/13/2022] Open
Abstract
Untreated HIV-1 infection leads to a slow decrease in CD4+ T cell lymphocytes over time resulting in increased susceptibility to opportunistic infections (acquired immunodeficiency syndrome, AIDS) and ultimately death of the infected individual. Initially, the host's immune response controls the infection, but cannot eliminate the HIV-1 from the host. Cytotoxic lymphocytes are the key effector cells in this response and can mediate crucial antiviral responses through the release of a set of proteases called granzymes towards HIV-1-infected cells. However, little is known about the immunological molecular mechanisms by which granzymes could control HIV-1. Since we noted that HIV-1 subtype C (HIV-1C) Gag with the tetrapeptide insertion PYKE contains a putative granzyme M (GrM) cleavage site (KEPL) that overlaps with the PYKE insertion, we analyzed the proteolytic activity of GrM towards Gag. Immunoblot analysis showed that GrM could cleave Gag proteins from HIV-1B and variants from HIV-1C of which the Gag-PYKE variant was cleaved with extremely high efficiency. The main cleavage site was directly after the insertion after leucine residue 483. GrM-mediated cleavage of Gag was also observed in co-cultures using cytotoxic lymphocytes as effector cells and this cleavage could be inhibited by a GrM inhibitor peptide. Altogether, our data indicate towards a noncytotoxic immunological mechanism by which GrM-positive cytotoxic lymphocytes target the HIV-1 Gag protein within infected cells to potentially control HIV-1 infection. This mechanism could be exploited in new therapeutic strategies to treat HIV-1-infected patients to improve immunological control of the infection.
Collapse
Affiliation(s)
- Elisa Saccon
- Division of Clinical Microbiology, ANA Futura Laboratory, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Flora Mikaeloff
- Division of Clinical Microbiology, ANA Futura Laboratory, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Pol Figueras Ivern
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Anders Sönnerborg
- Division of Clinical Microbiology, ANA Futura Laboratory, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.,Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
| | - Ujjwal Neogi
- Division of Clinical Microbiology, ANA Futura Laboratory, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
| | - Robert van Domselaar
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Martínez Cuesta L, Pérez SE. Perforin and granzymes in neurological infections: From humans to cattle. Comp Immunol Microbiol Infect Dis 2021; 75:101610. [PMID: 33453589 DOI: 10.1016/j.cimid.2021.101610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 01/14/2023]
Abstract
Perforin and granzymes are essential components of the cytotoxic granules present in cytotoxic T lymphocytes and natural killer cells. These proteins play a crucial role in a variety of conditions, including viral infections, tumor immune surveillance, and tissue rejection. Besides their beneficial effect in most of these situations, perforin and granzymes have also been associated with tissue damage and immune diseases. Moreover, it has been reported that perforin and granzymes released during viral infections could contribute to the pathogenesis of diseases. In this review, we summarize the information available on human perforin and granzymes and their relationship with neurological infections and immune disorders. Furthermore, we compare this information with that available for bovine and present data on perforin and granzymes expression in cattle infected with bovine alphaherpesvirus types1 and -5. To our knowledge, this is the first review analyzing the impact of perforin and granzymes on neurological infections caused by bovine herpesviruses.
Collapse
Affiliation(s)
- Lucía Martínez Cuesta
- Virology, SAMP Department, Centro de Investigación Veterinaria de Tandil (CIVETAN), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Pinto 399, Tandil, PC7000, Buenos Aires, Argentina
| | - Sandra Elizabeth Pérez
- Virology, SAMP Department, Centro de Investigación Veterinaria de Tandil (CIVETAN), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Pinto 399, Tandil, PC7000, Buenos Aires, Argentina.
| |
Collapse
|
24
|
Ahmad J, Ikram S, Hafeez AB, Durdagi S. Physics-driven identification of clinically approved and investigation drugs against human neutrophil serine protease 4 (NSP4): A virtual drug repurposing study. J Mol Graph Model 2020; 101:107744. [PMID: 33032202 DOI: 10.1016/j.jmgm.2020.107744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 01/19/2023]
Abstract
Neutrophils synthesize four immune associated serine proteases: Cathepsin G (CTSG), Elastase (ELANE), Proteinase 3 (PRTN3) and Neutrophil Serine Protease 4 (NSP4). While previously considered to be immune modulators, overexpression of neutrophil serine proteases correlates with various disease conditions. Therefore, identifying novel small molecules that can potentially control or inhibit the proteolytic activity of these proteases is crucial to revert or temper the aggravated disease phenotype. To the best of our knowledge, although there is limited data for inhibitors of other neutrophil protease members, there is no previous clinical study of a synthetic small molecule inhibitor targeting NSP4. In this study, an integrated molecular modeling algorithm was performed within a virtual drug repurposing study to identify novel inhibitors for NSP4, using clinically approved and investigation drugs library (∼8000 compounds). Based on our rigorous filtration, we found that following molecules Becatecarin, Iogulamide, Delprostenate and Iralukast are predicted to block the activity of NSP4 by interacting with core catalytic residues. The selected ligands were energetically more favorable compared to the reference molecule. The result of this study identifies promising molecules as potential lead candidates.
Collapse
Affiliation(s)
- Jamshaid Ahmad
- Center of Biotechnology & Microbiology, University of Peshawar, Pakistan.
| | - Saima Ikram
- Center of Biotechnology & Microbiology, University of Peshawar, Pakistan; Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Ahmer Bin Hafeez
- Center of Biotechnology & Microbiology, University of Peshawar, Pakistan
| | - Serdar Durdagi
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey.
| |
Collapse
|
25
|
Sordo-Bahamonde C, Lorenzo-Herrero S, Payer ÁR, Gonzalez S, López-Soto A. Mechanisms of Apoptosis Resistance to NK Cell-Mediated Cytotoxicity in Cancer. Int J Mol Sci 2020; 21:ijms21103726. [PMID: 32466293 PMCID: PMC7279491 DOI: 10.3390/ijms21103726] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cells are major contributors to immunosurveillance and control of tumor development by inducing apoptosis of malignant cells. Among the main mechanisms involved in NK cell-mediated cytotoxicity, the death receptor pathway and the release of granules containing perforin/granzymes stand out due to their efficacy in eliminating tumor cells. However, accumulated evidence suggest a profound immune suppression in the context of tumor progression affecting effector cells, such as NK cells, leading to decreased cytotoxicity. This diminished capability, together with the development of resistance to apoptosis by cancer cells, favor the loss of immunogenicity and promote immunosuppression, thus partially inducing NK cell-mediated killing resistance. Altered expression patterns of pro- and anti-apoptotic proteins along with genetic background comprise the main mechanisms of resistance to NK cell-related apoptosis. Herein, we summarize the main effector cytotoxic mechanisms against tumor cells, as well as the major resistance strategies acquired by tumor cells that hamper the extrinsic and intrinsic apoptotic pathways related to NK cell-mediated killing.
Collapse
Affiliation(s)
- Christian Sordo-Bahamonde
- Department of Functional Biology, Immunology, University of Oviedo, 33006 Oviedo, Spain; (S.L.-H.); (S.G.)
