1
|
Li J, Fan T, Wang D, Xiao C, Deng Z, Cai W, Ji Y, Li C, He J. SLAMF receptors: key regulators of tumor progression and emerging targets for cancer immunotherapy. Mol Cancer 2025; 24:145. [PMID: 40382610 PMCID: PMC12084948 DOI: 10.1186/s12943-025-02308-8] [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: 12/10/2024] [Accepted: 03/18/2025] [Indexed: 05/20/2025] Open
Abstract
The signaling lymphocytic activation molecule family (SLAMF) consists of nine distinct cell surface receptors predominantly expressed on immune cells, each characterized by unique structural features, expression patterns, downstream signaling pathways, and biological functions. These receptors play critical roles in modulating various immune cell activities within the tumor microenvironment, thereby shaping immune responses in cancer. Although accumulating evidence demonstrates their value as therapeutic targets for developing cancer immunotherapies, the full spectrum of SLAMF receptors in cancer remains incompletely understood. This review aims to provide a comprehensive overview of the molecular characteristics and immunomodulatory functions of each SLAMF receptor, underscoring their pivotal contributions to cancer progression. Furthermore, we also highlight their potential as promising targets for advancing cancer immunotherapeutic strategies. Finally, we discuss clinical trials evaluating the efficacy and safety of SLAMF receptor-based immunotherapies, emphasizing their translational relevance in the development of cancer treatments.
Collapse
Affiliation(s)
- Jia Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Di Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenpeng Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yu Ji
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
2
|
Liu A, Bai P, You H, Zhuang Z, Tian F, Weng H, Wei X, Tang L, Wang L, Liu C, Zhang J, Sun M, Zhang S, Shu X, Ge J. SLAMF7 Restrains Pro-Inflammatory Macrophage Activation to Counteract Doxorubicin-Induced Cardiotoxicity. JACC Basic Transl Sci 2025:S2452-302X(25)00073-7. [PMID: 40372307 DOI: 10.1016/j.jacbts.2025.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/14/2025] [Accepted: 02/20/2025] [Indexed: 05/16/2025]
Abstract
Doxorubicin-induced cardiotoxicity (DIC) poses a significant challenge in cancer treatment. This study investigated the role of SLAMF7 in DIC, particularly in macrophage-mediated inflammation. Using SLAMF7 knockout mice, we found that SLAMF7 deficiency exacerbates DIC and amplifies inflammatory responses. Mechanistically, SLAMF7 interacts with TNF receptor-associated factor 6 to attenuate nuclear factor κB signaling, reducing oxidative stress and proinflammatory cytokines. Notably, administering recombinant SLAMF7 protein effectively mitigated DIC. These findings underscore the critical role of SLAMF7 in protecting against DIC, positioning it as a promising therapeutic target.
Collapse
Affiliation(s)
- Ao Liu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Peiyuan Bai
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Hongmin You
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zehao Zhuang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Fangyan Tian
- Department of Ultrasound Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Haobo Weng
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Xuemei Wei
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Lu Tang
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Litao Wang
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaobao Liu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jinghong Zhang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Minmin Sun
- Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Shuning Zhang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xianhong Shu
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai Institute of Medical Imaging, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases and Institutes of Biomedical Sciences, Fudan University, Shanghai, China; State Key Laboratory of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai, China.
| |
Collapse
|
3
|
Altalbawy FMA, Babamuradova Z, Baldaniya L, Singh A, Singh KU, Ballal S, Sabarivani A, Sead FF, Alam R, Alshahrani MY. The multifaceted role of CS1 (SLAMF7) in immunoregulation: Implications for cancer therapy and autoimmune disorders. Exp Cell Res 2025; 447:114516. [PMID: 40073958 DOI: 10.1016/j.yexcr.2025.114516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/09/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
CS1 (SLAMF7), a pivotal immune receptor, plays a dual role in modulating immune responses in autoimmune diseases and cancer. In autoimmunity, aberrant CS1 signaling contributes to the activation of autoreactive lymphocytes, driving pathologies such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Conversely, in oncology, CS1 serves as a promising immunotherapeutic target, exemplified by the efficacy of the monoclonal antibody Elotuzumab in multiple myeloma. CS1 mediates immune cell functions through intricate signaling pathways, including interactions with EAT-2 and SAP adaptors, which influence cytotoxicity, cytokine production, and immune homeostasis. Beyond cancer and autoimmune diseases, soluble and membrane-bound forms of CS1 are emerging as biomarkers and potential therapeutic targets. Despite significant progress, gaps remain in understanding CS1\u2019s mechanisms, variability in expression, and role in other diseases. This study explores the multifaceted functions of CS1, proposing innovative strategies to leverage its therapeutic potential across diverse pathologies.
