1
|
Song Y, Zhang Y, Wang Z, Lin Y, Cao X, Han X, Li G, Hou A, Han S. CCL2 mediated IKZF1 expression promotes M2 polarization of glioma-associated macrophages through CD84-SHP2 pathway. Oncogene 2024; 43:2737-2749. [PMID: 39112517 DOI: 10.1038/s41388-024-03118-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024]
Abstract
The proneural-mesenchymal (PN-MES) transformation of glioma stem cells (GSCs) can significantly increase proliferation, invasion, chemotherapy tolerance, and recurrence. M2-like polarization of tumor-associated macrophages (TAMs) has a strong immunosuppressive effect, promoting tumor malignancy and angiogenesis. There is limited understanding on the interactions between GSCs and TAMs as well as their associated molecular mechanisms. In the present study, bioinformatics analysis, GSC and TAM co-culture, determination of TAM polarization phenotypes, and other in vitro experiments confirmed that CCL2 secreted by MES-GSCs promotes TAM-M2 polarization via the IKZF1-CD84-SHP2 pathway and PN-MES transformation of GSCs via the IKZF1-LRG1 pathway in TAMs. IKZF1 inhibitors could significantly reduce tumor volumes in animal glioma models and improve survival, as well as suppress TAM-M2 polarization and the GSC malignant phenotype. The results of this study indicate the important interaction between TAMs and GSCs in the glioma microenvironment as well as its role in tumor progression. The findings also suggest a novel target for follow-up clinical transformation research on the regulation of TAM function and GSCs malignant phenotype.
Collapse
Affiliation(s)
- Yifu Song
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yaochuan Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Zixun Wang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yibin Lin
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xu Cao
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaodi Han
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Guangyu Li
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Ana Hou
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Sheng Han
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
2
|
Kľoc D, Kurhajec S, Huniadi M, Sýkora J, Guman T, Šarišský M. SLAM Family Receptors in B Cell Chronic Lymphoproliferative Disorders. Int J Mol Sci 2024; 25:4014. [PMID: 38612827 PMCID: PMC11012012 DOI: 10.3390/ijms25074014] [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: 02/20/2024] [Revised: 03/28/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
The signaling lymphocytic activation molecule (SLAM) receptor family (SLAMF) consists of nine glycoproteins that belong to the CD2 superfamily of immunoglobulin (Ig) domain-containing molecules. SLAMF receptors modulate the differentiation and activation of a wide range of immune cells. Individual SLAMF receptors are expressed on the surface of hematopoietic stem cells, hematopoietic progenitor cells, B cells, T cells, NK cells, NKT cells, monocytes, macrophages, dendritic cells, neutrophils, and platelets. The expression of SLAMF receptors was studied during normal B cell maturation. Several SLAMF receptors were also detected in cancer cell lines of B-lymphoid origin and in pathological B cells from patients with B cell chronic lymphoproliferative disorders (B-CLPD), the most frequent hematological malignancies in adults. This review summarizes current knowledge on the expression of SLAMF receptors and their adaptor proteins SAP and EAT-2 in B-CLPD. Several SLAMF receptors could be regarded as potential diagnostic and differential diagnostic markers, prognostic factors, and targets for the development of novel drugs for patients with B-CLPD.
Collapse
Affiliation(s)
- Dominik Kľoc
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (D.K.); (M.H.)
| | - Slavomír Kurhajec
- Department of Pharmaceutical Technology, Pharmacognosy, and Botany, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia;
| | - Mykhailo Huniadi
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (D.K.); (M.H.)
| | - Ján Sýkora
- Department of Haematology and Oncohaematology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice and Louis Pasteur University Hospital Košice, Trieda SNP 1, 04011 Košice, Slovakia; (J.S.); (T.G.)
| | - Tomáš Guman
- Department of Haematology and Oncohaematology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice and Louis Pasteur University Hospital Košice, Trieda SNP 1, 04011 Košice, Slovakia; (J.S.); (T.G.)
| | - Marek Šarišský
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (D.K.); (M.H.)
| |
Collapse
|
3
|
Farhangnia P, Ghomi SM, Mollazadehghomi S, Nickho H, Akbarpour M, Delbandi AA. SLAM-family receptors come of age as a potential molecular target in cancer immunotherapy. Front Immunol 2023; 14:1174138. [PMID: 37251372 PMCID: PMC10213746 DOI: 10.3389/fimmu.2023.1174138] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
The signaling lymphocytic activation molecule (SLAM) family receptors were discovered in immune cells for the first time. The SLAM-family receptors are a significant player in cytotoxicity, humoral immune responses, autoimmune diseases, lymphocyte development, cell survival, and cell adhesion. There is growing evidence that SLAM-family receptors have been involved in cancer progression and heralded as a novel immune checkpoint on T cells. Previous studies have reported the role of SLAMs in tumor immunity in various cancers, including chronic lymphocytic leukemia, lymphoma, multiple myeloma, acute myeloid leukemia, hepatocellular carcinoma, head and neck squamous cell carcinoma, pancreas, lung, and melanoma. Evidence has deciphered that the SLAM-family receptors may be targeted for cancer immunotherapy. However, our understanding in this regard is not complete. This review will discuss the role of SLAM-family receptors in cancer immunotherapy. It will also provide an update on recent advances in SLAM-based targeted immunotherapies.
