1
|
Deng K, Yuan L, Xu Z, Qin F, Zheng Z, Huang L, Jiang W, Qin J, Sun Y, Zheng T, Ou X, Zheng L, Li S. Study of LY9 as a potential biomarker for prognosis and prediction of immunotherapy efficacy in lung adenocarcinoma. PeerJ 2024; 12:e17816. [PMID: 39193519 PMCID: PMC11348898 DOI: 10.7717/peerj.17816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/04/2024] [Indexed: 08/29/2024] Open
Abstract
Background Lymphocyte antigen 9 (LY9) participates in the development of several tumors and diseases but has not been reported yet in lung adenocarcinoma (LUAD). Methods First, we analyzed the expression and prognostic value of LY9 in pan-cancer, including LUAD. Additionally, we conducted a correlation analysis of LY9 expression in LUAD with immune cell infiltration using the TIMER database and the CIBERSORT algorithm, and with immune checkpoints using the GEPIA database. Also, we constructed a potential ceRNA network for LY9. Furthermore, we explored LY9-related pathways by Gene Set Enrichment Analysis (GSEA). Finally, validation of differential expression at the mRNA level was obtained from the GEO database. We collected LUAD tissues for Quantitative Real-time PCR (qRT-PCR) to verify the expression of LY9, CD8, and CD4 and calculated the correlation between them. We also conducted immunohistochemistry (IHC) to verify the protein expression of LY9. Results Results showed that LY9 was highly expressed in various tumors, including LUAD. Besides, patients with high LY9 expression presented longer overall survival (OS) and more multiple lymphocyte infiltrations. The expression of LY9 in LUAD strongly and positively correlates with multiple immune cell infiltration and immune checkpoints. The functional enrichment analysis indicated that LY9 was involved in multiple immune-related pathways and non-small cell lung cancer. Moreover, a ceRNA regulatory network of LINC00943-hsa-miR-141-3p-LY9 might be involved. Finally, GSE68465 dataset confirmed differential expression of LY9 mRNA levels in LUAD and the qRT-PCR results verified LY9 had a strong and positive correlation with CD4 and CD8 T cells. Unfortunately, IHC did not detect the expression of LY9 protein level in tumor tissues and WB experiments validated the protein expression of LY9 in the OCI-AML-2 cell line. Conclusions Therefore, we hypothesized that LY9 could serve as a potential, novel prognostic biomarker for LUAD and could predict immunotherapy efficacy at the mRNA level.
Collapse
Affiliation(s)
- Kun Deng
- Department of Thoracic and Cardiovascular Surgery, The Second People’s Hospital of Neijiang, Neijiang, Sichuan, China
| | - Liqiang Yuan
- Department of Thoracic and Cardiovascular Surgery, People’s Hospital of Deyang, Deyang, Sichuan, China
| | - Zhanyu Xu
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Fanglu Qin
- Department of Scientific Research, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhiwen Zheng
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liuliu Huang
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wei Jiang
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Junqi Qin
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yu Sun
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tiaozhan Zheng
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xinhuai Ou
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liping Zheng
- Catheterization Laboratory of Cardiovascular Institute, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Shikang Li
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
2
|
Snow KS, Raburn DJ, Price TM. Differential Gene Regulation of the Human Blastocyst Trophectoderm and Inner Cell Mass by Progesterone. Reprod Sci 2024; 31:1363-1372. [PMID: 38151652 DOI: 10.1007/s43032-023-01429-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/11/2023] [Indexed: 12/29/2023]
Abstract
Knowledge of action of progesterone (P4) on the human preimplantation embryo is lacking. The objective of this study was to determine expression of a mitochondrial P4 receptor (PR-M) in the trophectoderm (TE) and the inner cell mass (ICM) of the human blastocyst and to determine P4-induced gene expression during growth from the cleavage to the blastocyst stage. Previously cryopreserved cleavage stage embryos were treated with P4 (10-6 M) or vehicle until blastocyst development. Cells from the TE and the ICM of dissected euploid embryos underwent RNA-seq analysis, while other embryos were used for analysis of nuclear PR (nPR) and PR-M expression.PR-M expression was confirmed in the TE, the ICM, and a human embryonic stem cell line (HESC). Conversely, nPR expression was absent in the TE and the ICM with low expression in the HESC line. RNA-seq analysis revealed P4 effects greater in the TE with 183 significant pathway changes compared to 27 in the ICM. The TE response included significant upregulation of genes associated with DNA replication, cell cycle phase transition and others, exemplified by a 7.6-fold increase in the cell proliferation gene, F-Box Associated Domain Containing. The majority of ICM pathways were downregulated including chromosome separation, centromere complex assembly and chromatin remodeling at centromere. This study confirms that human blastocysts express PR-M in both the TE and the ICM, but lack expression of nPR. P4-induced gene regulation differs greatly in the two cell fractions with the predominant effect of cell proliferation in the TE and not the ICM.
Collapse
Affiliation(s)
- Kathryn Shaia Snow
- Division of Reproductive Endocrinology and Infertility Department of Obstetrics and Gynecology, Duke University, 5601 Arringdon Park Dr Suite 210, Morrisville, NC, 27560, USA
| | - Douglas J Raburn
- Division of Reproductive Endocrinology and Infertility Department of Obstetrics and Gynecology, Duke University, 5601 Arringdon Park Dr Suite 210, Morrisville, NC, 27560, USA
| | - Thomas M Price
- Division of Reproductive Endocrinology and Infertility Department of Obstetrics and Gynecology, Duke University, 5601 Arringdon Park Dr Suite 210, Morrisville, NC, 27560, USA.
| |
Collapse
|
3
|
Tojjari A, Giles FJ, Vilbert M, Saeed A, Cavalcante L. SLAM Modification as an Immune-Modulatory Therapeutic Approach in Cancer. Cancers (Basel) 2023; 15:4808. [PMID: 37835502 PMCID: PMC10571764 DOI: 10.3390/cancers15194808] [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: 09/08/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
In the field of oncology, the Signaling Lymphocyte Activation Molecule (SLAM) family is emerging as pivotal in modulating immune responses within tumor environments. The SLAM family comprises nine receptors, mainly found on immune cell surfaces. These receptors play complex roles in the interaction between cancer and the host immune system. Research suggests SLAM's role in both enhancing and dampening tumor-immune responses, influencing the progression and treatment outcomes of various cancers. As immunotherapy advances, resistance remains an issue. The nuanced roles of the SLAM family might provide answers. With the rise in technologies like single-cell RNA sequencing and advanced imaging, there is potential for precise SLAM-targeted treatments. This review stresses patient safety, the importance of thorough clinical trials, and the potential of SLAM-focused therapies to transform cancer care. In summary, SLAM's role in oncology signals a new direction for more tailored and adaptable cancer treatments.
Collapse
Affiliation(s)
- Alireza Tojjari
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15213, USA; (A.T.); (M.V.)
| | | | - Maysa Vilbert
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15213, USA; (A.T.); (M.V.)
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15213, USA; (A.T.); (M.V.)
| | | |
Collapse
|
4
|
Cao L, Morgun E, Genardi S, Visvabharathy L, Cui Y, Huang H, Wang CR. METTL14-dependent m 6A modification controls iNKT cell development and function. Cell Rep 2022; 40:111156. [PMID: 35926466 PMCID: PMC9495716 DOI: 10.1016/j.celrep.2022.111156] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 06/06/2022] [Accepted: 07/13/2022] [Indexed: 11/25/2022] Open
Abstract
N6-methyladenosine (m6A), the most common form of RNA modification, controls CD4+ T cell homeostasis by targeting the IL-7/STAT5/SOCS signaling pathways. The role of m6A modification in unconventional T cell development remains unknown. Using mice with T cell-specific deletion of RNA methyltransferase METTL14 (T-Mettl14−/−), we demonstrate that m6A modification is indispensable for iNKT cell homeostasis. Loss of METTL14-dependent m6A modification leads to the upregulation of apoptosis in double-positive thymocytes, which in turn decreases Vα14-Jα18 gene rearrangements, resulting in drastic reduction of iNKT numbers in the thymus and periphery. Residual T-Mettl14−/− iNKT cells exhibit increased apoptosis, impaired maturation, and decreased responsiveness to IL-2/IL-15 and TCR stimulation. Furthermore, METTL14 knockdown in mature iNKT cells diminishes their cytokine production, correlating with increased Cish expression and decreased TCR signaling. Collectively, our study highlights a critical role for METTL14-dependent-m6A modification in iNKT cell development and function. Cao et al. show that T cell-specific deletion of METTL14, a component of RNA m6A writer complex, leads to severe defects in iNKT cell development, survival, and function. Mechanistically, METTL14-dependent m6A modification controls iNKT cell development in a cell-intrinsic manner by regulating the apoptosis pathway and TCR signaling pathway.