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Correspondence: (C.S.-B.); (A.L.-S.)
| | - Seila Lorenzo-Herrero
- Department of Functional Biology, Immunology, University of Oviedo, 33006 Oviedo, Spain; (S.L.-H.); (S.G.)
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Ángel R. Payer
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Hematology, Hospital Universitario Central de Asturias (HUCA), 33011 Oviedo, Spain
| | - Segundo Gonzalez
- Department of Functional Biology, Immunology, University of Oviedo, 33006 Oviedo, Spain; (S.L.-H.); (S.G.)
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Alejandro López-Soto
- Instituto Universitario de Oncología del Principado de Asturias, IUOPA, 33006 Oviedo, Spain;
- Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Department of Biochemistry and Molecular Biology, University of Oviedo, 33006 Oviedo, Spain
- Correspondence: (C.S.-B.); (A.L.-S.)
| |
Collapse
|
26
|
Shan L, Li S, Meeldijk J, Blijenberg B, Hendriks A, van Boxtel KJWM, van den Berg SPH, Groves IJ, Potts M, Svrlanska A, Stamminger T, Wills MR, Bovenschen N. Killer cell proteases can target viral immediate-early proteins to control human cytomegalovirus infection in a noncytotoxic manner. PLoS Pathog 2020; 16:e1008426. [PMID: 32282833 PMCID: PMC7179929 DOI: 10.1371/journal.ppat.1008426] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 04/23/2020] [Accepted: 02/21/2020] [Indexed: 12/17/2022] Open
Abstract
Human cytomegalovirus (HCMV) is the most frequent viral cause of congenital defects and can trigger devastating disease in immune-suppressed patients. Cytotoxic lymphocytes (CD8+ T cells and NK cells) control HCMV infection by releasing interferon-γ and five granzymes (GrA, GrB, GrH, GrK, GrM), which are believed to kill infected host cells through cleavage of intracellular death substrates. However, it has recently been demonstrated that the in vivo killing capacity of cytotoxic T cells is limited and multiple T cell hits are required to kill a single virus-infected cell. This raises the question whether cytotoxic lymphocytes can use granzymes to control HCMV infection in a noncytotoxic manner. Here, we demonstrate that (primary) cytotoxic lymphocytes can block HCMV dissemination independent of host cell death, and interferon-α/β/γ. Prior to killing, cytotoxic lymphocytes induce the degradation of viral immediate-early (IE) proteins IE1 and IE2 in HCMV-infected cells. Intriguingly, both IE1 and/or IE2 are directly proteolyzed by all human granzymes, with GrB and GrM being most efficient. GrB and GrM cleave IE1 after Asp398 and Leu414, respectively, likely resulting in IE1 aberrant cellular localization, IE1 instability, and functional impairment of IE1 to interfere with the JAK-STAT signaling pathway. Furthermore, GrB and GrM cleave IE2 after Asp184 and Leu173, respectively, resulting in IE2 aberrant cellular localization and functional abolishment of IE2 to transactivate the HCMV UL112 early promoter. Taken together, our data indicate that cytotoxic lymphocytes can also employ noncytotoxic ways to control HCMV infection, which may be explained by granzyme-mediated targeting of indispensable viral proteins during lytic infection. Human cytomegalovirus (HCMV) is the leading viral cause of congenital defects, can trigger disease in immune-compromised patients, and plays roles in cancer development. Cytotoxic lymphocytes kill HCMV-infected cells via releasing a set of five cytotoxic serine proteases called granzymes. However, the killing capacity of cytotoxic cells is limited and multiple T cell hits are required to kill a single virus-infected cell. This raises the question whether cytotoxic lymphocytes can use granzymes to control HCMV infection in a noncytotoxic manner. Here, we show that cytotoxic lymphocytes can also use granzymes to inhibit HCMV replication in absence of cell death. All five granzymes cleave and inactivate both viral immediate-early (IE1/2) proteins, which are essential players for initiating HCMV infection. Our data support the model that cytotoxic cells employ granzymes to dampen HCMV replication prior to accumulation of sufficient hits to kill the infected cell.
Collapse
Affiliation(s)
- Liling Shan
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Shuang Li
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan Meeldijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bernadet Blijenberg
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Astrid Hendriks
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - Ian J. Groves
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Martin Potts
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Adriana Svrlanska
- Institute of Clinical and Molecular Virology, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | - Mark R. Wills
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
27
|
Figueiredo CR, Kalirai H, Sacco JJ, Azevedo RA, Duckworth A, Slupsky JR, Coulson JM, Coupland SE. Loss of BAP1 expression is associated with an immunosuppressive microenvironment in uveal melanoma, with implications for immunotherapy development. J Pathol 2020; 250:420-439. [PMID: 31960425 PMCID: PMC7216965 DOI: 10.1002/path.5384] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/28/2019] [Accepted: 01/14/2020] [Indexed: 12/22/2022]
Abstract
Immunotherapy using immune checkpoint inhibitors (ICIs) induces durable responses in many metastatic cancers. Metastatic uveal melanoma (mUM), typically occurring in the liver, is one of the most refractory tumours to ICIs and has dismal outcomes. Monosomy 3 (M3), polysomy 8q, and BAP1 loss in primary uveal melanoma (pUM) are associated with poor prognoses. The presence of tumour-infiltrating lymphocytes (TILs) within pUM and surrounding mUM - and some evidence of clinical responses to adoptive TIL transfer - strongly suggests that UMs are indeed immunogenic despite their low mutational burden. The mechanisms that suppress TILs in pUM and mUM are unknown. We show that BAP1 loss is correlated with upregulation of several genes associated with suppressive immune responses, some of which build an immune suppressive axis, including HLA-DR, CD38, and CD74. Further, single-cell analysis of pUM by mass cytometry confirmed the expression of these and other markers revealing important functions of infiltrating immune cells in UM, most being regulatory CD8+ T lymphocytes and tumour-associated macrophages (TAMs). Transcriptomic analysis of hepatic mUM revealed similar immune profiles to pUM with BAP1 loss, including the expression of IDO1. At the protein level, we observed TAMs and TILs entrapped within peritumoural fibrotic areas surrounding mUM, with increased expression of IDO1, PD-L1, and β-catenin (CTNNB1), suggesting tumour-driven immune exclusion and hence the immunotherapy resistance. These findings aid the understanding of how the immune response is organised in BAP1 - mUM, which will further enable functional validation of detected biomarkers and the development of focused immunotherapeutic approaches. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Carlos R Figueiredo
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
- Department of the Faculty of Medicine, MediCity Research Laboratory and Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Helen Kalirai
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
| | - Joseph J Sacco
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
- Department of Medical OncologyThe Clatterbridge Cancer CentreWirralUK
| | - Ricardo A Azevedo
- Department of Cancer BiologyThe University of Texas–MD Anderson Cancer CenterHoustonTXUSA
| | - Andrew Duckworth
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
| | - Joseph R Slupsky
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
| | - Judy M Coulson
- Department of Cellular and Molecular PhysiologyUniversity of LiverpoolLiverpoolUK
| | - Sarah E Coupland
- Department of Molecular and Clinical Cancer Medicine, ITMUniversity of LiverpoolLiverpoolUK
- Liverpool Clinical LaboratoriesRoyal Liverpool University HospitalLiverpoolUK
| |
Collapse
|
28
|
Elhelbawy NG, Nassar AAH, Eltorgoman AEA, Saber SM, Badr EA. Immunological microenvironment gene expression in patients with diffuse large B cell non Hodgkin lymphoma. Biochem Biophys Rep 2020; 21:100731. [PMID: 32025577 PMCID: PMC6997500 DOI: 10.1016/j.bbrep.2020.100731] [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: 12/19/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 12/17/2022] Open
Abstract
Background Non Hodgkin lymphoma (NHL) is one of the immune system cancers. The occurrence and progression of malignant lymphomas depends on cellular pathways deregulation. Understanding the relationship between the immune system at the genetic level and malignant transformation is critical to reach its etiology. Objective The aim of this work is to evaluate the expression of five immune related genes (PD-1, FOXP3, GrA, GrB and CD11c) in patients with diffuse large B cell non Hodgkin lymphoma (DLBCL). Materials and methods This study was conducted on fifty patients with DLBCL and fifty sex and age matched apparently healthy subjects. The participants were subjected to these laboratory investigations: complete blood count, serum lactate dehydrogenase and β2microglobulin (β2M) levels and determination of PD-1, FOXP3, GrA, GrB and CD11c gene expressions. Results The results of this study revealed that PD-1, FOXP3, GrA, GrB and CD11c gene expressions were significantly increased in DLBCL patients. Conclusion Patients with DLBCL have variablePD-1, FOXP3,GrA, GrB and CD11cgene expressions levels, which are correlated with the overall survival (OS) indicating that they can be good predictors of outcome in these patients.