Collapse
Affiliation(s)
- Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia; National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza 12613, Egypt
| | - Zarrina Babamuradova
- Internal Diseases of Pediatric Faculty, Samarkand State Medical University, Samarkand, Uzbekistan
| | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Kamred Udham Singh
- School of Computing, Graphic Era Hill University, Dehradun, India; Graphic Era Deemed to Be University, Dehradun, Uttarakhand, 248002, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - A Sabarivani
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Fadhil Faez Sead
- Department of Dentistry, College of Dentistry, The Islamic University, Najaf, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Rubyat Alam
- Applied Chemistry & Chemical Engineering, University of Dhaka, Bangladesh
| | - Mohammad Y Alshahrani
- Central Labs, King Khalid University, AlQura'a, Abha, Saudi Arabia; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| |
Collapse
|
4
|
He C, Pan Z, Liu Y, Zhou H, Li L. SLAMF7 is a key molecule that promotes M1 polarization in lung tissue macrophages of high-fat diet-fed asthma mice model. Int Immunopharmacol 2025; 149:114203. [PMID: 39904038 DOI: 10.1016/j.intimp.2025.114203] [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: 10/21/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
OBJECTIVE Investigating the regulatory role of Signaling lymphocyte activation molecule family 7 (SLAMF7) in the pathogenesis of asthma in a high fat-fed (HFD) mouse model, providing targets for treating obese asthma. METHODS We constructed a mouse model of obese asthma, and Quantitative real-time RT-PCR (qPCR) for the detection of mRNA levels of SLAMF7 and M1 polarization markers of macrophages. Lung tissue levels of SLAMF7 protein, macrophage M1 polarization markers, and neutrophil markers were measured by Western blotting. The proportions of SLAMF7+ macrophages and neutrophils in bronchoalveolar lavage fluid (BALF) were determined by flow cytometry. Neutrophil inflammatory cytokine levels were determined by Enzyme-linked immunosorbent assay (ELISA). Immunofluorescence performed the colocalization of SLAMF7 and inducible nitric oxide synthase (iNOS). The regulation of SLAMF7 on M1 polarization of macrophages was verified by cell experiments. RESULTS The group of HFD asthmatic mice had more severe airway inflammation and mucus secretion. They also had higher SLAMF7 levels, airway neutrophil inflammation and M1 polarization of macrophages in lung tissue. SLAMF7 overexpression increased M1 polarization, and SLAMF7 knockdown decreased M1 polarization. The expression change of SLAMF7 affects the expression of NR4A1 and RUNX3, inhibiting NR4A1 and promoting RUNX3. CONCLUSION SLAMF7 expression is increased in obese asthma mice, accompanied by neutrophil infiltration and enhanced M1 polarization. SLAMF7 promotes M1 polarization may be through the NR4A1-RUNX3 axis, suppressing NR4A1, and promoting RUNX3.
Collapse
Affiliation(s)
- Cengceng He
- Department of Respiratory Medicine & Clinical Allergy Center, Affiliated Children's Hospital of Jiangnan University, Wuxi, 299-1 Qingyang Road, Wuxi 214023, China
| | - Zhenzhen Pan
- Department of Respiratory Medicine & Clinical Allergy Center, Affiliated Children's Hospital of Jiangnan University, Wuxi, 299-1 Qingyang Road, Wuxi 214023, China
| | - Yanchen Liu
- Department of Respiratory Medicine & Clinical Allergy Center, Affiliated Children's Hospital of Jiangnan University, Wuxi, 299-1 Qingyang Road, Wuxi 214023, China
| | - Huan Zhou
- Department of Respiratory Medicine & Clinical Allergy Center, Affiliated Children's Hospital of Jiangnan University, Wuxi, 299-1 Qingyang Road, Wuxi 214023, China
| | - Ling Li
- Department of Respiratory Medicine & Clinical Allergy Center, Affiliated Children's Hospital of Jiangnan University, Wuxi, 299-1 Qingyang Road, Wuxi 214023, China.
| |
Collapse
|
5
|
Zhang Z, Zhang Y, Chen Z, Xia L. Emerging roles of SLAMF7 in immune cells and related diseases. Innate Immun 2025; 31:17534259251326700. [PMID: 40091370 PMCID: PMC11912174 DOI: 10.1177/17534259251326700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/21/2024] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
Immune cells are heterogeneous and perform different functions in different microenvironment, thus playing different roles in different stages of diseases. Studies have shown that immune cells are involved in the pathogenesis of many diseases, and there is a causal association of immune cells with disease states. Signaling Lymphocyte Activation Molecule family (SLAMF) members are a newly appreciated group of specific receptors that are mainly expressed in immune cells and whose role is to regulate the function of immune cells. SLAMF7, also known as CD319, has been widely reported in multiple myeloma, and in recent years, more and more studies have shown that SLAMF7 is widely involved in the function of immune cells and the progression of breast cancer, acquired immune deficiency syndrome, systemic lupus erythematosus and other immune cells-related diseases. However, the mechanisms underlying the regulatory role of SLAMF7 on immune cells, and the impact on the progression of immune cells-related diseases remain poorly elucidated. In this review, we summarize current knowledge about the role of SLAMF7 in immune cells and related diseases such as cancer, infectious disease, autoimmune disease and atherosclerosis, and the therapeutic strategy targeting SLAMF7 is also described. By better understanding the role and regulation of SLAMF7, we hope to provide new insights and directions for improving the diagnosis and treatment of inflammation.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ying Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zeyu Chen
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lin Xia
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Hematological Disease, Jiangsu University, Zhenjiang, China
| |
Collapse
|
6
|
Kared H, Tan C, Narang V, Tan SW, Xian CH, Wei ATS, Lum J, Ruiz-Mateos E, Rajasuriar R, Kamarulzaman A, Ng TP, Larbi A. SLAMF7 defines subsets of human effector CD8 T cells. Sci Rep 2024; 14:30779. [PMID: 39730488 DOI: 10.1038/s41598-024-80971-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024] Open
Abstract
Long-term control of viral replication relies on the efficient differentiation of memory T cells into effector T cells during secondary immune responses. Recent findings have identified T cell precursors for both memory and exhausted T cells, suggesting the existence of progenitor-like effector T cells. These cells can persist without antigenic challenge but expand and acquire effector functions upon recall immune responses. In this study, we demonstrate that the combination of SLAMF7 with either CD27 or TCF-1 effectively identifies progenitor-like effector CD8 T cells, while SLAMF7 with GPR56 or TOX defines effector CD8 T cells. These markers allow for the clear segregation of these distinct cell subsets. SLAMF7+ CD8T cells are dynamically modulated during viral infections, including HIV, HCV, CMV, and SARS-CoV-2, as well as during aging. We further characterize the SLAMF7 signature at both phenotypic and transcriptional levels. Notably, during aging, the SLAMF7 pathway becomes dysregulated, resulting in persistent phosphorylation of STAT1. Additionally, SLAMF7 ligation in the presence of IL-15 induces TCF-1 expression, which promotes the homeostatic proliferation of progenitor-like effector CD8 T cells.