Collapse
Affiliation(s)
- Pooya Farhangnia
- Immunology Research Center, Institute of Immunology and Infectious Disease, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Shamim Mollazadeh Ghomi
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Shabnam Mollazadehghomi
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hamid Nickho
- Immunology Research Center, Institute of Immunology and Infectious Disease, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahzad Akbarpour
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Advanced Cellular Therapeutics Facility (ACTF), Hematopoietic Cellular Therapy Program, Section of Hematology & Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, United States
| | - Ali-Akbar Delbandi
- Immunology Research Center, Institute of Immunology and Infectious Disease, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Romano A, Storti P, Marchica V, Scandura G, Notarfranchi L, Craviotto L, Di Raimondo F, Giuliani N. Mechanisms of Action of the New Antibodies in Use in Multiple Myeloma. Front Oncol 2021; 11:684561. [PMID: 34307150 PMCID: PMC8297441 DOI: 10.3389/fonc.2021.684561] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/09/2021] [Indexed: 12/19/2022] Open
Abstract
Monoclonal antibodies (mAbs) directed against antigen-specific of multiple myeloma (MM) cells have Fc-dependent immune effector mechanisms, such as complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC), and antibody-dependent cellular phagocytosis (ADCP), but the choice of the antigen is crucial for the development of effective immuno-therapy in MM. Recently new immunotherapeutic options in MM patients have been developed against different myeloma-related antigens as drug conjugate-antibody, bispecific T-cell engagers (BiTEs) and chimeric antigen receptor (CAR)-T cells. In this review, we will highlight the mechanism of action of immuno-therapy currently available in clinical practice to target CD38, SLAMF7, and BCMA, focusing on the biological role of the targets and on mechanisms of actions of the different immunotherapeutic approaches underlying their advantages and disadvantages with critical review of the literature data.
Collapse
Affiliation(s)
- Alessandra Romano
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
| | - Paola Storti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Grazia Scandura
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
| | | | - Luisa Craviotto
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Francesco Di Raimondo
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
- U.O.C. Ematologia, A.O.U. Policlinico–San Marco, Catania, Italy
| | | |
Collapse
|
5
|
Diacylglycerol Kinase alpha in X Linked Lymphoproliferative Disease Type 1. Int J Mol Sci 2021; 22:ijms22115816. [PMID: 34072296 PMCID: PMC8198409 DOI: 10.3390/ijms22115816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/31/2022] Open
Abstract
Diacylglycerol kinases are intracellular enzymes that control the balance between the secondary messengers diacylglycerol and phosphatidic acid. DGKα and DGKζ are the prominent isoforms that restrain the intensity of T cell receptor signalling by metabolizing PLCγ generated diacylglycerol. Thus, their activity must be tightly controlled to grant cellular homeostasis and refine immune responses. DGKα is specifically inhibited by strong T cell activating signals to allow for full diacylglycerol signalling which mediates T cell response. In X-linked lymphoproliferative disease 1, deficiency of the adaptor protein SAP results in altered T cell receptor signalling, due in part to persistent DGKα activity. This activity constrains diacylglycerol levels, attenuating downstream pathways such as PKCθ and Ras/MAPK and decreasing T cell restimulation induced cell death. This is a form of apoptosis triggered by prolonged T cell activation that is indeed defective in CD8+ cells of X-linked lymphoproliferative disease type 1 patients. Accordingly, inhibition or downregulation of DGKα activity restores in vitro a correct diacylglycerol dependent signal transduction, cytokines production and restimulation induced apoptosis. In animal disease models, DGKα inhibitors limit CD8+ expansion and immune-mediated tissue damage, suggesting the possibility of using inhibitors of diacylglycerol kinase as a new therapeutic approach.
Collapse
|
6
|
Research Progress on NK Cell Receptors and Their Signaling Pathways. Mediators Inflamm 2020; 2020:6437057. [PMID: 32774149 PMCID: PMC7396059 DOI: 10.1155/2020/6437057] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/25/2020] [Accepted: 06/20/2020] [Indexed: 12/17/2022] Open
Abstract
Natural killer cells (NK cells) play an important role in innate immunity. NK cells recognize self and nonself depending on the balance of activating receptors and inhibitory receptors. After binding to their ligands, NK cell receptors trigger subsequent signaling conduction and then determine whether NK is activated or inhibited. Furthermore, NK cell response includes cytotoxicity and cytokine release, which is tightly related to the activation of NK cell-activating receptors and the inhibition of inhibitory receptors on the surfaces of NK cells. The expression and function of NK cell surface receptors also alter in virus infection, tumor, and autoimmune diseases and influence the occurrence and development of diseases. So, it is important to understand the mechanism of recognition between NK receptors and their ligands in pathological conditions and the signaling pathways of NK cell receptors. This review mainly summarizes the research progress on NK cell surface receptors and their signal pathways.