Collapse
Affiliation(s)
- Liang Cao
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, 320 E. Superior Street, Searle 3-401, Chicago, IL 60611, USA
| | - Eva Morgun
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, 320 E. Superior Street, Searle 3-401, Chicago, IL 60611, USA
| | - Samantha Genardi
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, 320 E. Superior Street, Searle 3-401, Chicago, IL 60611, USA
| | - Lavanya Visvabharathy
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, 320 E. Superior Street, Searle 3-401, Chicago, IL 60611, USA
| | - Yongyong Cui
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, 320 E. Superior Street, Searle 3-401, Chicago, IL 60611, USA
| | - Haochu Huang
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Chyung-Ru Wang
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, 320 E. Superior Street, Searle 3-401, Chicago, IL 60611, USA.
| |
Collapse
|
5
|
Roncador G, Puñet-Ortiz J, Maestre L, Rodríguez-Lobato LG, Jiménez S, Reyes-García AI, García-González Á, García JF, Piris MÁ, Montes-Moreno S, Rodríguez-Justo M, Mena MP, Fernández de Larrea C, Engel P. CD229 (Ly9) a Novel Biomarker for B-Cell Malignancies and Multiple Myeloma. Cancers (Basel) 2022; 14:2154. [PMID: 35565280 PMCID: PMC9101303 DOI: 10.3390/cancers14092154] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 02/04/2023] Open
Abstract
CD229 (Ly9) homophilic receptor, which belongs to the SLAM family of cell-surface molecules, is predominantly expressed on B and T cells. It acts as a signaling molecule, regulating lymphocyte homoeostasis and activation. Studies of CD229 function indicate that this receptor functions as a regulator of the development of marginal-zone B cells and other innate-like T and B lymphocytes. The expression on leukemias and lymphomas remains poorly understood due to the lack of CD229 monoclonal antibodies (mAb) for immunohistochemistry application (IHC). In this study, we used a new mAb against the cytoplasmic region of CD229 to study the expression of CD229 on normal tissues and B-cell malignancies, including multiple myeloma (MM), using tissue microarrays. We showed CD229 to be restricted to hematopoietic cells. It was strongly expressed in all cases of MM and in most marginal-zone lymphomas (MZL). Moderate CD229 expression was also found in chronic lymphocyte leukemia (CLL), follicular (FL), classic mantle-cell (MCL) and diffuse large B-cell lymphoma. Given the high expression on myeloma cells, we also analyzed for the presence of soluble CD229 in the sera of these patients. Serum levels of soluble CD229 (sCD229) at the time of diagnosis in MM patients could be useful as a prognostic biomarker. In conclusion, our results indicate that CD229 represents not only a useful biomarker but also an attractive therapeutic target.
Collapse
Affiliation(s)
- Giovanna Roncador
- Monoclonal Antibodies Unit, Biotechnology Program, Spanish National Cancer Centre (CNIO), Centro de Investigación Médica en red Cancer (CIBERONC), 28029 Madrid, Spain; (G.R.); (L.M.); (S.J.); (A.I.R.-G.); (Á.G.-G.)
| | - Joan Puñet-Ortiz
- Immunology Unit, Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Barcelona, Casanova 172, 08036 Barcelona, Spain;
| | - Lorena Maestre
- Monoclonal Antibodies Unit, Biotechnology Program, Spanish National Cancer Centre (CNIO), Centro de Investigación Médica en red Cancer (CIBERONC), 28029 Madrid, Spain; (G.R.); (L.M.); (S.J.); (A.I.R.-G.); (Á.G.-G.)
| | - Luis Gerardo Rodríguez-Lobato
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (L.G.R.-L.); (M.-P.M.); (C.F.d.L.)
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Scherezade Jiménez
- Monoclonal Antibodies Unit, Biotechnology Program, Spanish National Cancer Centre (CNIO), Centro de Investigación Médica en red Cancer (CIBERONC), 28029 Madrid, Spain; (G.R.); (L.M.); (S.J.); (A.I.R.-G.); (Á.G.-G.)
| | - Ana Isabel Reyes-García
- Monoclonal Antibodies Unit, Biotechnology Program, Spanish National Cancer Centre (CNIO), Centro de Investigación Médica en red Cancer (CIBERONC), 28029 Madrid, Spain; (G.R.); (L.M.); (S.J.); (A.I.R.-G.); (Á.G.-G.)
| | - Álvaro García-González
- Monoclonal Antibodies Unit, Biotechnology Program, Spanish National Cancer Centre (CNIO), Centro de Investigación Médica en red Cancer (CIBERONC), 28029 Madrid, Spain; (G.R.); (L.M.); (S.J.); (A.I.R.-G.); (Á.G.-G.)
| | - Juan F. García
- Department of Pathology, MD Anderson Cancer Center Madrid, Centro de Investigación Médica en Red Cancer (CIBERONC), 28040 Madrid, Spain;
| | - Miguel Ángel Piris
- Department of Pathology, Fundación Jiménez Díaz, Centro de Investigación Médica en Red Cancer (CIBERONC), 28040 Madrid, Spain;
| | - Santiago Montes-Moreno
- Translational Haematopathology Laboratory, Anatomic Pathology Department, Hospital Universitario Marqués de Valdecilla/IDIVAL, CIBERONC, 39008 Santander, Spain;
| | - Manuel Rodríguez-Justo
- Department of Research Pathology, Cancer Institute, University Collage London, London WC1E 6DD, UK;
| | - Mari-Pau Mena
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (L.G.R.-L.); (M.-P.M.); (C.F.d.L.)
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Carlos Fernández de Larrea
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (L.G.R.-L.); (M.-P.M.); (C.F.d.L.)
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Pablo Engel
- Immunology Unit, Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Barcelona, Casanova 172, 08036 Barcelona, Spain;
| |
Collapse
|
6
|
T cell subtype profiling measures exhaustion and predicts anti-PD-1 response. Sci Rep 2022; 12:1342. [PMID: 35079117 PMCID: PMC8789795 DOI: 10.1038/s41598-022-05474-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/05/2022] [Indexed: 12/11/2022] Open
Abstract
Anti-PD-1 therapy can provide long, durable benefit to a fraction of patients. The on-label PD-L1 test, however, does not accurately predict response. To build a better biomarker, we created a method called T Cell Subtype Profiling (TCSP) that characterizes the abundance of T cell subtypes (TCSs) in FFPE specimens using five RNA models. These TCS RNA models are created using functional methods, and robustly discriminate between naïve, activated, exhausted, effector memory, and central memory TCSs, without the reliance on non-specific, classical markers. TCSP is analytically valid and corroborates associations between TCSs and clinical outcomes. Multianalyte biomarkers based on TCS estimates predicted response to anti-PD-1 therapy in three different cancers and outperformed the indicated PD-L1 test, as well as Tumor Mutational Burden. Given the utility of TCSP, we investigated the abundance of TCSs in TCGA cancers and created a portal to enable researchers to discover other TCSP-based biomarkers.