Collapse
Affiliation(s)
- Hassen Kared
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore.
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| | - Crystal Tan
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Vipin Narang
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Shu Wen Tan
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Chin Hui Xian
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Alicia Tay Seok Wei
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Josephine Lum
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Ezequiel Ruiz-Mateos
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Reena Rajasuriar
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Adeeba Kamarulzaman
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Tze Pin Ng
- Gerontology Research Programme and Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| |
Collapse
|
7
|
King EM, McClendon J, Trinh T, Matsuda JL, Lyn-Kew KH, McCubbrey AL, Mould KJ, Henson PM, Janssen WJ. Slamf7 is dispensable in mouse models of acute lung injury. ERJ Open Res 2024; 10:00345-2024. [PMID: 39377087 PMCID: PMC11456964 DOI: 10.1183/23120541.00345-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/14/2024] [Indexed: 10/09/2024] Open
Abstract
Slamf7 is expressed by monocyte-derived macrophages recruited to the lungs during injury. Whole-body and macrophage-specific knockouts of Slamf7 had no effect on the degree of inflammation in three mouse models of acute lung injury. https://bit.ly/3KgTJg1.
Collapse
Affiliation(s)
- Emily M. King
- University of Colorado Anschutz Medical Campus, Department of Medicine, Aurora, CO, USA
| | | | | | | | | | - Alexandra L. McCubbrey
- University of Colorado Anschutz Medical Campus, Department of Medicine, Aurora, CO, USA
- National Jewish Health, Department of Medicine, Denver, CO, USA
| | - Kara J. Mould
- University of Colorado Anschutz Medical Campus, Department of Medicine, Aurora, CO, USA
- National Jewish Health, Department of Medicine, Denver, CO, USA
| | - Peter M. Henson
- University of Colorado Anschutz Medical Campus, Department of Medicine, Aurora, CO, USA
- National Jewish Health, Department of Pediatrics, Denver, CO, USA
| | - William J. Janssen
- University of Colorado Anschutz Medical Campus, Department of Medicine, Aurora, CO, USA
- National Jewish Health, Department of Medicine, Denver, CO, USA
| |
Collapse
|
8
|
Xiao L, Zhang L, Guo C, Xin Q, Gu X, Jiang C, Wu J. "Find Me" and "Eat Me" signals: tools to drive phagocytic processes for modulating antitumor immunity. Cancer Commun (Lond) 2024; 44:791-832. [PMID: 38923737 PMCID: PMC11260773 DOI: 10.1002/cac2.12579] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Phagocytosis, a vital defense mechanism, involves the recognition and elimination of foreign substances by cells. Phagocytes, such as neutrophils and macrophages, rapidly respond to invaders; macrophages are especially important in later stages of the immune response. They detect "find me" signals to locate apoptotic cells and migrate toward them. Apoptotic cells then send "eat me" signals that are recognized by phagocytes via specific receptors. "Find me" and "eat me" signals can be strategically harnessed to modulate antitumor immunity in support of cancer therapy. These signals, such as calreticulin and phosphatidylserine, mediate potent pro-phagocytic effects, thereby promoting the engulfment of dying cells or their remnants by macrophages, neutrophils, and dendritic cells and inducing tumor cell death. This review summarizes the phagocytic "find me" and "eat me" signals, including their concepts, signaling mechanisms, involved ligands, and functions. Furthermore, we delineate the relationships between "find me" and "eat me" signaling molecules and tumors, especially the roles of these molecules in tumor initiation, progression, diagnosis, and patient prognosis. The interplay of these signals with tumor biology is elucidated, and specific approaches to modulate "find me" and "eat me" signals and enhance antitumor immunity are explored. Additionally, novel therapeutic strategies that combine "find me" and "eat me" signals to better bridge innate and adaptive immunity in the treatment of cancer patients are discussed.