Collapse
|
7
|
Wilson TJ, Clare S, Mikulin J, Johnson CM, Harcourt K, Lyons PA, Dougan G, Smith KGC. Signalling lymphocyte activation molecule family member 9 is found on select subsets of antigen-presenting cells and promotes resistance to Salmonella infection. Immunology 2020; 159:393-403. [PMID: 31880316 PMCID: PMC7078004 DOI: 10.1111/imm.13169] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/16/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
Signalling lymphocyte activation molecule family member 9 (SLAMF9) is an orphan receptor of the CD2/SLAM family of leucocyte surface proteins. Examination of SLAMF9 expression and function indicates that SLAMF9 promotes inflammation by specialized subsets of antigen‐presenting cells. Within healthy liver and circulating mouse peripheral blood mononuclear cells, SLAMF9 is expressed on CD11b+, Ly6C−, CD11clow, F4/80low, MHC‐II+, CX3CR1+ mononuclear phagocytes as well as plasmacytoid dendritic cells. In addition, SLAMF9 can be found on peritoneal B1 cells and small (F4/80low), but not large (F4/80high), peritoneal macrophages. Upon systemic challenge with Salmonella enterica Typhimurium, Slamf9−/− mice were impaired in their ability to clear the infection from the liver. In humans, SLAMF9 is up‐regulated upon differentiation of monocytes into macrophages, and lipopolysaccharide stimulation of PMA‐differentiated, SLAMF9 knockdown THP‐1 cells showed an essential role of SLAMF9 in production of granulocyte–macrophage colony‐stimulating factor, tumour necrosis factor‐α, and interleukin‐1β. Taken together, these data implicate SLAMF9 in the initiation of inflammation and clearance of bacterial infection.
Collapse
Affiliation(s)
- Timothy J Wilson
- Department of Microbiology, Miami University, Oxford, OH, USA.,Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Simon Clare
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Joseph Mikulin
- Department of Microbiology, Miami University, Oxford, OH, USA
| | | | | | - Paul A Lyons
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.,Cambridge Institute for Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Gordon Dougan
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.,Cambridge Institute for Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kenneth G C Smith
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.,Cambridge Institute for Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
Campbell KS, Cohen AD, Pazina T. Mechanisms of NK Cell Activation and Clinical Activity of the Therapeutic SLAMF7 Antibody, Elotuzumab in Multiple Myeloma. Front Immunol 2018; 9:2551. [PMID: 30455698 PMCID: PMC6230619 DOI: 10.3389/fimmu.2018.02551] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/17/2018] [Indexed: 12/28/2022] Open
Abstract
Multiple myeloma (MM) is a bone marrow plasma cell neoplasm and is the second most-common hematologic malignancy. Despite advances in therapy, MM remains largely incurable. Elotuzumab is a humanized IgG1 monoclonal antibody targeting SLAMF7, which is highly expressed on myeloma cells, and the antibody is approved for the treatment of relapsed and/or refractory (RR) MM in combination with lenalidomide and dexamethasone. Elotuzumab can stimulate robust antibody-dependent cellular cytotoxicity (ADCC) through engaging with FcγRIIIA (CD16) on NK cells and antibody-dependent cellular phagocytosis (ADCP) by macrophages. Interestingly, SLAMF7 is also expressed on cytolytic NK cells, which also express the requisite adaptor protein, EAT-2, to mediate activation signaling. Accumulating evidence indicates that antibody crosslinking of SLAMF7 on human and mouse NK cells can stimulate EAT-2-dependent activation of PLCγ, ERK, and intracellular calcium mobilization. The binding of SLAMF7 by elotuzumab can directly induce signal transduction in human NK cells, including co-stimulation of the calcium signaling triggered through other surface receptors, such as NKp46 and NKG2D. In RRMM patients, elotuzumab monotherapy did not produce objective responses, but did enhance the activity of approved standard of care therapies, including lenalidomide or bortezomib, which are known to enhance anti-tumor responses by NK cells. Taken together, these preclinical results and accumulating experience in the clinic provide compelling evidence that the mechanism of action of elotuzumab in MM patients involves the activation of NK cells through both CD16-mediated ADCC and direct co-stimulation via engagement with SLAMF7, as well as promoting ADCP by macrophages. We review the current understanding of how elotuzumab utilizes multiple mechanisms to facilitate immune-mediated attack of myeloma cells, as well as outline goals for future research.