Collapse
Affiliation(s)
- Nesreen G Elhelbawy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Menoufia University, Egypt
| | - Adel A H Nassar
- Department of Organic Chemistry, Faculty of Science, Menoufia University, Egypt
| | | | - Safa M Saber
- Chemist at Central Laboratory, Faculty of Medicine, Menoufia University, Egypt
| | - Eman Ae Badr
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Menoufia University, Egypt
| |
Collapse
|
29
|
Fernandes C, Suares D, Yergeri MC. Tumor Microenvironment Targeted Nanotherapy. Front Pharmacol 2018; 9:1230. [PMID: 30429787 PMCID: PMC6220447 DOI: 10.3389/fphar.2018.01230] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Recent developments in nanotechnology have brought new approaches to cancer diagnosis and therapy. While enhanced permeability and retention effect promotes nano-chemotherapeutics extravasation, the abnormal tumor vasculature, high interstitial pressure and dense stroma structure limit homogeneous intratumoral distribution of nano-chemotherapeutics and compromise their imaging and therapeutic effect. Moreover, heterogeneous distribution of nano-chemotherapeutics in non-tumor-stroma cells damages the non-tumor cells, and interferes with tumor-stroma crosstalk. This can lead not only to inhibition of tumor progression, but can also paradoxically induce acquired resistance and facilitate tumor cell proliferation and metastasis. Overall, the tumor microenvironment plays a vital role in regulating nano-chemotherapeutics distribution and their biological effects. In this review, the barriers in tumor microenvironment, its consequential effects on nano-chemotherapeutics, considerations to improve nano-chemotherapeutics delivery and combinatory strategies to overcome acquired resistance induced by tumor microenvironment have been summarized. The various strategies viz., nanotechnology based approach as well as ligand-mediated, redox-responsive, and enzyme-mediated based combinatorial nanoapproaches have been discussed in this review.
Collapse
Affiliation(s)
| | | | - Mayur C Yergeri
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's Narsee Monjee Institute of Management Studies - NMIMS, Mumbai, India
| |
Collapse
|
30
|
Spetz J, Presser AG, Sarosiek KA. T Cells and Regulated Cell Death: Kill or Be Killed. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 342:27-71. [PMID: 30635093 DOI: 10.1016/bs.ircmb.2018.07.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell death plays two major complementary roles in T cell biology: mediating the removal of cells that are targeted by T cells and the removal of T cells themselves. T cells serve as major actors in the adaptive immune response and function by selectively killing cells which are infected or dysfunctional. This feature is highly involved during homeostatic maintenance, and is relied upon and modulated in the context of cancer immunotherapy. The vital recognition and elimination of both autoreactive T cells and cells which are unable to recognize threats is a highly selective and regulated process. Moreover, detection of potential threats will result in the activation and expansion of T cells, which on resolution of the immune response will need to be eliminated. The culling of these T cells can be executed via a multitude of cell death pathways which are used in context-specific manners. Failure of these processes may result in an accumulation of misdirected or dysfunctional T cells, leading to complications such as autoimmunity or cancer. This review will focus on the role of cell death regulation in the maintenance of T cell homeostasis, as well as T cell-mediated elimination of infected or dysfunctional cells, and will summarize and discuss the current knowledge of the cellular mechanisms which are implicated in these processes.
Collapse
Affiliation(s)
- Johan Spetz
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| | - Adam G Presser
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| | - Kristopher A Sarosiek
- John B. Little Center for Radiation Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Science, Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
31
|
Mao FY, Zhao YL, Lv YP, Teng YS, Kong H, Liu YG, Wu XL, Hao CJ, Chen W, Duan MB, Han B, Ma Q, Wang TT, Peng LS, Zhang JY, Cheng P, Su CY, Fu XL, Zou QM, Guo G, Guo XL, Zhuang Y. CD45 +CD33 lowCD11b dim myeloid-derived suppressor cells suppress CD8 + T cell activity via the IL-6/IL-8-arginase I axis in human gastric cancer. Cell Death Dis 2018; 9:763. [PMID: 29988030 PMCID: PMC6037756 DOI: 10.1038/s41419-018-0803-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/14/2018] [Accepted: 05/31/2018] [Indexed: 12/26/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a prominent component of the pro-tumoral response. The phenotype of and mechanisms used by MDSCs is heterogeneous and requires more precise characterization in gastric cancer (GC) patients. Here, we have identified a novel subset of CD45+CD33lowCD11bdim MDSCs in the peripheral blood of GC patients compared to healthy individuals. CD45+CD33lowCD11bdim MDSCs morphologically resembled neutrophils and expressed high levels of the neutrophil marker CD66b. Circulating CD45+CD33lowCD11bdim MDSCs effectively suppressed CD8+ T cells activity through the inhibition of CD8+ T cell proliferation and interferon-γ (IFN-γ) and granzyme B (GrB) production. The proportion of CD45+CD33lowCD11bdim MDSCs also negatively correlated with the proportion of IFN-γ+CD8+ T cell in the peripheral blood of GC patients. GC patient serum-derived IL-6 and IL-8 activated and induced CD45+CD33lowCD11bdim MDSCs to express arginase I via the PI3K-AKT signaling pathway. This pathway contributed to CD8+ T cell suppression as it was partially rescued by the blockade of the IL-6/IL-8-arginase I axis. Peripheral blood CD45+CD33lowCD11bdim MDSCs, as well as IL-6, IL-8, and arginase I serum levels, positively correlated with GC progression and negatively correlated with overall patient survival. Altogether, our results highlight that a subset of neutrophilic CD45+CD33lowCD11bdim MDSCs is functionally immunosuppressive and activated via the IL-6/IL-8-arginase I axis in GC patients.