Collapse
Affiliation(s)
- Lingjun Xiao
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
| | - Louqian Zhang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
| | - Ciliang Guo
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
| | - Qilei Xin
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| | - Xiaosong Gu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| |
Collapse
|
9
|
Wu JJ, Zhang PA, Chen MZ, Zhang Y, Du WS, Li XN, Ji GC, Jiang LD, Jiao Y, Li X. Analysis of Key Genes and miRNA-mRNA Networks Associated with Glucocorticoids Treatment in Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2024; 19:589-605. [PMID: 38435123 PMCID: PMC10909375 DOI: 10.2147/copd.s441716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/21/2024] [Indexed: 03/05/2024] Open
Abstract
Background Some patients with chronic obstructive pulmonary disease (COPD) benefit from glucocorticoid (GC) treatment, but its mechanism is unclear. Objective With the help of the Gene Expression Omnibus (GEO) database, the key genes and miRNA-mRNA related to the treatment of COPD by GCs were discussed, and the potential mechanism was explained. Methods The miRNA microarray dataset (GSE76774) and mRNA microarray dataset (GSE36221) were downloaded, and differential expression analysis were performed. Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed on the differentially expressed genes (DEGs). The protein interaction network of the DEGs in the regulatory network was constructed with the STRING database, and the key genes were screened through Cytoscape. Potential downstream target genes regulated by differentially expressed miRNAs (DEMs) were predicted by the miRWalk3.0 database, and miRNA-mRNA regulatory networks were constructed. Finally, some research results were validated. Results ① Four DEMs and 83 DEGs were screened; ② GO and KEGG enrichment analysis mainly focused on the PI3K/Akt signalling pathway, ECM receptor interaction, etc.; ③ CD2, SLAMF7, etc. may be the key targets of GC in the treatment of COPD; ④ 18 intersection genes were predicted by the mirwalk 3.0 database, and 9 pairs of miRNA-mRNA regulatory networks were identified; ⑤ The expression of miR-320d-2 and TFCP2L1 were upregulated by dexamethasone in the COPD cell model, while the expression of miR-181a-2-3p and SLAMF7 were downregulated. Conclusion In COPD, GC may mediate the expression of the PI3K/Akt signalling pathway through miR-181a-2-3p, miR-320d-2, miR-650, and miR-155-5p, targeting its downstream signal factors. The research results provide new ideas for RNA therapy strategies of COPD, and also lay a foundation for further research.
Collapse
Affiliation(s)
- Jian-Jun Wu
- Respiratory Department, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Ping-An Zhang
- Respiratory Department, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Ming-Zhe Chen
- Infectious Disease Department, Henan Provincial Hospital of Traditional Chinese Medicine, Zhengzhou, Henan, People’s Republic of China
| | - Yi Zhang
- Respiratory Department, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Wei-Sha Du
- Respiratory Department, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Xiao-Ning Li
- Respiratory Department, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Guo-Chao Ji
- Respiratory Department, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Liang-Duo Jiang
- Respiratory Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Yang Jiao
- Respiratory Department, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Xin Li
- Glaucoma Department, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
10
|
Choe U, Pham Q, Kim YS, Yu L, Wang TTY. Identification and elucidation of cross talk between SLAM Family Member 7 (SLAMF7) and Toll-like receptor (TLR) pathways in monocytes and macrophages. Sci Rep 2023; 13:11007. [PMID: 37420084 PMCID: PMC10329007 DOI: 10.1038/s41598-023-37040-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/14/2023] [Indexed: 07/09/2023] Open
Abstract
To further elucidate the expression, regulation and function of Signaling Lymphocytic Activation Molecule Family (SLAMF) protein members in human monocytes and macrophages. Un-differentiated monocytic THP-1 cell (u-THP-1) and differentiated THP-1 macrophage (d-THP-1) were used as culture models in the study. Responses of cells to the differentiation agents phorbol ester (25 ng/ml) and TLR (Toll-like receptor) ligands were assessed. RT-PCR and Western blot analysis were used to determine mRNA and protein level. Pro-inflammatory cytokine mRNA expression levels and phagocytosis were used as functional markers. Data analyzed using t-test, one-way or two-way ANOVA followed by post hoc test. SLAMFs were differentially expressed in THP-1 cells. Differentiation of u-THP-1 to d-THP-1 led to significantly higher SLAMF7 mRNA and protein levels than other SLAMF. In addition, TLR stimuli increased SLAMF7 mRNA expression but not protein expression. Importantly, SLAMF7 agonist antibody and TLR ligands synergistically increased the mRNA expression levels of IL-1β, IL-6 and TNF-α, but had no effect on phagocytosis. SLAMF7 knocked-down in d-THP-1 significantly lowered TLR-induced mRNA expressions of pro-inflammatory markers. SLAM family proteins are differentially regulated by differentiation and TLRs. SLAMF7 enhanced TLR-mediated induction of pro-inflammatory cytokines in monocytes and macrophages but not phagocytosis.