Collapse
Affiliation(s)
- Kerry S Campbell
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Adam D Cohen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Tatiana Pazina
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, United States.,FSBSI "Institute of Experimental Medicine", St. Petersburg, Russia
| |
Collapse
|
9
|
Dragovich MA, Mor A. The SLAM family receptors: Potential therapeutic targets for inflammatory and autoimmune diseases. Autoimmun Rev 2018; 17:674-682. [PMID: 29729453 DOI: 10.1016/j.autrev.2018.01.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 01/18/2018] [Indexed: 12/20/2022]
Abstract
The signaling lymphocytic activation molecule (SLAM) family is comprised of nine distinct receptors (SLAMF1 through SLAMF9) that are expressed on hematopoietic cells. All of these receptors, with the exception of SLAMF4, are homotypic by nature as downstream signaling occurs when hematopoietic cells that express the same SLAM receptor interact. The SLAM family receptor function is largely controlled via SLAM associated protein (SAP) family adaptors. The SAP family adaptors consist of SAP, Ewing sarcoma associated transcript (EAT)-2, and EAT-2-related transducer (ERT). These adaptors associate with the cytoplasmic domain of the SLAM family receptors through phosphorylated tyrosines. Defects in SLAM family members and SAP adaptors have been implicated in causing immune deficiencies. This is exemplified in patients with X-linked lymphoproliferative (XLP) disease, where SAP undergoes a loss of function mutation. Furthermore, evidence has been accumulating that SLAM family members are potential targets for inflammatory and autoimmune diseases. This review will discuss the structure and function of the SLAM family receptors and SAP family adaptors, their role in immune regulation, and potential approaches to target this family of receptors therapeutically.
Collapse
Affiliation(s)
- Matthew A Dragovich
- Department of Medicine, Division of Rheumatology, NYU School of Medicine, New York, NY 10016, USA; Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| | - Adam Mor
- Department of Medicine, Division of Rheumatology, NYU School of Medicine, New York, NY 10016, USA; Perlmutter Cancer Center, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
10
|
Elotuzumab for the Treatment of Relapsed or Refractory Multiple Myeloma, with Special Reference to its Modes of Action and SLAMF7 Signaling. Mediterr J Hematol Infect Dis 2018. [PMID: 29531651 PMCID: PMC5841936 DOI: 10.4084/mjhid.2018.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Elotuzumab, targeting signaling lymphocytic activation molecule family 7 (SLAMF7), has been approved in combination with lenalidomide and dexamethasone (ELd) for relapsed/refractory multiple myeloma (MM) based on the findings of the phase III randomized trial ELOQUENT-2 (NCT01239797). Four-year follow-up analyses of ELOQUENT-2 have demonstrated that progression-free survival was 21% in ELd versus 14% in Ld. Elotuzumab binds a unique epitope on the membrane IgC2 domain of SLAMF7, exhibiting a dual mechanism of action: natural killer (NK) cell-mediated antibody-dependent cellular cytotoxicity (ADCC) and enhancement of NK cell activity. The ADCC is mediated through engagement between Fc portion of elotuzumab and FcgRIIIa/CD16 on NK cells. Enhanced NK cell cytotoxicity results from phosphorylation of the immunoreceptor tyrosine-based switch motif (ITSM) that is induced via elotuzumab binding and recruits the SLAM-associated adaptor protein EAT-2. The coupling of EAT-2 to the phospholipase Cg enzymes SH2 domain leads to enhanced Ca2+ influx and MAPK/Erk pathway activation, resulting in granule polarization and enhanced exocytosis in NK cells. Elotuzumab does not stimulate the proliferation of MM cells due to a lack of EAT-2. The inhibitory effects of elotuzumab on MM cell growth are not induced by the lack of CD45, even though SHP-2, SHP-1, SHIP-1, and Csk may be recruited to phosphorylated ITSM of SLAMF7. ELd improves PFS in patients with high-risk cytogenetics, i.e. t(4;14), del(17p), and 1q21 gain/amplification. Since the immune state is paralytic in advanced MM, the efficacy of ELd with minimal toxicity may bring forward for consideration of its use in the early stages of the disease.
Collapse
|
11
|
Jadwin JA, Curran TG, Lafontaine AT, White FM, Mayer BJ. Src homology 2 domains enhance tyrosine phosphorylation in vivo by protecting binding sites in their target proteins from dephosphorylation. J Biol Chem 2017; 293:623-637. [PMID: 29162725 DOI: 10.1074/jbc.m117.794412] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 11/17/2017] [Indexed: 02/03/2023] Open
Abstract
Phosphotyrosine (pTyr)-dependent signaling is critical for many cellular processes. It is highly dynamic, as signal output depends not only on phosphorylation and dephosphorylation rates but also on the rates of binding and dissociation of effectors containing phosphotyrosine-dependent binding modules such as Src homology 2 (SH2) and phosphotyrosine-binding (PTB) domains. Previous in vitro studies suggested that binding of SH2 and PTB domains can enhance protein phosphorylation by protecting the sites bound by these domains from phosphatase-mediated dephosphorylation. To test whether this occurs in vivo, we used the binding of growth factor receptor bound 2 (GRB2) to phosphorylated epidermal growth factor receptor (EGFR) as a model system. We analyzed the effects of SH2 domain overexpression on protein tyrosine phosphorylation by quantitative Western and far-Western blotting, mass spectrometry, and computational modeling. We found that SH2 overexpression results in a significant, dose-dependent increase in EGFR tyrosine phosphorylation, particularly of sites corresponding to the binding specificity of the overexpressed SH2 domain. Computational models using experimentally determined EGFR phosphorylation and dephosphorylation rates, and pTyr-EGFR and GRB2 concentrations, recapitulated the experimental findings. Surprisingly, both modeling and biochemical analyses suggested that SH2 domain overexpression does not result in a major decrease in the number of unbound phosphorylated SH2 domain-binding sites. Our results suggest that signaling via SH2 domain binding is buffered over a relatively wide range of effector concentrations and that SH2 domain proteins with overlapping binding specificities are unlikely to compete with one another for phosphosites in vivo.