Collapse
|
7
|
Quan Q, Xiong X, Wu S, Yu M. Identification of Immune-Related Key Genes in Ovarian Cancer Based on WGCNA. Front Genet 2021; 12:760225. [PMID: 34868239 PMCID: PMC8634599 DOI: 10.3389/fgene.2021.760225] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/20/2021] [Indexed: 12/21/2022] Open
Abstract
Background: Ovarian cancer (OV) is a fatal gynecologic malignancy and has poor survival rate in women over the age of forty. In our study, we aimed to identify genes related to immune microenvironment regulations and explore genes associated with OV prognosis. Methods: The RNA-seq data of GDC TCGA Ovarian Cancer cohort of 376 patients was retrieved from website. Weighted gene co-expression network analysis (WGCNA) and ESTIMATE algorithm were applied to identify the key genes associated with the immune scores. The correlation between key genes and 22 immune cell types were estimated by using CIBERSORT algorithms. Results: WGCNA showed that the pink module was most correlated with the immune score. Seven of 14 key genes (FCRL3, IFNG, KCNA3, LY9, PLA2G2D, THEMIS, and TRAT1) were significantly associated with the OS of OV patients. Correlation analysis showed our key genes positively related to M1 macrophages, CD8 T cells, plasma cells, regulatory T (Treg) cells and activated memory CD4 T cells, and negatively related to naive CD4 T cells, M0 macrophages, activated dendritic cells (DCs) and memory B cells. Kaplan-Meier survival analysis showed that lower abundances of neutrophils and higher abundances of M1 macrophages, plasma cells, T cells gamma delta (γδT) cells and follicular helper T (Tfh) cells predicted better OV prognosis. Conclusion: Forteen key genes related to the immune infiltrating of OV were identified, and seven of them were significantly related to prognosis. These key genes have potential roles in tumor infiltrating immune cells differentiation and proliferation. This study provided potential prognostic markers and immunotherapy targets for OV.
Collapse
Affiliation(s)
- Qingli Quan
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,School of Life Sciences, Fudan University, Shanghai, China
| | - Xinxin Xiong
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Shanyun Wu
- Department of Biology, Faculty of Science, University of British Columbia, Vancouver, BC, Canada
| | - Meixing Yu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
8
|
Agarwal S, Sharma A, Bouzeyen R, Deep A, Sharma H, Mangalaparthi KK, Datta KK, Kidwai S, Gowda H, Varadarajan R, Sharma RD, Thakur KG, Singh R. VapBC22 toxin-antitoxin system from Mycobacterium tuberculosis is required for pathogenesis and modulation of host immune response. SCIENCE ADVANCES 2020; 6:eaba6944. [PMID: 32537511 PMCID: PMC7269643 DOI: 10.1126/sciadv.aba6944] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 04/02/2020] [Indexed: 05/16/2023]
Abstract
Virulence-associated protein B and C toxin-antitoxin (TA) systems are widespread in prokaryotes, but their precise role in physiology is poorly understood. We have functionally characterized the VapBC22 TA system from Mycobacterium tuberculosis. Transcriptome analysis revealed that overexpression of VapC22 toxin in M. tuberculosis results in reduced levels of metabolic enzymes and increased levels of ribosomal proteins. Proteomics studies showed reduced expression of virulence-associated proteins and increased levels of cognate antitoxin, VapB22 in the ΔvapC22 mutant strain. Furthermore, both the ΔvapC22 mutant and VapB22 overexpression strains of M. tuberculosis were susceptible to killing upon exposure to oxidative stress and showed attenuated growth in guinea pigs and mice. Host transcriptome analysis suggests upregulation of the transcripts involved in innate immune responses and tissue remodeling in mice infected with the ΔvapC22 mutant strain. Together, we demonstrate that the VapBC22 TA system belongs to a key regulatory network and is essential for M. tuberculosis pathogenesis.
Collapse
Affiliation(s)
- Sakshi Agarwal
- Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Harya na-121001, India
| | - Arun Sharma
- Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Harya na-121001, India
| | - Rania Bouzeyen
- Institut Pasteur de Tunis, LTCII, LR11IPT02, Tunis 1002, Tunisia
| | - Amar Deep
- Structural Biology Laboratory, Council of Scientific and Industrial Research–Institute of Microbial Technology, Chandigarh 160036, India
| | - Harsh Sharma
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurugr am-122413, India
| | | | | | - Saqib Kidwai
- Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Harya na-121001, India
| | - Harsha Gowda
- Institute of Bioinformatics, Bangalore 560066, India
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | | | - Ravi Datta Sharma
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurugr am-122413, India
| | - Krishan Gopal Thakur
- Structural Biology Laboratory, Council of Scientific and Industrial Research–Institute of Microbial Technology, Chandigarh 160036, India
| | - Ramandeep Singh
- Tuberculosis Research Laboratory, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Harya na-121001, India
- Corresponding author.
| |
Collapse
|
9
|
SLAM-SAP-Fyn: Old Players with New Roles in iNKT Cell Development and Function. Int J Mol Sci 2019; 20:ijms20194797. [PMID: 31569599 PMCID: PMC6801923 DOI: 10.3390/ijms20194797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 12/25/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are a unique T cell lineage that develop in the thymus and emerge with a memory-like phenotype. Accordingly, following antigenic stimulation, they can rapidly produce copious amounts of Th1 and Th2 cytokines and mediate activation of several immune cells. Thus, it is not surprising that iNKT cells play diverse roles in a broad range of diseases. Given their pivotal roles in host immunity, it is crucial that we understand the mechanisms that govern iNKT cell development and effector functions. Over the last two decades, several studies have contributed to the current knowledge of iNKT cell biology and activity. Collectively, these studies reveal that the thymic development of iNKT cells, their lineage expansion, and functional properties are tightly regulated by a complex network of transcription factors and signaling molecules. While prior studies have clearly established the importance of the SLAM-SAP-Fyn signaling axis in iNKT cell ontogenesis, recent studies provide exciting mechanistic insights into the role of this signaling cascade in iNKT cell development, lineage fate decisions, and functions. Here we summarize the previous literature and discuss the more recent studies that guide our understanding of iNKT cell development and functional responses.
Collapse
|
10
|
Wang N, Yigit B, van der Poel CE, Cuenca M, Carroll MC, Herzog RW, Engel P, Terhorst C. The Checkpoint Regulator SLAMF3 Preferentially Prevents Expansion of Auto-Reactive B Cells Generated by Graft-vs.-Host Disease. Front Immunol 2019; 10:831. [PMID: 31057553 PMCID: PMC6482334 DOI: 10.3389/fimmu.2019.00831] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/29/2019] [Indexed: 12/16/2022] Open
Abstract
Absence of the mouse cell surface receptor SLAMF3 in SLAMF3-/- mice suggested that this receptor negatively regulates B cell homeostasis by modulating activation thresholds of B cell subsets. Here, we examine whether anti-SLAMF3 affects both B and T cell subsets during immune responses to haptenated ovalbumin [NP-OVA] and in the setting of chronic graft vs. host disease (cGVHD) induced by transferring B6.C-H2bm12/KhEg (bm12) CD4+ T cells into B6 WT mice. We find that administering αSLAMF3 to NP-OVA immunized B6 mice primarily impairs antibody responses and Germinal center B cell [GC B] numbers, whilst CXCR5+, PD-1+, and ICOS+ T follicular helper (TFH) cells are not significantly affected. By contrast, administering αSLAMF3 markedly enhanced autoantibody production upon induction of cGVHD by the transfer of bm12 CD4+ T cells into B6 recipients. Surprisingly, αSLAMF3 accelerated both the differentiation of GC B and donor-derived TFH cells initiated by cGVHD. The latter appeared to be induced by decreased numbers of donor-derived Treg and T follicular regulatory (TFR) cells. Collectively, these data show that control of anti-SLAMF3-induced signaling is requisite to prevent autoantibody responses during cGVHD, but reduces responses to foreign antigens.