Collapse
Affiliation(s)
- Uyory Choe
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, 20742, USA
| | - Quynhchi Pham
- U.S. Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Beltsville, MD, 20705, USA
| | - Young S Kim
- Cancer Prevention Science Branch, Division of Cancer Prevention, NCI, Rockville, MD, 20850, USA
| | - Liangli Yu
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, 20742, USA
| | - Thomas T Y Wang
- U.S. Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics and Immunology Laboratory, Beltsville, MD, 20705, USA.
| |
Collapse
|
11
|
Wu Y, Wang Q, Li M, Lao J, Tang H, Ming S, Wu M, Gong S, Li L, Liu L, Huang X. SLAMF7 regulates the inflammatory response in macrophages during polymicrobial sepsis. J Clin Invest 2023; 133:150224. [PMID: 36749634 PMCID: PMC10014109 DOI: 10.1172/jci150224] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 01/27/2023] [Indexed: 02/08/2023] Open
Abstract
Uncontrolled inflammation occurred in sepsis results in multiple organ injuries and shock, which contributes to the death of patients with sepsis. However, the regulatory mechanisms that restrict excessive inflammation are still elusive. Here, we identified an Ig-like receptor called signaling lymphocyte activation molecular family 7 (SLAMF7) as a key suppressor of inflammation during sepsis. We found that the expression of SLAMF7 on monocytes/macrophages was significantly elevated in patients with sepsis and in septic mice. SLAMF7 attenuated TLR-dependent MAPK and NF-κB signaling activation in macrophages by cooperating with Src homology 2-containing inositol-5'‑phosphatase 1 (SHIP1). Furthermore, SLAMF7 interacted with SHIP1 and TNF receptor-associated factor 6 (TRAF6) to inhibit K63 ubiquitination of TRAF6. In addition, we found that tyrosine phosphorylation sites within the intracellular domain of SLAMF7 and the phosphatase domain of SHIP1 were indispensable for the interaction between SLAMF7, SHIP1, and TRAF6 and SLAMF7-mediated modulation of cytokine production. Finally, we demonstrated that SLAMF7 protected against lethal sepsis and endotoxemia by downregulating macrophage proinflammatory cytokines and suppressing inflammation-induced organ damage. Taken together, our findings reveal a negative regulatory role of SLAMF7 in polymicrobial sepsis, thus providing sights into the treatment of sepsis.
Collapse
Affiliation(s)
- Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Qiaohua Wang
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Miao Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Juanfeng Lao
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Huishu Tang
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Siqi Ming
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital of the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Minhao Wu
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Sitang Gong
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Linhai Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong Province, China
| | - Lei Liu
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital of the Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province, China.,National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital of the Southern University of Science and Technology, Shenzhen, Guangdong Province, China.,The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong Province, China
| |
Collapse
|
12
|
Powers SB, Ahmed NG, Jose R, Brezgiel M, Aryal S, Bowman WP, Mathew PA, Mathew SO. Differential Expression of LLT1, SLAM Receptors CS1 and 2B4 and NCR Receptors NKp46 and NKp30 in Pediatric Acute Lymphoblastic Leukemia (ALL). Int J Mol Sci 2023; 24:ijms24043860. [PMID: 36835271 PMCID: PMC9959214 DOI: 10.3390/ijms24043860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) represents the most common pediatric cancer. Most patients (85%) develop B-cell ALL; however, T-cell ALL tends to be more aggressive. We have previously identified 2B4 (SLAMF4), CS1 (SLAMF7) and LLT1 (CLEC2D) that can activate or inhibit NK cells upon the interaction with their ligands. In this study, the expression of 2B4, CS1, LLT1, NKp30 and NKp46 was determined. The expression profiles of these immune receptors were analyzed in the peripheral blood mononuclear cells of B-ALL and T-ALL subjects by single-cell RNA sequencing data obtained from the St. Jude PeCan data portal that showed increased expression of LLT1 in B-ALL and T-ALL subjects. Whole blood was collected from 42 pediatric ALL subjects at diagnosis and post-induction chemotherapy and 20 healthy subjects, and expression was determined at the mRNA and cell surface protein level. A significant increase in cell surface LLT1 expression in T cells, monocytes and NK cells was observed. Increased expression of CS1 and NKp46 was observed on monocytes of ALL subjects at diagnosis. A decrease of LLT1, 2B4, CS1 and NKp46 on T cells of ALL subjects was also observed post-induction chemotherapy. Furthermore, mRNA data showed altered expression of receptors in ALL subjects pre- and post-induction chemotherapy treatment. The results indicate that the differential expression of the receptors/ligand may play a role in the T-cell- and NK-cell-mediated immune surveillance of pediatric ALL.