Collapse
Affiliation(s)
- Fang-Yuan Mao
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yong-Liang Zhao
- Department of General Surgery and Centre of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yi-Pin Lv
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yong-Sheng Teng
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hui Kong
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yu-Gang Liu
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiao-Long Wu
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Chuan-Jie Hao
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Weisan Chen
- La Trobe Institute of Molecular Science, School of Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Mu-Bing Duan
- La Trobe Institute of Molecular Science, School of Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Bin Han
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Qiang Ma
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Ting-Ting Wang
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Liu-Sheng Peng
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jin-Yu Zhang
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ping Cheng
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Chong-Yu Su
- Department of General Surgery and Centre of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xiao-Long Fu
- Department of General Surgery and Centre of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Quan-Ming Zou
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Gang Guo
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiao-Lan Guo
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China.
| | - Yuan Zhuang
- National Engineering Research Centre of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China.
| |
Collapse
|
32
|
Yang J, Vrettou C, Connelley T, Morrison WI. Identification and annotation of bovine granzyme genes reveals a novel granzyme encoded within the trypsin-like locus. Immunogenetics 2018; 70:585-597. [PMID: 29947943 PMCID: PMC6096847 DOI: 10.1007/s00251-018-1062-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/09/2018] [Indexed: 11/26/2022]
Abstract
Granzymes are a family of serine proteases found in the lytic granules of cytotoxic T lymphocytes and natural killer (NK) cells, which are involved in killing of susceptible target cells. Most information on granzymes and their enzymatic specificities derive from studies in humans and mice. Although granzymes shared by both species show a high level of conservation, the complement of granzyme genes differs between the species. The aim of this study was to identify granzyme genes expressed in cattle, determine their genomic locations and analyse their sequences to predict likely functional specificities. Orthologues of the five granzyme genes found in humans (A, B, H, K and M) were identified, as well a novel gene designated granzyme O, most closely related to granzyme A. An orthologue of granzyme O was found in pigs and a non-function version was detected in the human genome. Use of specific PCRs demonstrated that all of these genes, including granzyme O, are expressed in activated subsets of bovine lymphocytes, with particularly high levels in CD8 T cells. Consistent with findings in humans and mice, the granzyme-encoding genes were located on three distinct genomic loci, which correspond to different proteolytic enzymatic activities, namely trypsin-like, chymotrypsin-like and metase-like. Analysis of amino acid sequences indicated that the granzyme proteins have broadly similar enzymatic specificities to their human and murine counterparts but indicated that granzyme B has a different secondary specificity. These findings provide the basis for further work to examine their role in the cytotoxic activity of bovine CD8 T cells.
Collapse
Affiliation(s)
- Jie Yang
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH8 9YL, UK.,Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, NW3 2QG, UK
| | - Christina Vrettou
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH8 9YL, UK
| | - Tim Connelley
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH8 9YL, UK
| | - W Ivan Morrison
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH8 9YL, UK.
| |
Collapse
|
33
|
Hoves S, Ooi CH, Wolter C, Sade H, Bissinger S, Schmittnaegel M, Ast O, Giusti AM, Wartha K, Runza V, Xu W, Kienast Y, Cannarile MA, Levitsky H, Romagnoli S, De Palma M, Rüttinger D, Ries CH. Rapid activation of tumor-associated macrophages boosts preexisting tumor immunity. J Exp Med 2018; 215:859-876. [PMID: 29436396 PMCID: PMC5839760 DOI: 10.1084/jem.20171440] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/20/2017] [Accepted: 12/21/2017] [Indexed: 12/14/2022] Open
Abstract
Combined CSF-1R+CD40 antibody therapy induces profound and rapid TAM reprogramming before TAMs are eliminated. This combination of cancer immunotherapies tailored to activate the innate immune system creates an inflamed tumor microenvironment ultimately leading to tumor eradication by the adaptive immunity. Depletion of immunosuppressive tumor-associated macrophages (TAMs) or reprogramming toward a proinflammatory activation state represent different strategies to therapeutically target this abundant myeloid population. In this study, we report that inhibition of colony-stimulating factor-1 receptor (CSF-1R) signaling sensitizes TAMs to profound and rapid reprogramming in the presence of a CD40 agonist before their depletion. Despite the short-lived nature of macrophage hyperactivation, combined CSF-1R+CD40 stimulation of macrophages is sufficient to create a proinflammatory tumor milieu that reinvigorates an effective T cell response in transplanted tumors that are either responsive or insensitive to immune checkpoint blockade. The central role of macrophages in regulating preexisting immunity is substantiated by depletion experiments, transcriptome analysis of ex vivo sorted TAMs, and gene expression profiling of whole tumor lysates at an early treatment time point. This approach enabled the identification of specific combination-induced changes among the pleiotropic activation spectrum of the CD40 agonist. In patients, CD40 expression on human TAMs was detected in mesothelioma and colorectal adenocarcinoma.
Collapse
Affiliation(s)
- Sabine Hoves
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Chia-Huey Ooi
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany.,Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Carsten Wolter
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Hadassah Sade
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Stefan Bissinger
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Martina Schmittnaegel
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Oliver Ast
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Anna M Giusti
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Katharina Wartha
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Valeria Runza
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Wei Xu
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Yvonne Kienast
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Michael A Cannarile
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Hyam Levitsky
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Solange Romagnoli
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Dominik Rüttinger
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Carola H Ries
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
34
|
Kok HM, van den Hoogen LL, van Roon JAG, Adriaansen EJM, Fritsch-Stork RDE, Nguyen TQ, Goldschmeding R, Radstake TRDJ, Bovenschen N. Systemic and local granzyme B levels are associated with disease activity, kidney damage and interferon signature in systemic lupus erythematosus. Rheumatology (Oxford) 2017; 56:2129-2134. [DOI: 10.1093/rheumatology/kex332] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
35
|
Vermeulen JF, Van Hecke W, Adriaansen EJM, Jansen MK, Bouma RG, Villacorta Hidalgo J, Fisch P, Broekhuizen R, Spliet WGM, Kool M, Bovenschen N. Prognostic relevance of tumor-infiltrating lymphocytes and immune checkpoints in pediatric medulloblastoma. Oncoimmunology 2017; 7:e1398877. [PMID: 29399402 PMCID: PMC5790383 DOI: 10.1080/2162402x.2017.1398877] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/19/2017] [Accepted: 10/24/2017] [Indexed: 12/20/2022] Open
Abstract
Pediatric medulloblastomas are the most frequently diagnosed embryonal tumors of the central nervous system. Current therapies cause severe neurological and cognitive side effects including secondary malignancies. Cellular immunotherapy might be key to improve survival and to avoid morbidity. Efficient killing of tumor cells using immunotherapy requires to overcome cancer-associated strategies to evade cytotoxic immune responses. Here, we examined the immune response and immune evasion strategies in pediatric medulloblastomas. Cytotoxic T-cells, infiltrating medulloblastomas with variable activation status, showed no correlation with overall survival of the patients. We found limited numbers of PD1+ T-cells and complete absence of PD-L1 on medulloblastomas. Medulloblastomas downregulated immune recognition molecules MHC-I and CD1 d. Intriguingly, expression of granzyme inhibitors SERPINB1 and SERPINB4 was acquired in 23% and 50% of the tumors, respectively. Concluding, pediatric medulloblastomas exploit multiple immune evasion strategies to overcome immune surveillance. Absence of PD-L1 expression in medulloblastoma suggest limited or no added value for immunotherapy with PD1/PD-L1 blockers.