Collapse
Affiliation(s)
- Ninghai Wang
- Division of Immunology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Burcu Yigit
- Division of Immunology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Cees E van der Poel
- Program in Cellular and Molecular Medicine, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Marta Cuenca
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Michael C Carroll
- Program in Cellular and Molecular Medicine, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Roland W Herzog
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Pablo Engel
- Immunology Unit, Department of Cell Biology, Immunology and Neurosciences, Medical School, University of Barcelona, Barcelona, Spain
| | - Cox Terhorst
- Division of Immunology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, United States
| |
Collapse
|
11
|
Angulo A, Cuenca M, Martínez-Vicente P, Engel P. Viral CD229 (Ly9) homologs as new manipulators of host immunity. J Leukoc Biol 2019; 105:947-954. [PMID: 30791129 DOI: 10.1002/jlb.2mr1018-413r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/30/2019] [Accepted: 02/01/2019] [Indexed: 12/27/2022] Open
Abstract
The signaling lymphocytic activation molecule family (SLAMF) of receptors plays crucial roles during innate and adaptive immune responses. The SLAMF member CD229 (Ly9, SLAMF3) is a homophilic receptor predominantly expressed on the surface of B and T cells. CD229 acts as a cosignaling molecule, regulating lymphocyte homoeostasis and activation. To promote viral replication and survival in their hosts, viruses have developed sophisticated mechanisms to combat and avoid immune surveillance. Many of these strategies rely on host defense genes captured during the process of virus-host coevolution. In particular, large DNA viruses devote a wide range of proteins to interfere with almost every host immune pathway. Given that CD229 is critically involved in regulating immune responses, it is not surprising that viruses have designed tactics to mimic or interfere with this receptor. The discovery, in recent years, that some viruses have hijacked CD229 genes from their hosts, incorporating them as an integral part of their genomes, or have evolved proteins to directly target CD229, indicates that this is the case. While it is still an emerging area of research, the present review discusses these viral molecules and their potential in immune modulation. A more detailed understanding of the mechanisms of action and the functional implications of these new viral CD229 mimics may not only provide seminal information on viral immune evasion mechanisms but also, unveil unrecognized aspects of CD229 immune functions.
Collapse
Affiliation(s)
- Ana Angulo
- Immunology Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Barcelona, Spain
| | - Marta Cuenca
- Immunology Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Barcelona, Spain
| | - Pablo Martínez-Vicente
- Immunology Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Barcelona, Spain
| | - Pablo Engel
- Immunology Unit, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques, August Pi i Sunyer, Barcelona, Spain
| |
Collapse
|
12
|
Shissler SC, Webb TJ. The ins and outs of type I iNKT cell development. Mol Immunol 2018; 105:116-130. [PMID: 30502719 DOI: 10.1016/j.molimm.2018.09.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 09/14/2018] [Accepted: 09/29/2018] [Indexed: 01/07/2023]
Abstract
Natural killer T (NKT) cells are innate-like lymphocytes that bridge the gap between the innate and adaptive immune responses. Like innate immune cells, they have a mature, effector phenotype that allows them to rapidly respond to threats, compared to adaptive cells. NKT cells express T cell receptors (TCRs) like conventional T cells, but instead of responding to peptide antigen presented by MHC class I or II, NKT cell TCRs recognize glycolipid antigen in the context of CD1d. NKT cells are subdivided into classes based on their TCR and antigen reactivity. This review will focus on type I iNKT cells that express a semi invariant Vα14Jα18 TCR and respond to the canonical glycolipid antigen, α-galactosylceramide. The innate-like effector functions of these cells combined with their T cell identity make their developmental path quite unique. In addition to the extrinsic factors that affect iNKT cell development such as lipid:CD1d complexes, co-stimulation, and cytokines, this review will provide a comprehensive delineation of the cell intrinsic factors that impact iNKT cell development, differentiation, and effector functions - including TCR rearrangement, survival and metabolism signaling, transcription factor expression, and gene regulation.
Collapse
Affiliation(s)
- Susannah C Shissler
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore St. HSF-1 Room 380, Baltimore, MD 21201, USA.
| | - Tonya J Webb
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore St. HSF-1 Room 380, Baltimore, MD 21201, USA
| |
Collapse
|
13
|
Puñet-Ortiz J, Sáez Moya M, Cuenca M, Caleiras E, Lazaro A, Engel P. Ly9 (CD229) Antibody Targeting Depletes Marginal Zone and Germinal Center B Cells in Lymphoid Tissues and Reduces Salivary Gland Inflammation in a Mouse Model of Sjögren's Syndrome. Front Immunol 2018; 9:2661. [PMID: 30519241 PMCID: PMC6251324 DOI: 10.3389/fimmu.2018.02661] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/29/2018] [Indexed: 01/02/2023] Open
Abstract
Sjögren's Syndrome (SjS) is a common chronic autoimmune disease characterized by the B cell hyperactivation, lymphocyte infiltration, and tissue damage of exocrine glands. It can also present life-threatening extraglandular manifestations, such as pulmonary and hepatic involvement, renal inflammation and marginal zone (MZ) B cell lymphoma. Several biologic agents have been tested in SjS but none has shown significant efficacy. Here, we report the effects of Ly9 (CD229) antibody targeting, a cell surface molecule that belongs to the SLAM family of immunomodulatory receptors, using NOD.H-2h4 mice as a model of SjS-like disease. Female mice were treated with anti-Ly9 antibody or isotype control at week 24, when all mice present SjS related autoantibodies, salivary gland infiltrates, and marginal zone (MZ) B cell pool enlargement. Antibody injection depleted key lymphocyte subsets involved in SjS pathology such as MZ, B1, and germinal center B cells in spleen and draining lymph nodes without inducing a general immunosuppression. Importantly, mice receiving anti-Ly9 mAb showed a reduced lymphocyte infiltrate within salivary glands. This reduction may be, in part, explained by the down-regulation of L-selectin and alfa4/beta7 integrin induced by the anti-Ly9 antibody. Furthermore, levels of anti-nuclear autoantibodies were reduced after anti-Ly9 treatment. These data indicate that Ly9 is a potential therapeutic target for the treatment of SjS.
Collapse
Affiliation(s)
- Joan Puñet-Ortiz
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - Manuel Sáez Moya
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - Marta Cuenca
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - Eduardo Caleiras
- Histopathology Unit, Biotechnology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Adriana Lazaro
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain
| | - Pablo Engel
- Immunology Unit, Department of Biomedical Sciences, Medical School, University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| |
Collapse
|
14
|
Li S, Wong EM, Bui M, Nguyen TL, Joo JHE, Stone J, Dite GS, Dugué PA, Milne RL, Giles GG, Saffery R, Southey MC, Hopper JL. Inference about causation between body mass index and DNA methylation in blood from a twin family study. Int J Obes (Lond) 2018; 43:243-252. [PMID: 29777239 DOI: 10.1038/s41366-018-0103-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 03/19/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Several studies have reported DNA methylation in blood to be associated with body mass index (BMI), but few have investigated causal aspects of the association. We used a twin family design to assess this association at two life points and applied a novel analytical approach to appraise the evidence for causality. METHODS The methylation profile of DNA from peripheral blood was measured for 479 Australian women from 130 twin families. Linear regression was used to estimate the associations of DNA methylation at ~410,000 cytosine-guanine dinucleotides (CpGs), and of the average DNA methylation at ~20,000 genes, with current BMI, BMI at age 18-21 years, and the change between the two (BMI change). A novel regression-based methodology for twins, Inference about Causation through Examination of Familial Confounding (ICE FALCON), was used to assess causation. RESULTS At a 5% false discovery rate, nine, six and 12 CpGs at 24 loci were associated with current BMI, BMI at age 18-21 years and BMI change, respectively. The average DNA methylation of the BHLHE40 and SOCS3 loci was associated with current BMI, and of the PHGDH locus with BMI change. From the ICE FALCON analyses with BMI as the predictor and DNA methylation as the outcome, a woman's DNA methylation level was associated with her co-twin's BMI, and the association disappeared after conditioning on her own BMI, consistent with BMI causing DNA methylation. To the contrary, using DNA methylation as the predictor and BMI as the outcome, a woman's BMI was not associated with her co-twin's DNA methylation level, consistent with DNA methylation not causing BMI. CONCLUSION For middle-aged women, peripheral blood DNA methylation at several genomic locations is associated with current BMI, BMI at age 18-21 years and BMI change. Our study suggests that BMI has a causal effect on peripheral blood DNA methylation.