Collapse
Affiliation(s)
- Sheila B. Powers
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Nourhan G. Ahmed
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Roslin Jose
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Marissa Brezgiel
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Subhash Aryal
- School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - W. Paul Bowman
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Cook Children’s Medical Center, 801 7th Avenue, Fort Worth, TX 76104, USA
| | - Porunelloor A. Mathew
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Stephen O. Mathew
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- Correspondence:
| |
Collapse
|
13
|
O'Connell P, Blake MK, Godbehere S, Amalfitano A, Aldhamen YA. SLAMF7 modulates B cells and adaptive immunity to regulate susceptibility to CNS autoimmunity. J Neuroinflammation 2022; 19:241. [PMID: 36199066 PMCID: PMC9533612 DOI: 10.1186/s12974-022-02594-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 09/08/2022] [Indexed: 12/02/2022] Open
Abstract
Background Multiple sclerosis (MS) is a chronic, debilitating condition characterized by CNS autoimmunity stemming from a complex etiology involving both environmental and genetic factors. Our current understanding of MS points to dysregulation of the immune system as the pathogenic culprit, however, it remains unknown as to how the many genes associated with increased susceptibility to MS are involved. One such gene linked to MS susceptibility and known to regulate immune function is the self-ligand immune cell receptor SLAMF7. Methods We subjected WT and SLAMF7−/− mice to multiple EAE models, compared disease severity, and comprehensively profiled the CNS immune landscape of these mice. We identified all SLAMF7-expressing CNS immune cells and compared the entire CNS immune niche between genotypes. We performed deep phenotyping and in vitro functional studies of B and T cells via spectral cytometry and BioPlex assays. Adoptive transfer studies involving the transfer of WT and SLAMF7−/− B cells into B cell-deficient mice (μMT) were also performed. Finally, B–T cell co-culture studies were performed, and a comparative cell–cell interaction network derived from scRNA-seq data of SLAMF7+ vs. SLAMF7− human CSF immune cells was constructed. Results We found SLAMF7−/− mice to be more susceptible to EAE compared to WT mice and found SLAMF7 to be expressed on numerous CNS immune cell subsets. Absence of SLAMF7 did not grossly alter the CNS immune landscape, but allowed for altered immune cell subset infiltration during EAE in a model-dependent manner. Global lack of SLAMF7 expression increased myeloid cell activation states along with augmented T cell anti-MOG immunity. B cell profiling studies revealed increased activation states of specific plasma and B cell subsets in SLAMF7−/− mice during EAE, and functional co-culture studies determined that SLAMF7−/− B cells induce exaggerated T cell activation. Adoptive transfer studies revealed that the increased susceptibility of SLAMF7−/− mice to EAE is partly B cell dependent and reconstruction of the human CSF SLAMF7-interactome found B cells to be critical to cell–cell communication between SLAMF7-expressing cells. Conclusions Our studies have identified novel roles for SLAMF7 in CNS immune regulation and B cell function, and illuminate underpinnings of the genetic association between SLAMF7 and MS. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02594-9.
Collapse
Affiliation(s)
- Patrick O'Connell
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4108 Biomedical and Physical Sciences Building, East Lansing, MI, 48824, USA
| | - Maja K Blake
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4108 Biomedical and Physical Sciences Building, East Lansing, MI, 48824, USA
| | - Sarah Godbehere
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4108 Biomedical and Physical Sciences Building, East Lansing, MI, 48824, USA
| | - Andrea Amalfitano
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4108 Biomedical and Physical Sciences Building, East Lansing, MI, 48824, USA.,Department of Pediatrics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Yasser A Aldhamen
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4108 Biomedical and Physical Sciences Building, East Lansing, MI, 48824, USA.
| |
Collapse
|
14
|
Chen Y, Liang L, Wu C, Cao Z, Xia L, Meng J, Wang Z. Epigenetic Control of Vascular Smooth Muscle Cell Function in Atherosclerosis: A Role for DNA Methylation. DNA Cell Biol 2022; 41:824-837. [PMID: 35900288 DOI: 10.1089/dna.2022.0278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis is a complex vascular inflammatory disease in which multiple cell types are involved, including vascular smooth muscle cells (VSMCs). In response to vascular injury and inflammatory stimuli, VSMCs undergo a "phenotypic switching" characterized by extracellular matrix secretion, loss of contractility, and abnormal proliferation and migration, which play a key role in the progression of atherosclerosis. DNA methylation modification is an important epigenetic mechanism that plays an important role in atherosclerosis. Studies investigating abnormal DNA methylation in patients with atherosclerosis have determined a specific DNA methylation profile, and proposed multiple pathways and genes involved in the etiopathogenesis of atherosclerosis. Recent studies have also revealed that DNA methylation modification controls VSMC function by regulating gene expression involved in atherosclerosis. In this review, we summarize the recent advances regarding the epigenetic control of VSMC function by DNA methylation in atherosclerosis and provide insights into the development of VSMC-centered therapeutic strategies.
Collapse
Affiliation(s)
- Yanjun Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Lingli Liang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Chunyan Wu
- The Third Affiliated Hospital of University of South China, Hengyang, China
| | - Zitong Cao
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Linzhen Xia
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| | - Jun Meng
- Functional Department, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Zuo Wang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
15
|
Richardson K, Keam SP, Zhu JJ, Meyran D, D’Souza C, Macdonald S, Campbell K, Robbins M, Bezman NA, Todd K, Quach H, Ritchie DS, Harrison SJ, Prince HM, Trapani JA, Jenkins MR, Beavis PA, Darcy PK, Neeson PJ. The efficacy of combination treatment with elotuzumab and lenalidomide is dependent on crosstalk between natural killer cells, monocytes and myeloma cells. Haematologica 2022; 108:83-97. [PMID: 35770527 PMCID: PMC9827168 DOI: 10.3324/haematol.2021.279930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Indexed: 02/04/2023] Open
Abstract
Patients with refractory relapsed multiple myeloma respond to combination treatment with elotuzumab and lenalidomide. The mechanisms underlying this observation are not fully understood. Furthermore, biomarkers predictive of response have not been identified to date. To address these issues, we used a humanized myeloma mouse model and adoptive transfer of human natural killer (NK) cells to show that elotuzumab and lenalidomide treatment controlled myeloma growth, and this was mediated through CD16 on NK cells. In co-culture studies, we showed that peripheral blood mononuclear cells from a subset of patients with refractory relapsed multiple myeloma were effective killers of OPM2 myeloma cells when treated with elotuzumab and lenalidomide, and this was associated with significantly increased expression of CD54 on OPM2 cells. Furthermore, elotuzumab- and lenalidomide-induced OPM2 cell killing and increased OPM2 CD54 expression were dependent on both monocytes and NK cells, and these effects were not mediated by soluble factors alone. At the transcript level, elotuzumab and lenalidomide treatment significantly increased OPM2 myeloma cell expression of genes for trafficking and adhesion molecules, NK cell activation ligands and antigen presentation molecules. In conclusion, our findings suggest that multiple myeloma patients require elotuzumab- and lenalidomide-mediated upregulation of CD54 on autologous myeloma cells, in combination with NK cells and monocytes to mediate an effective anti-tumor response. Furthermore, our data suggest that increased myeloma cell CD54 expression levels could be a powerful predictive biomarker for response to elotuzumab and lenalidomide treatment.