Collapse
Affiliation(s)
- Joshua A Jadwin
- From the Raymond and Beverly Sackler Laboratory of Molecular Medicine, Department of Genetics and Genome Sciences, and the Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, Connecticut 06030 and
| | - Timothy G Curran
- the Department of Biological Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Adam T Lafontaine
- From the Raymond and Beverly Sackler Laboratory of Molecular Medicine, Department of Genetics and Genome Sciences, and the Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, Connecticut 06030 and
| | - Forest M White
- the Department of Biological Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Bruce J Mayer
- From the Raymond and Beverly Sackler Laboratory of Molecular Medicine, Department of Genetics and Genome Sciences, and the Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, Connecticut 06030 and
| |
Collapse
|
12
|
T Cell Costimulation by CD6 Is Dependent on Bivalent Binding of a GADS/SLP-76 Complex. Mol Cell Biol 2017; 37:MCB.00071-17. [PMID: 28289074 PMCID: PMC5440646 DOI: 10.1128/mcb.00071-17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/01/2017] [Indexed: 12/22/2022] Open
Abstract
The cell surface receptor CD6 regulates T cell activation in both activating and inhibitory manners. The adaptor protein SLP-76 is recruited to the phosphorylated CD6 cytoplasmic Y662 residue during T cell activation, providing an activating signal to T cells. In this study, a biochemical approach identified the SH2 domain-containing adaptor protein GADS as the dominant interaction partner for the CD6 cytoplasmic Y629 residue. Functional experiments in human Jurkat and primary T cells showed that both mutations Y629F and Y662F abolished costimulation by CD6. In addition, a restraint on T cell activation by CD6 was revealed in primary T cells expressing CD6 mutated at Y629 and Y662. These data are consistent with a model in which bivalent recruitment of a GADS/SLP-76 complex is required for costimulation by CD6.
Collapse
|
13
|
Shi T, Niepel M, McDermott JE, Gao Y, Nicora CD, Chrisler WB, Markillie LM, Petyuk VA, Smith RD, Rodland KD, Sorger PK, Qian WJ, Wiley HS. Conservation of protein abundance patterns reveals the regulatory architecture of the EGFR-MAPK pathway. Sci Signal 2016; 9:rs6. [PMID: 27405981 DOI: 10.1126/scisignal.aaf0891] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Various genetic mutations associated with cancer are known to alter cell signaling, but it is not clear whether they dysregulate signaling pathways by altering the abundance of pathway proteins. Using a combination of RNA sequencing and ultrasensitive targeted proteomics, we defined the primary components-16 core proteins and 10 feedback regulators-of the epidermal growth factor receptor (EGFR)-mitogen-activated protein kinase (MAPK) pathway in normal human mammary epithelial cells and then quantified their absolute abundance across a panel of normal and breast cancer cell lines as well as fibroblasts. We found that core pathway proteins were present at very similar concentrations across all cell types, with a variance similar to that of proteins previously shown to display conserved abundances across species. In contrast, EGFR and transcriptionally controlled feedback regulators were present at highly variable concentrations. The absolute abundance of most core proteins was between 50,000 and 70,000 copies per cell, but the adaptors SOS1, SOS2, and GAB1 were found at far lower amounts (2000 to 5000 copies per cell). MAPK signaling showed saturation in all cells between 3000 and 10,000 occupied EGFRs, consistent with the idea that adaptors limit signaling. Our results suggest that the relative stoichiometry of core MAPK pathway proteins is very similar across different cell types, with cell-specific differences mostly restricted to variable amounts of feedback regulators and receptors. The low abundance of adaptors relative to EGFR could be responsible for previous observations that only a fraction of total cell surface EGFR is capable of rapid endocytosis, high-affinity binding, and mitogenic signaling.