Collapse
Affiliation(s)
- Shuai Li
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| | - Ee Ming Wong
- Genetic Epidemiology Laboratory, Department of Clinical Pathology, University of Melbourne, Parkville, VIC, Australia.,Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Minh Bui
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| | - Tuong L Nguyen
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| | - Ji-Hoon Eric Joo
- Genetic Epidemiology Laboratory, Department of Clinical Pathology, University of Melbourne, Parkville, VIC, Australia.,Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Jennifer Stone
- Centre for Genetic Origins of Health and Disease, Curtin University and the University of Western Australia, Perth, WA, Australia
| | - Gillian S Dite
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| | - Pierre-Antoine Dugué
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia.,Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Roger L Milne
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia.,Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Graham G Giles
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia.,Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC, Australia.,Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Melissa C Southey
- Genetic Epidemiology Laboratory, Department of Clinical Pathology, University of Melbourne, Parkville, VIC, Australia.,Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
15
|
Cuenca M, Puñet-Ortiz J, Ruart M, Terhorst C, Engel P. Ly9 (SLAMF3) receptor differentially regulates iNKT cell development and activation in mice. Eur J Immunol 2017; 48:99-105. [PMID: 28980301 DOI: 10.1002/eji.201746925] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 08/22/2017] [Accepted: 09/25/2017] [Indexed: 11/11/2022]
Abstract
Invariant natural killer T (iNKT) cells develop into three subsets (NKT1, NKT2, and NKT17) expressing a distinct transcription factor profile, which regulates cytokine secretion upon activation. iNKT cell development in the thymus is modulated by signaling lymphocytic activation molecule family (SLAMF) receptors. In contrast to other SLAMF members, Ly9 (SLAMF3) is a non-redundant negative regulator of iNKT cell development. Here, we show that Ly9 influences iNKT cell lineage differentiation. Ly9-deficient mice on a BALB/c background contained a significantly expanded population of thymic NKT2 cells, while NKT1 cells were nearly absent in BALB/c.Ly9-/- thymus. Conversely, the number of peripheral NKT1 cells in BALB/c.Ly9-/- mice was comparable to that in wild-type mice, indicating that the homeostasis of the different iNKT cell subsets may have distinct requirements depending on their tissue localization. Importantly, Ly9 absence also promoted NKT2 cell differentiation in the NKT1-skewed C57BL/6 background. Furthermore, treatment of wild-type mice with an agonistic monoclonal antibody directed against Ly9 impaired IL-4 and IFN-γ production and reduced by half the number of spleen iNKT cells, with a significant decrease in the proportion of NKT2 cells. Thus, anti-Ly9 targeting could represent a novel therapeutic approach to modulate iNKT cell numbers and activation.
Collapse
Affiliation(s)
- Marta Cuenca
- Immunology Unit, Department of Biomedical Sciences, University of Barcelona Medical School, Barcelona, Spain
| | - Joan Puñet-Ortiz
- Immunology Unit, Department of Biomedical Sciences, University of Barcelona Medical School, Barcelona, Spain
| | - Maria Ruart
- Barcelona Hepatic Hemodynamic Laboratory, Liver Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, University of Barcelona Medical School, Barcelona, Spain
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Pablo Engel
- Immunology Unit, Department of Biomedical Sciences, University of Barcelona Medical School, Barcelona, Spain
| |
Collapse
|
16
|
Waide EH, Tuggle CK, Serão NVL, Schroyen M, Hess A, Rowland RRR, Lunney JK, Plastow G, Dekkers JCM. Genomewide association of piglet responses to infection with one of two porcine reproductive and respiratory syndrome virus isolates. J Anim Sci 2017; 95:16-38. [PMID: 28177360 DOI: 10.2527/jas.2016.0874] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is a devastating disease in the swine industry. Identification of host genetic factors that enable selection for improved performance during PRRS virus (PRRSV) infection would reduce the impact of this disease on animal welfare and production efficiency. We conducted genomewide association study (GWAS) analyses of data from 13 trials of approximately 200 commercial crossbred nursery-age piglets that were experimentally infected with 1 of 2 type 2 isolates of PRRSV (NVSL 97-7985 [NVSL] and KS2006-72109 [KS06]). Phenotypes analyzed were viral load (VL) in blood during the first 21 d after infection (dpi) and weight gain (WG) from 0 to 42 dpi. We accounted for the previously identified QTL in the region on SSC4 in our models to increase power to identify additional regions. Many regions identified by single-SNP analyses were not identified using Bayes-B, but both analyses identified the same regions on SSC3 and SSC5 to be associated with VL in the KS06 trials and on SSC6 in the NVSL trials ( < 5 × 10); for WG, regions on SSC5 and SSC17 were associated in the NVSL trials ( < 3 × 10). No regions were identified with either method for WG in the KS06 trials. Except for the region on SSC4, which was associated with VL for both isolates (but only with WG for NVSL), identified regions did not overlap between the 2 PRRSV isolate data sets, despite high estimates of the genetic correlation between isolates for traits based on these data. We also identified genomic regions whose associations with VL or WG interacted with either PRRSV isolate or with genotype at the SSC4 QTL. Gene ontology (GO) annotation terms for genes located near moderately associated SNP ( < 0.003) were enriched for multiple immunologically (VL) and metabolism- (WG) related GO terms. The biological relevance of these regions suggests that, although it may increase the number of false positives, the use of single-SNP analyses and a relaxed threshold also increased the identification of true positives. In conclusion, although only the SSC4 QTL was associated with response to both PRRSV isolates, genes near associated SNP were enriched for the same GO terms across PRRSV isolates, suggesting that host responses to these 2 isolates are affected by the actions of many genes that function together in similar biological processes.
Collapse
|
17
|
Baglaenko Y, Cruz Tleugabulova M, Gracey E, Talaei N, Manion KP, Chang NH, Ferri DM, Mallevaey T, Wither JE. Invariant NKT Cell Activation Is Potentiated by Homotypic trans-Ly108 Interactions. THE JOURNAL OF IMMUNOLOGY 2017; 198:3949-3962. [PMID: 28373584 DOI: 10.4049/jimmunol.1601369] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 03/07/2017] [Indexed: 01/27/2023]
Abstract
Invariant NKT (iNKT) cells are innate lymphocytes that respond to glycolipids presented by the MHC class Ib molecule CD1d and are rapidly activated to produce large quantities of cytokines and chemokines. iNKT cell development uniquely depends on interactions between double-positive thymocytes that provide key homotypic interactions between signaling lymphocyte activation molecule (SLAM) family members. However, the role of SLAM receptors in the differentiation of iNKT cell effector subsets and activation has not been explored. In this article, we show that C57BL/6 mice containing the New Zealand Black Slam locus have profound alterations in Ly108, CD150, and Ly9 expression that is associated with iNKT cell hyporesponsiveness. This loss of function was only apparent when dendritic cells and iNKT cells had a loss of SLAM receptor expression. Using small interfering RNA knockdowns and peptide-blocking strategies, we demonstrated that trans-Ly108 interactions between dendritic cells and iNKT cells are critical for robust activation. LY108 costimulation similarly increased human iNKT cell activation. Thus, in addition to its established role in iNKT cell ontogeny, Ly108 regulates iNKT cell function in mice and humans.
Collapse
Affiliation(s)
- Yuriy Baglaenko
- Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; and
| | | | - Eric Gracey
- Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; and
| | - Nafiseh Talaei
- Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; and
| | - Kieran Patricia Manion
- Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; and
| | - Nan-Hua Chang
- Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8, Canada
| | - Dario Michael Ferri
- Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; and
| | - Thierry Mallevaey
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; and
| | - Joan E Wither
- Krembil Research Institute, University Health Network, Toronto, Ontario M5T 2S8, Canada; .,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; and.,Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
18
|
Radhakrishnan SV, Bhardwaj N, Luetkens T, Atanackovic D. Novel anti-myeloma immunotherapies targeting the SLAM family of receptors. Oncoimmunology 2017. [PMID: 28638731 DOI: 10.1080/2162402x.2017.1308618] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Treatment for multiple myeloma (MM) has significantly advanced in the last decade with the introduction of proteasome inhibitors and immunomodulatory therapies. Unfortunately, MM continues to cause significant morbidity and most patients eventually succumb to the disease. As in other areas of cancer, immunotherapy in MM has also evolved and holds promise to deliver long-lasting remissions or even cure. The signaling lymphocyte activation molecules (SLAM) family of surface proteins represents a group of potential targets for immunotherapy in MM as some of the family members are expressed consistently on plasma cells and also on myeloma propagating pre-plasma cells. Here, we review the SLAM family members in detail, describe their tissue distribution, biologic pathways, as well as relevant pre-clinical studies and clinical trials in MM. Our review demonstrates the value of SLAM family receptors as potential targets for anti-myeloma immunotherapies and outlines how immunotherapeutic approaches can be developed.