Collapse
Affiliation(s)
- Kelden Richardson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Simon P. Keam
- Tumor Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Joe Jiang Zhu
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Deborah Meyran
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Criselle D’Souza
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Sean Macdonald
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Kerry Campbell
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Michael Robbins
- Translational Medicine, Bristol-Myers Squibb, Cambridge, MA, USA,°Current address: io904 LLC, Jacksonville Beach, FL, USA
| | - Natalie A. Bezman
- Oncology Discovery Research, Bristol-Myers Squibb, Redwood City, CA, USA
| | - Kirsten Todd
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Hang Quach
- Department of Haematology, St Vincent’s Hospital, Melbourne, Australia,Faculty of Medicine, The University of Melbourne, Melbourne, Australia
| | - David S. Ritchie
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
| | - Simon J. Harrison
- Faculty of Medicine, The University of Melbourne, Melbourne, Australia,Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
| | - H. Miles Prince
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia,Faculty of Medicine, The University of Melbourne, Melbourne, Australia,Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
| | - Joseph A. Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Misty R. Jenkins
- Faculty of Medicine, The University of Melbourne, Melbourne, Australia,Immunology Division, Walter and Eliza Hall Institute, Melbourne, Australia,Institute for Molecular Science, La Trobe University, Bundoora, Australia
| | - Paul A. Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Phillip K. Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Paul J. Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia,P. Neeson
| |
Collapse
|
16
|
O'Connell P, Blake MK, Pepelyayeva Y, Hyslop S, Godbehere S, Angarita AM, Pereira-Hicks C, Amalfitano A, Aldhamen YA. Adenoviral delivery of an immunomodulatory protein to the tumor microenvironment controls tumor growth. Mol Ther Oncolytics 2022; 24:180-193. [PMID: 35036523 PMCID: PMC8741417 DOI: 10.1016/j.omto.2021.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/04/2021] [Indexed: 11/15/2022] Open
Abstract
Targeted modulation of the immune system against tumors can achieve responses in otherwise refractory cancers, which has spurred efforts aimed at optimizing such strategies. To this end, we have previously investigated cancer immunotherapy approaches using recombinant adenovirus vectors, as well as via modulation of the self-ligand receptor SLAMF7. Here, we present a gene transfer-based immunotherapy approach using targeted expression of a SLAMF7-Fc fusion construct directly into tumors at high concentrations via a recombinant adenoviral vector (Ad-SF7-Fc). Using multiple murine cancer models, we show that Ad-SF7-Fc can induce tumor control via augmentation of innate immunity; specifically, induction of type I interferons and activation of dendritic cells (DCs) and macrophages. Analogously, we find that modulating SLAMF7 signaling via an adenoviral vector expressing its intracellular adaptor, EAT-2, is also capable of inducing tumor control. Finally, we employ a novel in vivo prediction approach and dataset integration with machine learning to dissect how Ad-SF7-Fc modulates cell-type-specific responses in the tumor microenvironment to achieve tumor control. Thus, our novel combinatorial cancer immunotherapy highlights the benefit of multimodal immune modulation and lays a framework for combination with complementary approaches capable of inducing adaptive immune responses.