Collapse
Affiliation(s)
- Jeroen F Vermeulen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wim Van Hecke
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Mieke K Jansen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rianne G Bouma
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Paul Fisch
- Institute of Clinical Pathology, University Medical Center Freiburg, Freiburg, Germany
| | - Roel Broekhuizen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wim G M Spliet
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel Kool
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,Hopp Children's Cancer Center at NCT Heidelberg (KiTZ), Heidelberg, Germany
| | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
36
|
de Jong HK, Garcia-Laorden MI, Hoogendijk AJ, Parry CM, Maude RR, Dondorp AM, Faiz MA, van der Poll T, Wiersinga WJ. Expression of intra- and extracellular granzymes in patients with typhoid fever. PLoS Negl Trop Dis 2017; 11:e0005823. [PMID: 28749963 PMCID: PMC5549753 DOI: 10.1371/journal.pntd.0005823] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 08/08/2017] [Accepted: 07/20/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Typhoid fever, caused by the intracellular pathogen Salmonella (S.) enterica serovar Typhi, remains a major cause of morbidity and mortality worldwide. Granzymes are serine proteases promoting cytotoxic lymphocytes mediated eradication of intracellular pathogens via the induction of cell death and which can also play a role in inflammation. We aimed to characterize the expression of extracellular and intracellular granzymes in patients with typhoid fever and whether the extracellular levels of granzyme correlated with IFN-γ release. METHODS AND PRINCIPAL FINDINGS We analyzed soluble protein levels of extracellular granzyme A and B in healthy volunteers and patients with confirmed S. Typhi infection on admission and day of discharge, and investigated whether this correlated with interferon (IFN)-γ release, a cytokine significantly expressed in typhoid fever. The intracellular expression of granzyme A, B and K in subsets of lymphocytic cells was determined using flow cytometry. Patients demonstrated a marked increase of extracellular granzyme A and B in acute phase plasma and a correlation of both granzymes with IFN-γ release. In patients, lower plasma levels of granzyme B, but not granzyme A, were found at day of discharge compared to admission, indicating an association of granzyme B with stage of disease. Peripheral blood mononuclear cells of typhoid fever patients had a higher percentage of lymphocytic cells expressing intracellular granzyme A and granzyme B, but not granzyme K, compared to controls. CONCLUSION The marked increase observed in extra- and intracellular levels of granzyme expression in patients with typhoid fever, and the correlation with stage of disease, suggests a role for granzymes in the host response to this disease.
Collapse
Affiliation(s)
- Hanna K. de Jong
- Department of Internal Medicine, Division of Infectious Diseases and Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, the Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Maria Isabel Garcia-Laorden
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Arie J. Hoogendijk
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Christopher M. Parry
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Center for Tropical Medicine, Nuffield Department of Clinical Medicine, Churchill Hospital, Oxford, United Kingdom
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | - Rapeephan R. Maude
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Arjen M. Dondorp
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Center for Tropical Medicine, Nuffield Department of Clinical Medicine, Churchill Hospital, Oxford, United Kingdom
- Chittagong Medical College Hospital, Chittagong, Bangladesh
| | - Mohammed Abul Faiz
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Dev Care Foundation, Dhaka, Bangladesh
| | - Tom van der Poll
- Department of Internal Medicine, Division of Infectious Diseases and Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, the Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Willem Joost Wiersinga
- Department of Internal Medicine, Division of Infectious Diseases and Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, the Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
37
|
Arias M, Martínez-Lostao L, Santiago L, Ferrandez A, Granville DJ, Pardo J. The Untold Story of Granzymes in Oncoimmunology: Novel Opportunities with Old Acquaintances. Trends Cancer 2017; 3:407-422. [PMID: 28718416 DOI: 10.1016/j.trecan.2017.04.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 12/16/2022]
Abstract
For more than 20 years perforin and granzymes (GZMs) have been recognized as key cell death executors of cytotoxic T (Tc) and natural killer (NK) cells during cancer immunosurveillance. In immune surveillance, perforin and GZMB, the most potent cytotoxic molecules, act mainly as antitumoral and anti-infectious factors. However, when expressed by immune regulatory cells they may contribute to immune evasion of specific cancer types. By contrast, the other major granzyme, GZMA, seems not to play a major role in Tc/NK cell-mediated cytotoxicity, but acts as a proinflammatory cytokine that might contribute to cancer development. Members of the GZM family also regulate other biological processes unrelated to cell death, such as angiogenesis, vascular integrity, extracellular matrix remodeling, and barrier function, all of which contribute to cancer initiation and progression. Thus, a new paradigm is emerging in the field of oncoimmunology. Can GZMs act as protumoral factors under some circumstances? We review the diverse roles of GZMs in cancer progression, and new therapeutic opportunities emerging from targeting these protumoral roles.
Collapse
Affiliation(s)
- Maykel Arias
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain; These authors contributed equally to this work
| | - Luis Martínez-Lostao
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain; Department of Biochemistry and Molecular and Cell Biology, and Department of Microbiology, Preventive Medicine, and Public Health, University of Zaragoza, 50009 Zaragoza, Spain; Servicio de Inmunología Hospital Clínico Universitario Lorenzo Blesa, Zaragoza, Spain; Nanoscience Institute of Aragon (INA), University of Zaragoza, 50018 Zaragoza, Spain; These authors contributed equally to this work
| | - Llipsy Santiago
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Angel Ferrandez
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain; Servicio de Aparato Digestivo, Hospital Clínico Universitario Lorenzo Blesa, Zaragoza, Spain
| | - David J Granville
- International Collaboration on Repair Discoveries (ICORD), Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Julián Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain; Department of Biochemistry and Molecular and Cell Biology, and Department of Microbiology, Preventive Medicine, and Public Health, University of Zaragoza, 50009 Zaragoza, Spain; Nanoscience Institute of Aragon (INA), University of Zaragoza, 50018 Zaragoza, Spain; Aragon I+D Foundation (ARAID), Zaragoza, Spain.
| |
Collapse
|
38
|
Granzyme K‐deficient mice show no evidence of impaired antiviral immunity. Immunol Cell Biol 2017; 95:676-683. [DOI: 10.1038/icb.2017.35] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 04/13/2017] [Accepted: 04/13/2017] [Indexed: 01/16/2023]
|
39
|
van Eck JA, Shan L, Meeldijk J, Hack CE, Bovenschen N. A novel proinflammatory role for granzyme A. Cell Death Dis 2017; 8:e2630. [PMID: 28230859 PMCID: PMC5386495 DOI: 10.1038/cddis.2017.56] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Jacqueline A van Eck
- Department of Pathology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Liling Shan
- Department of Pathology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Jan Meeldijk
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - C Erik Hack
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| |
Collapse
|
40
|
Juno JA, van Bockel D, Kent SJ, Kelleher AD, Zaunders JJ, Munier CML. Cytotoxic CD4 T Cells-Friend or Foe during Viral Infection? Front Immunol 2017; 8:19. [PMID: 28167943 PMCID: PMC5253382 DOI: 10.3389/fimmu.2017.00019] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/05/2017] [Indexed: 01/03/2023] Open
Abstract
CD4 T cells with cytotoxic function were once thought to be an artifact due to long-term in vitro cultures but have in more recent years become accepted and reported in the literature in response to a number of viral infections. In this review, we focus on cytotoxic CD4 T cells in the context of human viral infections and in some infections that affect mice and non-human primates. We examine the effector mechanisms used by cytotoxic CD4 cells, the phenotypes that describe this population, and the transcription factors and pathways that lead to their induction following infection. We further consider the cells that are the predominant targets of this effector subset and describe the viral infections in which CD4 cytotoxic T lymphocytes have been shown to play a protective or pathologic role. Cytotoxic CD4 T cells are detected in the circulation at much higher levels than previously realized and are now recognized to have an important role in the immune response to viral infections.