Collapse
Affiliation(s)
- Sabarinath Venniyil Radhakrishnan
- Multiple Myeloma Program & Cancer Immunotherapy, Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Neelam Bhardwaj
- Multiple Myeloma Program & Cancer Immunotherapy, Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Tim Luetkens
- Multiple Myeloma Program & Cancer Immunotherapy, Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Djordje Atanackovic
- Multiple Myeloma Program & Cancer Immunotherapy, Hematology and Hematologic Malignancies, University of Utah/Huntsman Cancer Institute, Salt Lake City, UT, USA
| |
Collapse
|
19
|
Gapin L. Development of invariant natural killer T cells. Curr Opin Immunol 2016; 39:68-74. [PMID: 26802287 DOI: 10.1016/j.coi.2016.01.001] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 12/16/2015] [Accepted: 01/02/2016] [Indexed: 01/09/2023]
Abstract
Invariant natural killer T (iNKT) cells develop into functionally distinct subsets. Each subset expresses a unique combination of transcription factors that regulate cytokine gene transcription upon activation. The tissue distribution and localization within tissues also varies between subsets. Importantly, the relative abundance of the various subsets is directly responsible for altering several immunological parameters, which subsequently affect the immune response. Here, I review recent advances in our understanding of the molecular regulation of iNKT cell subset development.
Collapse
Affiliation(s)
- Laurent Gapin
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical campus and National Jewish Health, Aurora, CO 80045, USA.
| |
Collapse
|
20
|
van Driel BJ, Liao G, Engel P, Terhorst C. Responses to Microbial Challenges by SLAMF Receptors. Front Immunol 2016; 7:4. [PMID: 26834746 PMCID: PMC4718992 DOI: 10.3389/fimmu.2016.00004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/06/2016] [Indexed: 12/24/2022] Open
Abstract
The SLAMF family (SLAMF) of cell surface glycoproteins is comprised of nine glycoproteins and while SLAMF1, 3, 5, 6, 7, 8, and 9 are self-ligand receptors, SLAMF2 and SLAMF4 interact with each other. Their interactions induce signal transduction networks in trans, thereby shaping immune cell-cell communications. Collectively, these receptors modulate a wide range of functions, such as myeloid cell and lymphocyte development, and T and B cell responses to microbes and parasites. In addition, several SLAMF receptors serve as microbial sensors, which either positively or negatively modulate the function of macrophages, dendritic cells, neutrophils, and NK cells in response to microbial challenges. The SLAMF receptor-microbe interactions contribute both to intracellular microbicidal activity as well as to migration of phagocytes to the site of inflammation. In this review, we describe the current knowledge on how the SLAMF receptors and their specific adapters SLAM-associated protein and EAT-2 regulate innate and adaptive immune responses to microbes.
Collapse
Affiliation(s)
- Boaz Job van Driel
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Gongxian Liao
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Pablo Engel
- Immunology Unit, Department of Cell Biology, Immunology and Neurosciences, Medical School, University of Barcelona , Barcelona , Spain
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
21
|
Cuenca M, Romero X, Sintes J, Terhorst C, Engel P. Targeting of Ly9 (CD229) Disrupts Marginal Zone and B1 B Cell Homeostasis and Antibody Responses. THE JOURNAL OF IMMUNOLOGY 2015; 196:726-37. [PMID: 26667173 DOI: 10.4049/jimmunol.1501266] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 11/10/2015] [Indexed: 12/16/2022]
Abstract
Marginal zone (MZ) and B1 B cells have the capacity to respond to foreign Ags more rapidly than conventional B cells, providing early immune responses to blood-borne pathogens. Ly9 (CD229, SLAMF3), a member of the signaling lymphocytic activation molecule family receptors, has been implicated in the development and function of innate T lymphocytes. In this article, we provide evidence that in Ly9-deficient mice splenic transitional 1, MZ, and B1a B cells are markedly expanded, whereas development of B lymphocytes in bone marrow is unaltered. Consistent with an increased number of these B cell subsets, we detected elevated levels of IgG3 natural Abs and a striking increase of T-independent type II Abs after immunization with 2,4,6-trinitrophenyl-Ficoll in the serum of Ly9-deficient mice. The notion that Ly9 could be a negative regulator of innate-like B cell responses was supported by the observation that administering an mAb directed against Ly9 to wild-type mice selectively eliminated splenic MZ B cells and significantly reduced the numbers of B1 and transitional 1 B cells. In addition, Ly9 mAb dramatically diminished in vivo humoral responses and caused a selective downregulation of the CD19/CD21/CD81 complex on B cells and concomitantly an impaired B cell survival and activation in an Fc-independent manner. We conclude that altered signaling caused by the absence of Ly9 or induced by anti-Ly9 may negatively regulate development and function of innate-like B cells by modulating B cell activation thresholds. The results suggest that Ly9 could serve as a novel target for the treatment of B cell-related diseases.
Collapse
Affiliation(s)
- Marta Cuenca
- Immunology Unit, Department of Cell Biology, Immunology and Neurosciences, Medical School, University of Barcelona, Barcelona 08036, Spain; and
| | - Xavier Romero
- Immunology Unit, Department of Cell Biology, Immunology and Neurosciences, Medical School, University of Barcelona, Barcelona 08036, Spain; and
| | - Jordi Sintes
- Immunology Unit, Department of Cell Biology, Immunology and Neurosciences, Medical School, University of Barcelona, Barcelona 08036, Spain; and
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Pablo Engel
- Immunology Unit, Department of Cell Biology, Immunology and Neurosciences, Medical School, University of Barcelona, Barcelona 08036, Spain; and
| |
Collapse
|
22
|
Margraf S, Garner LI, Wilson TJ, Brown MH. A polymorphism in a phosphotyrosine signalling motif of CD229 (Ly9, SLAMF3) alters SH2 domain binding and T-cell activation. Immunology 2015. [PMID: 26221972 PMCID: PMC4610628 DOI: 10.1111/imm.12513] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Signalling lymphocyte activation molecule (SLAM) family members regulate activation and inhibition in the innate and adaptive immune systems. Genome‐wide association studies identified their genetic locus (1q23) as highly polymorphic and associated with susceptibility to systemic lupus erythematosus (SLE). Here we show that the Val602 variant of the non‐synonymous single nucleotide polymorphism (SNP) rs509749 in the SLAM family member CD229 (Ly9, SLAMF3) has a two‐fold lower affinity compared with the SLE‐associated Met602 variant for the small adaptor protein SAP. Comparison of the two variants in T‐cell lines revealed the Val602 variant to be significantly more highly expressed than CD229 Met602. Activation was diminished in cells expressing CD229 Val602 compared with CD229 Met602 as measured by up‐regulation of CD69. There was no correlation between homozygosity at rs509749 and activation in peripheral blood mononuclear cells from healthy donors. These findings identify potential mechanisms by which a single SNP can perturb fine‐tuning in the immune system with significant functional consequences.
Collapse
Affiliation(s)
- Stefanie Margraf
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Lee I Garner
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Timothy J Wilson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Marion H Brown
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
23
|
Abstract
Memory T cells are usually considered to be a feature of a successful immune response against a foreign antigen, and such cells can mediate potent immunity. However, in mice, alternative pathways have been described, through which naïve T cells can acquire the characteristics and functions of memory T cells without encountering specific foreign antigen or the typical signals required for conventional T cell differentiation. Such cells reflect a response to the internal rather the external environment, and hence such cells are called innate memory T cells. In this review, we describe how innate memory subsets were identified, the signals that induce their generation and their functional properties and potential role in the normal immune response. The existence of innate memory T cells in mice raises questions about whether parallel populations exist in humans, and we discuss the evidence for such populations during human T cell development and differentiation.