Collapse
Affiliation(s)
- Tujin Shi
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Mario Niepel
- HMS LINCS Center and Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Jason E McDermott
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Yuqian Gao
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Carrie D Nicora
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - William B Chrisler
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Lye M Markillie
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99352 USA
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA. Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99352 USA
| | - Karin D Rodland
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Peter K Sorger
- HMS LINCS Center and Laboratory of Systems Pharmacology, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - H Steven Wiley
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99352 USA.
| |
Collapse
|
14
|
Chaudhary A, Leite M, Kulasekara BR, Altura MA, Ogahara C, Weiss E, Fu W, Blanc MP, O'Keeffe M, Terhorst C, Akey JM, Miller SI. Human Diversity in a Cell Surface Receptor that Inhibits Autophagy. Curr Biol 2016; 26:1791-801. [PMID: 27345162 DOI: 10.1016/j.cub.2016.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/27/2016] [Accepted: 05/03/2016] [Indexed: 02/01/2023]
Abstract
Mutations in genes encoding autophagy proteins have been associated with human autoimmune diseases, suggesting that diversity in autophagy responses could be associated with disease susceptibility or severity. A cellular genome-wide association study (GWAS) screen was performed to explore normal human diversity in responses to rapamycin, a microbial product that induces autophagy. Cells from several human populations demonstrated variability in expression of a cell surface receptor, CD244 (SlamF4, 2B4), that correlated with changes in rapamycin-induced autophagy. High expression of CD244 and receptor activation with its endogenous ligand CD48 inhibited starvation- and rapamycin-induced autophagy by promoting association of CD244 with the autophagy complex proteins Vps34 and Beclin-1. The association of CD244 with this complex reduced Vps34 lipid kinase activity. Lack of CD244 is associated with auto-antibody production in mice, and lower expression of human CD244 has previously been implicated in severity of human rheumatoid arthritis and systemic lupus erythematosus, indicating that increased autophagy as a result of low levels of CD244 may alter disease outcomes.
Collapse
Affiliation(s)
- Anu Chaudhary
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Mara Leite
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | | | - Melissa A Altura
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Cassandra Ogahara
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Eli Weiss
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Wenqing Fu
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Marie-Pierre Blanc
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Michael O'Keeffe
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Joshua M Akey
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Samuel I Miller
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA; Department of Immunology, University of Washington, Seattle, WA 98195, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
15
|
Wu N, Veillette A. SLAM family receptors in normal immunity and immune pathologies. Curr Opin Immunol 2015; 38:45-51. [PMID: 26682762 DOI: 10.1016/j.coi.2015.11.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/13/2015] [Accepted: 11/16/2015] [Indexed: 12/23/2022]
Abstract
The signaling lymphocytic activation molecule (SLAM) family is a group of six receptors restricted to hematopoietic cells. Most of these receptors are self-ligands, and thus are triggered in the context of interactions between hematopoietic cells. By way of their cytoplasmic domain, SLAM-related receptors associate with the SLAM-associated protein (SAP) family of adaptors, which control the signals and functions of SLAM family receptors. Recent findings have provided new insights into the key roles of SLAM family receptors in normal immunity, their involvement in human diseases and their usefulness as drug targets to treat human malignancies. These data are reviewed herein.
Collapse
Affiliation(s)
- Ning Wu
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada H2W 1R7.
| | - André Veillette
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada H2W 1R7; Department of Medicine, University of Montréal, Montréal, Québec, Canada H3T 1J4; Department of Medicine, McGill University, Montréal, Québec, Canada H3G 1Y6.
| |
Collapse
|
16
|
Muccio VE, Saraci E, Gilestro M, Gattei V, Zucchetto A, Astolfi M, Ruggeri M, Marzanati E, Passera R, Palumbo A, Boccadoro M, Omedè P. Multiple myeloma: New surface antigens for the characterization of plasma cells in the era of novel agents. CYTOMETRY PART B-CLINICAL CYTOMETRY 2015; 90:81-90. [DOI: 10.1002/cyto.b.21279] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 07/17/2015] [Accepted: 07/29/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Vittorio Emanuele Muccio
- Divisione Universitaria Di Ematologia; AOU Città Della Salute E Della Scienza Di Torino, Torino, Italy
| | - Elona Saraci
- Divisione Universitaria Di Ematologia; AOU Città Della Salute E Della Scienza Di Torino, Torino, Italy