Collapse
Affiliation(s)
- Jennifer A Juno
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne , Melbourne, VIC , Australia
| | - David van Bockel
- Immunovirology and Pathogenesis Program, The Kirby Institute for Infection and Immunity in Society, University of New South Wales Australia , Sydney, NSW , Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia; Melbourne Sexual Health Centre, Department of Infectious Diseases, Alfred Health, Central Clinical School, Monash University, Melbourne, VIC, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, VIC, Australia
| | - Anthony D Kelleher
- Immunovirology and Pathogenesis Program, The Kirby Institute for Infection and Immunity in Society, University of New South Wales Australia, Sydney, NSW, Australia; St Vincent's Hospital, Sydney, NSW, Australia
| | - John J Zaunders
- Immunovirology and Pathogenesis Program, The Kirby Institute for Infection and Immunity in Society, University of New South Wales Australia, Sydney, NSW, Australia; St Vincent's Hospital, Sydney, NSW, Australia
| | - C Mee Ling Munier
- Immunovirology and Pathogenesis Program, The Kirby Institute for Infection and Immunity in Society, University of New South Wales Australia , Sydney, NSW , Australia
| |
Collapse
|
41
|
Granzymes A and K differentially potentiate LPS-induced cytokine response. Cell Death Discov 2016; 2:16084. [PMID: 28028441 PMCID: PMC5149580 DOI: 10.1038/cddiscovery.2016.84] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 09/01/2016] [Accepted: 09/30/2016] [Indexed: 01/09/2023] Open
Abstract
Granzymes are serine proteases that, upon release from cytotoxic cells, induce apoptosis in tumor cells and virally infected cells. In addition, a role of granzymes in inflammation is emerging. Recently, we have demonstrated that extracellular granzyme K (GrK) potentiates lipopolysaccharide (LPS)-induced cytokine response from monocytes. GrK interacts with LPS, disaggregates LPS micelles, and stimulates LPS-CD14 binding and Toll-like receptor signaling. Here we show that human GrA also potentiates cytokine responses in human monocytes initiated by LPS or Gram-negative bacteria. Similar to GrK, this effect is independent of GrA catalytic activity. Unlike GrK, however, GrA does not bind to LPS, has little influence on LPS micelle disaggregation, and does not augment LPS-CD14 complex formation. We conclude that GrA and GrK differentially modulate LPS-Toll-like receptor signaling in monocytes, suggesting functional redundancy among cytotoxic lymphocyte proteases in the anti-bacterial innate immune response.
Collapse
|
42
|
Munier CML, van Bockel D, Bailey M, Ip S, Xu Y, Alcantara S, Liu SM, Denyer G, Kaplan W, Suzuki K, Croft N, Purcell A, Tscharke D, Cooper DA, Kent SJ, Zaunders JJ, Kelleher AD. The primary immune response to Vaccinia virus vaccination includes cells with a distinct cytotoxic effector CD4 T-cell phenotype. Vaccine 2016; 34:5251-5261. [PMID: 27639281 DOI: 10.1016/j.vaccine.2016.09.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 08/09/2016] [Accepted: 09/06/2016] [Indexed: 11/17/2022]
Abstract
BACKGROUND Smallpox was eradicated by a global program of inoculation with Vaccinia virus (VV). Robust VV-specific CD4 T-cell responses during primary infection are likely essential to controlling VV replication. Although there is increasing interest in cytolytic CD4 T-cells across many viral infections, the importance of these cells during acute VV infection is unclear. METHODS We undertook a detailed functional and genetic characterization of CD4 T-cells during acute VV-infection of humans. VV-specific T-cells were identified by up-regulation of activation markers directly ex vivo and through cytokine and co-stimulatory molecule expression. At day-13-post primary inoculation with VV, CD38highCD45RO+ CD4 T-cells were purified by cell sorting, RNA isolated and analysed by microarray. Differential expression of up-regulated genes in activated CD4 T-cells was confirmed at the mRNA and protein levels. We compared analyses of VV-specific CD4 T-cells to studies on 12 subjects with primary HIV infection (PHI). VV-specific T-cells lines were established from PBMCs collected post vaccination and checked for cytotoxicity potential. RESULTS A median 11.9% CD4 T-cells were CD38highCD45RO+ at day-13 post-VV inoculation, compared to 3.0% prior and 10.4% during PHI. Activated CD4 T-cells had an up-regulation of genes related to cytolytic function, including granzymes K and A, perforin, granulysin, TIA-1, and Rab27a. No difference was seen between CD4 T-cell expression of perforin or TIA-1 to VV and PHI, however granzyme k was more dominant in the VV response. At 25:1 effector to target ratio, two VV-specific T-cell lines exhibited 62% and 30% cytotoxicity respectively and CD107a degranulation. CONCLUSIONS We show for the first time that CD4 CTL are prominent in the early response to VV. Understanding the role of CD4 CTL in the generation of an effective anti-viral memory may help develop more effective vaccines for diseases such as HIV.
Collapse
Affiliation(s)
- C Mee Ling Munier
- The Kirby Institute for infection and immunity in society, UNSW Australia, Sydney, NSW, Australia.