Collapse
Affiliation(s)
- Stephen C Jameson
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA.
| | - You Jeong Lee
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Kristin A Hogquist
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA.
| |
Collapse
|
24
|
Tsaih SW, Presa M, Khaja S, Ciecko AE, Serreze DV, Chen YG. A locus on mouse chromosome 13 inversely regulates CD1d expression and the development of invariant natural killer T-cells. Genes Immun 2015; 16:221-30. [PMID: 25654212 PMCID: PMC4409484 DOI: 10.1038/gene.2014.81] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/17/2014] [Accepted: 12/18/2014] [Indexed: 12/12/2022]
Abstract
Invariant natural killer T (iNKT)-cell development is controlled by many polymorphic genes present in commonly used mouse inbred strains. Development of type 1 diabetes (T1D) in NOD mice partly results from their production of fewer iNKT-cells compared to non-autoimmune prone control strains including ICR. We previously identified several iNKT-cell quantitative trait genetic loci co-localized with known mouse and human T1D regions in a (NOD × ICR)F2 cross. To further dissect the mechanisms underlying the impaired iNKT-cell compartment in NOD mice, we carried out a series of bone marrow transplantation as well as additional genetic mapping studies. We found that impaired iNKT-cell development in NOD mice was mainly due to the inability of their double-positive (DP) thymocytes to efficiently select this T-cell population. Interestingly, we observed higher levels of CD1d expression by NOD than ICR DP thymocytes. The genetic control of the inverse relationship between the CD1d expression level on DP thymocytes and the frequency of thymic iNKT-cells was further mapped to a region on Chromosome 13 between 60.12 Mb and 70.59 Mb. The NOD allele was found to promote CD1d expression and suppress iNKT-cell development. Our results indicate that genetically controlled physiological variation of CD1d expression levels modulates iNKT-cell development.
Collapse
Affiliation(s)
- S-W Tsaih
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - M Presa
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - S Khaja
- 1] Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI, USA [2] Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - A E Ciecko
- 1] Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI, USA [2] Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Y-G Chen
- 1] Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI, USA [2] Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
25
|
Oghumu S, Terrazas CA, Varikuti S, Kimble J, Vadia S, Yu L, Seveau S, Satoskar AR. CXCR3 expression defines a novel subset of innate CD8+ T cells that enhance immunity against bacterial infection and cancer upon stimulation with IL-15. FASEB J 2014; 29:1019-28. [PMID: 25466888 DOI: 10.1096/fj.14-264507] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Innate CD8(+) T cells are a heterogeneous population with developmental pathways distinct from conventional CD8(+) T cells. However, their biology, classification, and functions remain incompletely understood. We recently demonstrated the existence of a novel population of chemokine (C-X-C motif) receptor 3 (CXCR3)-positive innate CD8(+) T cells. Here, we investigated the functional properties of this subset and identified effector molecules and pathways which mediate their function. Adoptive transfer of IL-15 activated CXCR3(+) innate CD8(+) T cells conferred increased protection against Listeria monocytogenes infection in susceptible IFN-γ(-/-) mice compared with similarly activated CXCR3(-) subset. This was associated with enhanced proliferation and IFN-γ production in CXCR3(+) cells. Further, CXCR3(+) innate cells showed enhanced cytotoxicity against a tumor cell line in vitro. In depth analysis of the CXCR3(+) subset showed increased gene expression of Ccl5, Klrc1, CtsW, GP49a, IL-2Rβ, Atp5e, and Ly6c but reduced IFN-γR2 and Art2b. Ingenuity pathway analysis revealed an up-regulation of genes associated with T-cell activation, proliferation, cytotoxicity, and translational initiation in CXCR3(+) populations. Our results demonstrate that CXCR3 expression in innate CD8(+) T cells defines a subset with enhanced cytotoxic potential and protective antibacterial immune functions. Immunotherapeutic approaches against infectious disease and cancer could utilize CXCR3(+) innate CD8(+) T-cell populations as novel clinical intervention strategies.
Collapse
Affiliation(s)
- Steve Oghumu
- *Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio, USA; Department of Oral Biology, The Ohio State University College of Dentistry, Columbus, Ohio, USA; and Department of Microbiology, Center for Biostatistics, and Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Cesar A Terrazas
- *Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio, USA; Department of Oral Biology, The Ohio State University College of Dentistry, Columbus, Ohio, USA; and Department of Microbiology, Center for Biostatistics, and Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Sanjay Varikuti
- *Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio, USA; Department of Oral Biology, The Ohio State University College of Dentistry, Columbus, Ohio, USA; and Department of Microbiology, Center for Biostatistics, and Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Jennifer Kimble
- *Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio, USA; Department of Oral Biology, The Ohio State University College of Dentistry, Columbus, Ohio, USA; and Department of Microbiology, Center for Biostatistics, and Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Stephen Vadia
- *Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio, USA; Department of Oral Biology, The Ohio State University College of Dentistry, Columbus, Ohio, USA; and Department of Microbiology, Center for Biostatistics, and Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Lianbo Yu
- *Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio, USA; Department of Oral Biology, The Ohio State University College of Dentistry, Columbus, Ohio, USA; and Department of Microbiology, Center for Biostatistics, and Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Stephanie Seveau
- *Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio, USA; Department of Oral Biology, The Ohio State University College of Dentistry, Columbus, Ohio, USA; and Department of Microbiology, Center for Biostatistics, and Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Abhay R Satoskar
- *Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio, USA; Department of Oral Biology, The Ohio State University College of Dentistry, Columbus, Ohio, USA; and Department of Microbiology, Center for Biostatistics, and Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
26
|
De Calisto J, Wang N, Wang G, Yigit B, Engel P, Terhorst C. SAP-Dependent and -Independent Regulation of Innate T Cell Development Involving SLAMF Receptors. Front Immunol 2014; 5:186. [PMID: 24795728 PMCID: PMC4005954 DOI: 10.3389/fimmu.2014.00186] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 04/08/2014] [Indexed: 12/24/2022] Open
Abstract
Signaling lymphocytic activation molecule (SLAM)-associated protein (SAP) plays an essential role in the immune system mediating the function of several members of the SLAM family (SLAMF) of receptors, whose expression is essential for T, NK, and B-cell responses. Additionally, the expression of SAP in double-positive thymocytes is mandatory for natural killer T (NKT) cells and, in mouse, for innate CD8+ T cell development. To date, only two members of the SLAMF of receptors, Slamf1 and Slamf6, have been shown to positively cooperate during NKT cell differentiation in mouse. However, it is less clear whether other members of this family may also participate in the development of these innate T cells. Here, we show that Slamf[1 + 6]−/− and Slamf[1 + 5 + 6]−/−B6 mice have ~70% reduction of NKT cells compared to wild-type B6 mice. Unexpectedly, the proportion of innate CD8+ T cells slightly increased in the Slamf[1 + 5 + 6]−/−, but not in the Slamf[1 + 6]−/− strain, suggesting that Slamf5 may function as a negative regulator of innate CD8+ T cell development. Accordingly, Slamf5−/− B6 mice showed an exclusive expansion of innate CD8+ T cells, but not NKT cells. Interestingly, the SAP-independent Slamf7−/− strain showed an expansion of both splenic innate CD8+ T cells and thymic NKT cells. On the other hand, and similar to what was recently shown in Slamf3−/− BALB/c mice, the proportions of thymic promyelocytic leukemia zinc finger (PLZFhi) NKT cells and innate CD8+ T cells significantly increased in the SAP-independent Slamf8−/− BALB/c strain. In summary, these results show that NKT and innate CD8+ T cell development can be regulated in a SAP-dependent and -independent fashion by SLAMF receptors, in which Slamf1, Slamf6, and Slamf8 affect development of NKT cells, and that Slamf5, Slamf7, and Slamf8 affect the development of innate CD8+ T cells.