| | - Milena Gilestro
- Divisione Universitaria Di Ematologia; AOU Città Della Salute E Della Scienza Di Torino, Torino, Italy
| | - Valter Gattei
- Clinical and Experimental Onco-Hematology Unit; Centro Di Riferimento Oncologico I.R.C.C.S, Torino, Italy
| | - Antonella Zucchetto
- Clinical and Experimental Onco-Hematology Unit; Centro Di Riferimento Oncologico I.R.C.C.S, Torino, Italy
| | - Monica Astolfi
- Divisione Universitaria Di Ematologia; AOU Città Della Salute E Della Scienza Di Torino, Torino, Italy
| | - Marina Ruggeri
- Divisione Universitaria Di Ematologia; AOU Città Della Salute E Della Scienza Di Torino, Torino, Italy
| | - Eleonora Marzanati
- Divisione Universitaria Di Ematologia; AOU Città Della Salute E Della Scienza Di Torino, Torino, Italy
| | - Roberto Passera
- AOU Città Della Salute E Della Scienza Di Torino, Divisione Di Medicina Nucleare; Università Di Torino, Torino, Italy
| | - Antonio Palumbo
- Divisione Universitaria Di Ematologia; AOU Città Della Salute E Della Scienza Di Torino, Torino, Italy
| | - Mario Boccadoro
- Divisione Universitaria Di Ematologia; AOU Città Della Salute E Della Scienza Di Torino, Torino, Italy
| | - Paola Omedè
- Divisione Universitaria Di Ematologia; AOU Città Della Salute E Della Scienza Di Torino, Torino, Italy
| |
Collapse
|
17
|
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
|
18
|
Kim JR, Mathew SO, Mathew PA. Blimp-1/PRDM1 regulates the transcription of human CS1 (SLAMF7) gene in NK and B cells. Immunobiology 2015; 221:31-9. [PMID: 26310579 DOI: 10.1016/j.imbio.2015.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 07/30/2015] [Accepted: 08/11/2015] [Indexed: 10/23/2022]
Abstract
CS1 (CRACC/CD319/SLAMF7) is a member of SLAM (Signaling Lymphocyte Activation Molecule) family receptors and is expressed on NK cells, a subset of CD8(+) T lymphocytes, activated monocytes, mature dendritic cells and activated B cells. In NK cells, CS1 signaling induces cytolytic function of NK cells against targets whereas in B cells CS1 induces proliferation and autocrine cytokine production. CS1 is upregulated in multiple myeloma cells and contributes to clonogenic growth and tumorigenicity. However, the mechanism of CS1 upregulation is unknown. In this study, we analyzed the transcriptional regulation of human CS1 gene in NK and B cells. The promoter region of CS1 contains a Blimp-1/PRDM1 binding site and relative luciferase activities of successive deletion mutants of CS1 promoter were different between Blimp-1/PRDM1-positive and Blimp-1/PRDM1-negative cells. Proximal region of CS1 promoter contains a CAAT box and atypical TATA-box that might result in common transcription initiation at -29 nucleotides upstream of the ATG translation start codon. Electrophoretic Mobility Shift Assay (EMSA) and Chromatin Immunoprecipitation (ChIP) assays revealed Blimp-1/PRDM1 binds to the CS1 promoter region. Mutating the Blimp-1/PRDM1 site at -750 to -746 decreased the transcriptional activity of CS1 promoter implicating a trans-activating function of Blimp-1/PRDM1 in human CS1 gene regulation. The finding that Blimp-1/PRDM1 enhances transcription of CS1 gene in multiple myeloma cells may help in developing novel strategies for therapeutic intervention in multiple myeloma.
Collapse
Affiliation(s)
- Jong R Kim
- Department of Cell Biology and Immunology and Institute for Cancer Research, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Stephen O Mathew
- Department of Cell Biology and Immunology and Institute for Cancer Research, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Porunelloor A Mathew
- Department of Cell Biology and Immunology and Institute for Cancer Research, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA.
| |
Collapse
|
19
|
O’Keeffe MS, Song JH, Liao G, De Calisto J, Halibozek PJ, Mora JR, Bhan AK, Wang N, Reinecker HC, Terhorst C. SLAMF4 Is a Negative Regulator of Expansion of Cytotoxic Intraepithelial CD8+ T Cells That Maintains Homeostasis in the Small Intestine. Gastroenterology 2015; 148:991-1001.e4. [PMID: 25678452 PMCID: PMC4409516 DOI: 10.1053/j.gastro.2015.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 01/25/2015] [Accepted: 02/04/2015] [Indexed: 01/19/2023]
Abstract
BACKGROUND & AIMS Intraepithelial T lymphocyte cells (IEL) are the first immune cells to respond to pathogens; they help maintain the integrity of the epithelial barrier. We studied the function of the mouse glycoprotein Signaling Lymphocyte Activation Molecule Family receptor (SLAMF) 4 (encoded by Slamf4) on the surface of CD8αβ αβ T-cell receptor (TCR)(+) IELs, and the roles of these cells in homeostasis of the small intestine in mice. METHODS SLAMF4(-) CD8(+) αβTCR(+) cells isolated from spleens of OT-I Rag1(-/-) mice were induced to express gut-homing receptors and transferred to C57BL/6J mice; levels of SLAMF4(+) cells were measured in small intestine tissues. After administration of anti-CD3 or antigen, with or without anti-SLAM4, to C57BL/6J and Slamf4(-/-) mice, CD8αβ αβTCR(+) IELs were collected; cytokine production and cytotoxicity were measured. Depletion of CX3CR1(+) phagocytes was assessed in mice by live-cell confocal imaging or by cytofluorometry; small intestine tissues were analyzed by histology and inflammation was quantified. RESULTS Splenic CD8(+) αβTCR(+) cells began to express SLAMF4 only after migrating to the