| | - David van Bockel
- The Kirby Institute for infection and immunity in society, UNSW Australia, Sydney, NSW, Australia
| | - Michelle Bailey
- The Kirby Institute for infection and immunity in society, UNSW Australia, Sydney, NSW, Australia
| | - Susanna Ip
- The Kirby Institute for infection and immunity in society, UNSW Australia, Sydney, NSW, Australia
| | - Yin Xu
- The Kirby Institute for infection and immunity in society, UNSW Australia, Sydney, NSW, Australia
| | - Sheilajen Alcantara
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Sue Min Liu
- The Garvan Institute, Sydney, NSW, Australia
| | - Gareth Denyer
- School of Molecular Bioscience, Faculty of Science, The University of Sydney, NSW, Australia
| | | | | | - Kazuo Suzuki
- The Kirby Institute for infection and immunity in society, UNSW Australia, Sydney, NSW, Australia; St Vincent's Hospital, Sydney, NSW, Australia
| | - Nathan Croft
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Anthony Purcell
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - David Tscharke
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - David A Cooper
- The Kirby Institute for infection and immunity in society, UNSW Australia, Sydney, NSW, Australia; St Vincent's Hospital, Sydney, NSW, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia; Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Health, Central Clinical School, Monash University Melbourne, VIC, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, VIC, Australia
| | - John J Zaunders
- The Kirby Institute for infection and immunity in society, UNSW Australia, Sydney, NSW, Australia; St Vincent's Hospital, Sydney, NSW, Australia
| | - Anthony D Kelleher
- The Kirby Institute for infection and immunity in society, UNSW Australia, Sydney, NSW, Australia; St Vincent's Hospital, Sydney, NSW, Australia
| |
Collapse
|
43
|
van Kempen PMW, Noorlag R, Swartz JE, Bovenschen N, Braunius WW, Vermeulen JF, Van Cann EM, Grolman W, Willems SM. Oropharyngeal squamous cell carcinomas differentially express granzyme inhibitors. Cancer Immunol Immunother 2016; 65:575-85. [PMID: 26993499 PMCID: PMC4840222 DOI: 10.1007/s00262-016-1819-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 02/26/2016] [Indexed: 01/10/2023]
Abstract
Objectives Patients with human papillomavirus (HPV)-positive oropharyngeal squamous cell carcinomas (OPSCCs) have an improved prognosis compared to HPV-negative OPSCCs. Several theories have been proposed to explain this relatively good prognosis. One hypothesis is a difference in immune response. In this study, we compared tumor-infiltrating CD3+, CD4+, CD8+ T-cells, and granzyme inhibitors (SERPINB1, SERPINB4, and SERPINB9) between HPV-positive and HPV-negative tumors and the relation with survival. Methods Protein expression of tumor-infiltrating lymphocytes (TILs) (CD3, CD4, and CD8) and granzyme inhibitors was analyzed in 262 OPSCCs by immunohistochemistry (IHC). Most patients (67 %) received primary radiotherapy with or without chemotherapy. Cox regression analysis was carried out to compare overall survival (OS) of patients with low and high TIL infiltration and expression of granzyme inhibitors. Results HPV-positive OPSCCs were significantly more heavily infiltrated by TILs (p < 0.001) compared to HPV-negative OPSCCs. A high level of CD3+ TILs was correlated with a favorable outcome in the total cohort and in HPV-positive OPSCCs, while it reached no significance in HPV-negative OPSCCs. There was expression of all three granzyme inhibitors in OPSCCs. No differences in expression were found between HPV-positive and HPV-negative OPSCCs. Within the group of HPV-positive tumors, a high expression of SERPINB1 was associated with a significantly worse overall survival. Conclusion HPV-positive OPSCCs with a low count of CD3+ TILs or high expression of SERPINB1 have a worse OS, comparable with HPV-negative OPSCCs. This suggests that the immune system plays an important role in the carcinogenesis of the virally induced oropharynx tumors.
Collapse
Affiliation(s)
- Pauline M W van Kempen
- Department of Otorhinolaryngology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| | - Rob Noorlag
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Justin E Swartz
- Department of Otorhinolaryngology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Weibel W Braunius
- Department of Otorhinolaryngology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.,Department of Head and Neck Surgical Oncology, Cancer Center University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Jeroen F Vermeulen
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Ellen M Van Cann
- Department of Head and Neck Surgical Oncology, Cancer Center University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Wilko Grolman
- Department of Otorhinolaryngology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Stefan M Willems
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| |
Collapse
|
44
|
Pediatric Primitive Neuroectodermal Tumors of the Central Nervous System Differentially Express Granzyme Inhibitors. PLoS One 2016; 11:e0151465. [PMID: 26963506 PMCID: PMC4786147 DOI: 10.1371/journal.pone.0151465] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 02/29/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Central nervous system (CNS) primitive neuroectodermal tumors (PNETs) are malignant primary brain tumors that occur in young infants. Using current standard therapy, up to 80% of the children still dies from recurrent disease. Cellular immunotherapy might be key to improve overall survival. To achieve efficient killing of tumor cells, however, immunotherapy has to overcome cancer-associated strategies to evade the cytotoxic immune response. Whether CNS-PNETs can evade the immune response remains unknown. METHODS We examined by immunohistochemistry the immune response and immune evasion strategies in pediatric CNS-PNETs. RESULTS Here, we show that CD4+, CD8+, γδ-T-cells, and Tregs can infiltrate pediatric CNS-PNETs, although the activation status of cytotoxic cells is variable. Pediatric CNS-PNETs evade immune recognition by downregulating cell surface MHC-I and CD1d expression. Intriguingly, expression of SERPINB9, SERPINB1, and SERPINB4 is acquired during tumorigenesis in 29%, 29%, and 57% of the tumors, respectively. CONCLUSION We show for the first time that brain tumors express direct granzyme inhibitors (serpins) as a potential mechanism to overcome cellular cytotoxicity, which may have consequences for cellular immunotherapy.
Collapse
|
45
|
Wensink AC, Wiewel MA, Jongeneel LH, Boes M, van der Poll T, Hack CE, Bovenschen N. Granzyme M and K release in human experimental endotoxemia. Immunobiology 2016; 221:773-7. [PMID: 26928607 DOI: 10.1016/j.imbio.2016.02.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/16/2016] [Accepted: 02/16/2016] [Indexed: 01/31/2023]
Abstract
Granzymes are serine proteases involved in killing of tumor cells and virally infected cells. However, granzymes are also upregulated in blood under inflammatory conditions and contribute to cytokine release and processing. Here, we show that granzyme M (GrM) and to a lesser extent GrK are transiently elevated in the circulation following LPS administration in humans. GrM is released upon stimulation of whole blood with LPS or the gram-negative bacteria Escherichia coli BL21, Pseudomonas aeruginosa, and Neisseria meningitidis. GrK is only released upon stimulation with P. aeruginosa. Thus, GrM and GrK are differentially released in response to LPS and gram-negative bacteria.
Collapse
Affiliation(s)
- Annette C Wensink
- Department of Pathology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands; Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Maryse A Wiewel
- Center for Experimental and Molecular Medicine, Amsterdam Medical Center, University of Amsterdam, Amsterdam 1100 DD, The Netherlands
| | - Lieneke H Jongeneel
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands; Department of Pediatrics, University Medical Center Utrecht, Utrecht 3584CX, The Netherlands
| | - Marianne Boes
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands; Department of Pediatrics, University Medical Center Utrecht, Utrecht 3584CX, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Amsterdam Medical Center, University of Amsterdam, Amsterdam 1100 DD, The Netherlands
| | - C Erik Hack
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands; Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands.