Collapse
Affiliation(s)
- Jaime De Calisto
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Ninghai Wang
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Guoxing Wang
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Burcu Yigit
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Pablo Engel
- Immunology Unit, Department of Cell Biology, Immunology and Neurosciences, Medical School , University of Barcelona, Barcelona , Spain
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
27
|
Marcq I, Nyga R, Cartier F, Amrathlal RS, Ossart C, Ouled-Haddou H, Ghamlouch H, Galmiche A, Chatelain D, Lamotte L, Debuysscher V, Fuentes V, Nguyen-Khac E, Regimbeau JM, Marolleau JP, Latour S, Bouhlal H. Identification of SLAMF3 (CD229) as an inhibitor of hepatocellular carcinoma cell proliferation and tumour progression. PLoS One 2013; 8:e82918. [PMID: 24376606 PMCID: PMC3869749 DOI: 10.1371/journal.pone.0082918] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 10/29/2013] [Indexed: 01/26/2023] Open
Abstract
Although hepatocellular carcinoma (HCC) is one of the most common malignancies and constitutes the third leading cause of cancer-related deaths, the underlying molecular mechanisms are not fully understood. In the present study, we demonstrate for the first time that hepatocytes express signalling lymphocytic activation molecule family member 3 (SLAMF3/CD229) but not other SLAMF members. We provide evidence to show that SLAMF3 is involved in the control of hepatocyte proliferation and in hepatocellular carcinogenesis. SLAMF3 expression is significantly lower in primary human HCC samples and HCC cell lines than in human healthy primary hepatocytes. In HCC cell lines, the restoration of high levels of SLAMF3 expression inhibited cell proliferation and migration and enhanced apoptosis. Furthermore, SLAMF3 expression was associated with inhibition of HCC xenograft progression in the nude mouse model. The restoration of SLAMF3 expression levels also decreased the phosphorylation of MAPK ERK1/2, JNK and mTOR. In samples from resected HCC patients, SLAMF3 expression levels were significantly lower in tumorous tissues than in peritumoral tissues. Our results identify SLAMF3 as a specific marker of normal hepatocytes and provide evidence for its potential role in the control of proliferation of HCC cells.
Collapse
Affiliation(s)
- Ingrid Marcq
- INSERM UMR925 and EA 4666 UFR de Médecine, CAP-Santé (FED 4231), Université de Picardie Jules Verne, Amiens, France
| | - Rémy Nyga
- INSERM UMR925 and EA 4666 UFR de Médecine, CAP-Santé (FED 4231), Université de Picardie Jules Verne, Amiens, France
| | - Flora Cartier
- INSERM UMR925 and EA 4666 UFR de Médecine, CAP-Santé (FED 4231), Université de Picardie Jules Verne, Amiens, France
- INSERM U1053, Laboratoire de Physiologie du Cancer du Foie, Université Bordeaux Segalen, 146, rue Léo Saignat, Bordeaux, France
| | - Rabbind Singh Amrathlal
- INSERM UMR925 and EA 4666 UFR de Médecine, CAP-Santé (FED 4231), Université de Picardie Jules Verne, Amiens, France
| | - Christèle Ossart
- Service d’hématologie Clinique et de thérapie cellulaire Centre Hospitalier Universitaire sud, Amiens, France
| | - Hakim Ouled-Haddou
- INSERM UMR925 and EA 4666 UFR de Médecine, CAP-Santé (FED 4231), Université de Picardie Jules Verne, Amiens, France
| | - Hussein Ghamlouch
- INSERM UMR925 and EA 4666 UFR de Médecine, CAP-Santé (FED 4231), Université de Picardie Jules Verne, Amiens, France
| | - Antoine Galmiche
- Service de Biochimie, Centre Hospitalier Universitaire sud, Amiens, France
| | - Denis Chatelain
- Service d’Anatomie Pathologique, Centre Hospitalier Universitaire sud, Amiens, France
| | - Luciane Lamotte
- INSERM UMR925 and EA 4666 UFR de Médecine, CAP-Santé (FED 4231), Université de Picardie Jules Verne, Amiens, France
| | - Véronique Debuysscher
- INSERM UMR925 and EA 4666 UFR de Médecine, CAP-Santé (FED 4231), Université de Picardie Jules Verne, Amiens, France
| | - Vincent Fuentes
- INSERM UMR925 and EA 4666 UFR de Médecine, CAP-Santé (FED 4231), Université de Picardie Jules Verne, Amiens, France
- Service d’Immunologie, Centre Hospitalier Universitaire sud, Amiens, France
| | - Eric Nguyen-Khac
- Service Hepato-Gastroenterologie, Centre Hospitalier Universitaire sud, Amiens, France
| | - Jean-Marc Regimbeau
- Service de chirurgie digestive Centre Hospitalier Universitaire sud, Amiens, France
| | - Jean-Pierre Marolleau
- INSERM UMR925 and EA 4666 UFR de Médecine, CAP-Santé (FED 4231), Université de Picardie Jules Verne, Amiens, France
- Service d’hématologie Clinique et de thérapie cellulaire Centre Hospitalier Universitaire sud, Amiens, France
| | - Sylvain Latour
- IRNEM U768, Hôpital Necker enfants maladies, Paris, France
| | - Hicham Bouhlal
- INSERM UMR925 and EA 4666 UFR de Médecine, CAP-Santé (FED 4231), Université de Picardie Jules Verne, Amiens, France
- Service d’hématologie Clinique et de thérapie cellulaire Centre Hospitalier Universitaire sud, Amiens, France
- * E-mail:
| |
Collapse
|
28
|
Yousefi M, Duplay P. CD28 controls the development of innate-like CD8+ T cells by promoting the functional maturation of NKT cells. Eur J Immunol 2013; 43:3017-27. [PMID: 23896981 DOI: 10.1002/eji.201343627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/20/2013] [Accepted: 07/24/2013] [Indexed: 11/08/2022]
Abstract
NK T cells(NKT cells) share functional characteristics and homing properties that are distinct from conventional T cells. In this study, we investigated the contribution of CD28 in the functional development of γδ NKT and αβ NKT cells in mice. We show that CD28 promotes the thymic maturation of promyelocytic leukemia zinc finger(+) IL-4(+) NKT cells and upregulation of LFA-1 expression on NKT cells. We demonstrate that the developmental defect of γδ NKT cells in CD28-deficient mice is cell autonomous. Moreover, we show in both wild-type C57BL/6 mice and in downstream of tyrosine kinase-1 transgenic mice, a mouse model with increased numbers of γδ NKT cells, that CD28-mediated regulation of thymic IL-4(+) NKT cells promotes the differentiation of eomesodermin(+) CD44(high) innate-like CD8(+) T cells. These findings reveal a previously unappreciated mechanism by which CD28 controls NKT-cell homeostasis and the size of the innate-like CD8(+) T-cell pool.
Collapse
Affiliation(s)
- Mitra Yousefi
- Institut National de la Recherche Scientifique-Institut Armand-Frappier, Université du Québec, Laval, Canada
| | | |
Collapse
|
29
|
de Salort J, Cuenca M, Terhorst C, Engel P, Romero X. Ly9 (CD229) Cell-Surface Receptor is Crucial for the Development of Spontaneous Autoantibody Production to Nuclear Antigens. Front Immunol 2013; 4:225. [PMID: 23914190 PMCID: PMC3728625 DOI: 10.3389/fimmu.2013.00225] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/18/2013] [Indexed: 01/07/2023] Open
Abstract
The Signaling Lymphocyte Activation Molecule Family (SLAMF) genes, which encode cell-surface receptors that modulate innate and adaptive immune responses, lay within a genomic region of human and mouse chromosome 1 that confers a predisposition for the development of systemic lupus erythematosus (SLE). Herein, we demonstrate that the SLAMF member Ly9 arises as a novel receptor contributing to the reinforcement of tolerance. Specifically, Ly9-deficient mice spontaneously developed features of systemic autoimmunity such as the production of anti-nuclear antibodies (ANA), -dsDNA, and -nucleosome autoantibodies, independently of genetic background [(B6.129) or (BALB/c.129)]. In aged (10- to 12-month-old) Ly9−/− mice key cell subsets implicated in autoimmunity were expanded, e.g., T follicular helper (Tfh) as well as germinal center (GC) B cells. More importantly, in vitro functional experiments showed that Ly9 acts as an inhibitory receptor of IFN-γ producing CD4+ T cells. Taken together, our findings reveal that the Ly9 receptor triggers cell intrinsic safeguarding mechanisms to prevent a breach of tolerance, emerging as a new non-redundant inhibitory cell-surface receptor capable of disabling autoantibody responses.
Collapse
Affiliation(s)
- Jose de Salort
- Immunology Unit, Department of Cell Biology, Immunology and Neurosciences, Medical School, University of Barcelona , Barcelona , Spain
| | | | | | | | | |
Collapse
|