small intestine. Injection of C57BL/6J mice with anti-SLAMF4 and anti-CD3 increased levels of interleukin 10 and interferon gamma secretion by IEL, compared with injection of anti-CD3 only. Similarly, the number of granzyme B(+) cytotoxic CD8(+) αβTCR(+) IELs increased in Slamf4(-/-) mice after injection of anti-CD3 and anti-SLAMF4, administration of antigen, or injection of anti-CD3. Surprisingly, in vivo activation of CD8αβ(+) IELs with anti-CD3 or antigen caused transient depletion of CX3CR1(+) phagocytes, which was prolonged by co-injection with anti-SLAMF4 or in Slamf4(-/-) mice. Anti-CD3 aggravated inflammation in the small intestines of Slamf4(-/-) mice and Eat2a(-/-)Eat2b(-/-) mice, indicated by flattened villi and crypt hyperplasia. CONCLUSIONS In mice, the intestinal environment induces SLAMF4 expression and localization to the surface of CD8(+) αβTCR(+) IELs. Signaling via SLAMF4 controls expansion of cytotoxic CD8αβ(+) IELs, which regulate the reversible depletion of lamina propria phagocytes and inflammation in the small intestine.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/deficiency
- Adaptor Proteins, Signal Transducing/genetics
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- CX3C Chemokine Receptor 1
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Cytokines/immunology
- Cytokines/metabolism
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Homeostasis
- Hyperplasia
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Intestine, Small/immunology
- Intestine, Small/metabolism
- Intestine, Small/pathology
- Lymphocyte Activation
- Mice, Inbred C57BL
- Mice, Knockout
- Phagocytes/immunology
- Phagocytes/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
- Receptors, Immunologic/deficiency
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Signal Transduction
- Signaling Lymphocytic Activation Molecule Family
- Spleen/immunology
- Spleen/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
Collapse
Affiliation(s)
- Michael S. O’Keeffe
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02115. USA
| | - Joo-Hye Song
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114. USA
| | - Gongxian Liao
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02115. USA. Tel: 1-617-735-4141, Fax: (617) 735-4140
| | - Jaime De Calisto
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02115. USA
| | - Peter J. Halibozek
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02115. USA
| | - J. Rodrigo Mora
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114. USA
| | - Atul K. Bhan
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ninghai Wang
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02115. USA
| | - Hans-Christian Reinecker
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114. USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02115. USA. Tel: (617) 735-4131; Fax: (617) 735-4135
| |
Collapse
|
20
|
Wang N, Halibozek PJ, Yigit B, Zhao H, O'Keeffe MS, Sage P, Sharpe A, Terhorst C. Negative Regulation of Humoral Immunity Due to Interplay between the SLAMF1, SLAMF5, and SLAMF6 Receptors. Front Immunol 2015; 6:158. [PMID: 25926831 PMCID: PMC4396446 DOI: 10.3389/fimmu.2015.00158] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 03/23/2015] [Indexed: 12/30/2022] Open
Abstract
Whereas the SLAMF-associated protein (SAP) is involved in differentiation of T follicular helper (Tfh) cells and antibody responses, the precise requirements of SLAMF receptors in humoral immune responses are incompletely understood. By analyzing mice with targeted disruptions of the Slamf1, Slamf5, and Slamf6 genes, we found that both T-dependent and T-independent antibody responses were twofold higher compared to those in single knockout mice. These data suggest a suppressive synergy of SLAMF1, SLAMF5, and SLAMF6 in humoral immunity, which contrasts the decreased antibody responses resulting from a defective GC reaction in the absence of the adapter SAP. In adoptive co-transfer assays, both [Slamf1 + 5 + 6]−/− B and T cells were capable of inducing enhanced antibody responses, but more pronounced enhancement was observed after adoptive transfer of [Slamf1 + 5 + 6]−/− B cells compared to that of [Slamf1 + 5 + 6]−/− T cells. In support of [Slamf1 + 5 + 6]−/− B cell intrinsic activity, [Slamf1 + 5 + 6]−/− mice also mounted significantly higher antibody responses to T-independent type 2 antigen. Furthermore, treatment of mice with anti-SLAMF6 monoclonal antibody results in severe inhibition of the development of Tfh cells and GC B cells, confirming a suppressive effect of SLAMF6. Taken together, these results establish SLAMF1, SLAMF5, and SLAMF6 as important negative regulators of humoral immune response, consistent with the notion that SLAM family receptors have dual functions in immune responses.
Collapse
Affiliation(s)
- Ninghai Wang
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Peter J Halibozek
- 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
| | - Hui Zhao
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Michael S O'Keeffe
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Peter Sage
- Department of Microbiology and Immunology, Harvard Medical School , Boston, MA , USA
| | - Arlene Sharpe
- Department of Microbiology and Immunology, Harvard Medical School , Boston, MA , USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| |
Collapse
|