| |
Collapse
|
46
|
Akula S, Thorpe M, Boinapally V, Hellman L. Granule Associated Serine Proteases of Hematopoietic Cells - An Analysis of Their Appearance and Diversification during Vertebrate Evolution. PLoS One 2015; 10:e0143091. [PMID: 26569620 PMCID: PMC4646688 DOI: 10.1371/journal.pone.0143091] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 10/20/2015] [Indexed: 12/02/2022] Open
Abstract
Serine proteases are among the most abundant granule constituents of several hematopoietic cell lineages including mast cells, neutrophils, cytotoxic T cells and NK cells. These proteases are stored in their active form in the cytoplasmic granules and in mammals are encoded from four different chromosomal loci: the chymase locus, the met-ase locus, the T cell tryptase and the mast cell tryptase locus. In order to study their appearance during vertebrate evolution we have performed a bioinformatic analysis of related genes and gene loci from a large panel of metazoan animals from sea urchins to placental mammals for three of these loci: the chymase, met-ase and granzyme A/K loci. Genes related to mammalian granzymes A and K were the most well conserved and could be traced as far back to cartilaginous fish. Here, the granzyme A and K genes were found in essentially the same chromosomal location from sharks to humans. However in sharks, no genes clearly identifiable as members of the chymase or met-ase loci were found. A selection of these genes seemed to appear with bony fish, but sometimes in other loci. Genes related to mammalian met-ase locus genes were found in bony fish. Here, the most well conserved member was complement factor D. However, genes distantly related to the neutrophil proteases were also identified in this locus in several bony fish species, indicating that this locus is also old and appeared at the base of bony fish. In fish, a few of the chymase locus-related genes were found in a locus with bordering genes other than the mammalian chymase locus and some were found in the fish met-ase locus. This indicates that a convergent evolution rather than divergent evolution has resulted in chymase locus-related genes in bony fish.
Collapse
Affiliation(s)
- Srinivas Akula
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, The Biomedical Center, Box 596, SE-751 24, Uppsala, Sweden
| | - Michael Thorpe
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, The Biomedical Center, Box 596, SE-751 24, Uppsala, Sweden
| | - Vamsi Boinapally
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, The Biomedical Center, Box 596, SE-751 24, Uppsala, Sweden
| | - Lars Hellman
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, The Biomedical Center, Box 596, SE-751 24, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
47
|
|
48
|
Dotiwala F, Fellay I, Filgueira L, Martinvalet D, Lieberman J, Walch M. A High Yield and Cost-efficient Expression System of Human Granzymes in Mammalian Cells. J Vis Exp 2015:e52911. [PMID: 26132420 DOI: 10.3791/52911] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
When cytotoxic T lymphocytes (CTL) or natural killer (NK) cells recognize tumor cells or cells infected with intracellular pathogens, they release their cytotoxic granule content to eliminate the target cells and the intracellular pathogen. Death of the host cells and intracellular pathogens is triggered by the granule serine proteases, granzymes (Gzms), delivered into the host cell cytosol by the pore forming protein perforin (PFN) and into bacterial pathogens by the prokaryotic membrane disrupting protein granulysin (GNLY). To investigate the molecular mechanisms of target cell death mediated by the Gzms in experimental in-vitro settings, protein expression and purification systems that produce high amounts of active enzymes are necessary. Mammalian secreted protein expression systems imply the potential to produce correctly folded, fully functional protein that bears posttranslational modification, such as glycosylation. Therefore, we used a cost-efficient calcium precipitation method for transient transfection of HEK293T cells with human Gzms cloned into the expression plasmid pHLsec. Gzm purification from the culture supernatant was achieved by immobilized nickel affinity chromatography using the C-terminal polyhistidine tag provided by the vector. The insertion of an enterokinase site at the N-terminus of the protein allowed the generation of active protease that was finally purified by cation exchange chromatography. The system was tested by producing high levels of cytotoxic human Gzm A, B and M and should be capable to produce virtually every enzyme in the human body in high yields.
Collapse
Affiliation(s)
- Farokh Dotiwala
- Cellular and Molecular Medicine Program, Boston Children's Hospital and Harvard Medical School
| | | | | | | | - Judy Lieberman
- Cellular and Molecular Medicine Program, Boston Children's Hospital and Harvard Medical School
| | | |
Collapse
|
49
|
Wensink AC, Hack CE, Bovenschen N. Granzymes regulate proinflammatory cytokine responses. THE JOURNAL OF IMMUNOLOGY 2015; 194:491-7. [PMID: 25556251 DOI: 10.4049/jimmunol.1401214] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Granzymes (Grs) are serine proteases mainly produced by cytotoxic lymphocytes and are traditionally considered to cause apoptosis in tumor cells and virally infected cells. However, the cytotoxicity of several Grs is currently being debated, and additional, predominantly extracellular, functions of Grs in inflammation are emerging. Extracellular soluble Grs are elevated in the circulation of patients with autoimmune diseases and infections. Additionally, Grs are expressed by several types of immune cells other than cytotoxic lymphocytes. Recent research has revealed novel immunomodulatory functions of Grs. In this review, we provide a comprehensive overview on the role of Grs in inflammation, highlighting their role in cytokine induction and processing.
Collapse
Affiliation(s)
- Annette C Wensink
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; and Laboratory of Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - C Erik Hack
- Laboratory of Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; and Laboratory of Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| |
Collapse
|
50
|
Infection history determines the differentiation state of human CD8+ T cells. J Virol 2015; 89:5110-23. [PMID: 25717102 DOI: 10.1128/jvi.03478-14] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 02/17/2015] [Indexed: 01/17/2023] Open
Abstract
UNLABELLED After the resolution of the acute phase of infection, otherwise quiescent antigen-experienced CD8(+) T cells confer rapid protection upon reinfection with viral pathogens or, in the case of persistent viruses, help to maintain control of the infection. Depending on the type of virus, antigen-specific CD8(+) T cells have distinct traits, ranging from typical memory cell properties in the case of rapidly cleared viruses to immediate effector functions for persistent viruses. We here show that both the differentiation stage, defined by the expression of cell surface markers, such as CD45RA, CCR7, CD28, and CD27, and distinct expression levels of T-bet and eomesodermin (Eomes) predict the functional profile of antigen-experienced CD8(+) T cells. Furthermore, virus-specific CD8(+) T cells targeting different respiratory syncytial virus-, influenza A virus-, Epstein-Barr virus (EBV)-, human cytomegalovirus (hCMV)-, and HIV-1-specific epitopes adopt distinct T-bet and Eomes expression patterns that appear to be installed early during the primary response. Importantly, the associations between surface phenotype, T-bet/Eomes expression levels, and the expression of markers that predict CD8(+) T-cell function change according to viral infection history, particularly against the background of HIV-1 and, to lesser extent, of human cytomegalovirus and/or Epstein-Barr virus infection. Thus, the functionality of human antigen-experienced CD8(+) T cells follows at least two dimensions, one outlined by the surface phenotype and another by the T-bet/Eomes expression levels, which are determined by previous or persistent viral challenges. IMPORTANCE Functional human CD8(+) T-cell subsets have been defined using surface markers like CD45RA, CCR7, CD28, and CD27. However, the induction of function-defining traits, like granzyme B expression, is controlled by transcription factors like T-bet and Eomes. Here, we describe how T-bet and Eomes levels distinctly relate to the expression of molecules predictive for CD8(+) T-cell function in a surface phenotype-independent manner. Importantly, we found that central memory and effector memory CD8(+) T-cell subsets differentially express T-bet, Eomes, and molecules predictive for function according to viral infection history, particularly so in the context of HIV-1 infection and, to lesser extent, of latent EBV- and/or hCMV-infected, otherwise healthy adults. Finally, we show that the distinct phenotypes and T-bet/Eomes levels of different virus-specific CD8(+) T-cell populations are imprinted early during the acute phase of primary infection in vivo. These findings broaden our understanding of CD8(+) T-cell differentiation.
Collapse
|