Collapse
Affiliation(s)
- Patrick O'Connell
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4194 Biomedical and Physical Sciences Building, East Lansing, MI 48824, USA
| | - Maja K. Blake
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4194 Biomedical and Physical Sciences Building, East Lansing, MI 48824, USA
| | - Yuliya Pepelyayeva
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4194 Biomedical and Physical Sciences Building, East Lansing, MI 48824, USA
| | - Sean Hyslop
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4194 Biomedical and Physical Sciences Building, East Lansing, MI 48824, USA
| | - Sarah Godbehere
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4194 Biomedical and Physical Sciences Building, East Lansing, MI 48824, USA
| | - Ariana M. Angarita
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4194 Biomedical and Physical Sciences Building, East Lansing, MI 48824, USA
| | - Cristiane Pereira-Hicks
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4194 Biomedical and Physical Sciences Building, East Lansing, MI 48824, USA
| | - Andrea Amalfitano
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4194 Biomedical and Physical Sciences Building, East Lansing, MI 48824, USA
- Department of Pediatrics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Yasser A. Aldhamen
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, 567 Wilson Road, 4194 Biomedical and Physical Sciences Building, East Lansing, MI 48824, USA
| |
Collapse
|
17
|
Simmons DP, Nguyen HN, Gomez-Rivas E, Jeong Y, Jonsson AH, Chen AF, Lange JK, Dyer GS, Blazar P, Earp BE, Coblyn JS, Massarotti EM, Sparks JA, Todd DJ, Accelerating Medicines Partnership ® (AMP ®) RA/SLE Network, Rao DA, Kim EY, Brenner MB. SLAMF7 engagement superactivates macrophages in acute and chronic inflammation. Sci Immunol 2022; 7:eabf2846. [PMID: 35148199 PMCID: PMC8991457 DOI: 10.1126/sciimmunol.abf2846] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Macrophages regulate protective immune responses to infectious microbes, but aberrant macrophage activation frequently drives pathological inflammation. To identify regulators of vigorous macrophage activation, we analyzed RNA-seq data from synovial macrophages and identified SLAMF7 as a receptor associated with a superactivated macrophage state in rheumatoid arthritis. We implicated IFN-γ as a key regulator of SLAMF7 expression and engaging SLAMF7 drove a strong wave of inflammatory cytokine expression. Induction of TNF-α after SLAMF7 engagement amplified inflammation through an autocrine signaling loop. We observed SLAMF7-induced gene programs not only in macrophages from rheumatoid arthritis patients but also in gut macrophages from patients with active Crohn's disease and in lung macrophages from patients with severe COVID-19. This suggests a central role for SLAMF7 in macrophage superactivation with broad implications in human disease pathology.
Collapse
Affiliation(s)
- Daimon P. Simmons
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Hung N. Nguyen
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Emma Gomez-Rivas
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Yunju Jeong
- Harvard Medical School, Boston, MA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - A. Helena Jonsson
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Antonia F. Chen
- Harvard Medical School, Boston, MA
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA
| | - Jeffrey K. Lange
- Harvard Medical School, Boston, MA
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA
| | - George S. Dyer
- Harvard Medical School, Boston, MA
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA
| | - Philip Blazar
- Harvard Medical School, Boston, MA
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA
| | - Brandon E. Earp
- Harvard Medical School, Boston, MA
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA
| | - Jonathan S. Coblyn
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Elena M. Massarotti
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Jeffrey A. Sparks
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Derrick J. Todd
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | | | - Deepak A. Rao
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Edy Y. Kim
- Harvard Medical School, Boston, MA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Michael B. Brenner
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| |
Collapse
|
18
|
Talker SC, Barut GT, Lischer HE, Rufener R, von Münchow L, Bruggmann R, Summerfield A. Monocyte biology conserved across species: Functional insights from cattle. Front Immunol 2022; 13:889175. [PMID: 35967310 PMCID: PMC9373011 DOI: 10.3389/fimmu.2022.889175] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/30/2022] [Indexed: 12/24/2022] Open
Abstract
Similar to human monocytes, bovine monocytes can be split into CD14highCD16- classical, CD14highCD16high intermediate and CD14-/dimCD16high nonclassical monocytes (cM, intM, and ncM, respectively). Here, we present an in-depth analysis of their steady-state bulk- and single-cell transcriptomes, highlighting both pronounced functional specializations and transcriptomic relatedness. Bulk gene transcription indicates pro-inflammatory and antibacterial roles of cM, while ncM and intM appear to be specialized in regulatory/anti-inflammatory functions and tissue repair, as well as antiviral responses and T-cell immunomodulation. Notably, intM stood out by high expression of several genes associated with antigen presentation. Anti-inflammatory and antiviral functions of ncM are further supported by dominant oxidative phosphorylation and selective strong responses to TLR7/8 ligands, respectively. Moreover, single-cell RNA-seq revealed previously unappreciated heterogeneity within cM and proposes intM as a transient differentiation intermediate between cM and ncM.
Collapse
Affiliation(s)
- Stephanie C. Talker
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- *Correspondence: Stephanie C. Talker,
| | - G. Tuba Barut
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Heidi E.L. Lischer
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Reto Rufener
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Artur Summerfield
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
19
|
Mao Y, Huang P, Wang Y, Wang M, Li MD, Yang Z. Genome-wide methylation and expression analyses reveal the epigenetic landscape of immune-related diseases for tobacco smoking. Clin Epigenetics 2021; 13:215. [PMID: 34886889 PMCID: PMC8662854 DOI: 10.1186/s13148-021-01208-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Smoking is a major causal risk factor for lung cancer, chronic obstructive pulmonary disease (COPD), cardiovascular disease (CVD), and is the main preventable cause of deaths in the world. The components of cigarette smoke are involved in immune and inflammatory processes, which may increase the prevalence of cigarette smoke-related diseases. However, the underlying molecular mechanisms linking smoking and diseases have not been well explored. This study was aimed to depict a global map of DNA methylation and gene expression changes induced by tobacco smoking and to explore the molecular mechanisms between smoking and human diseases through whole-genome bisulfite sequencing (WGBS) and RNA-sequencing (RNA-seq). RESULTS We performed WGBS on 72 samples (36 smokers and 36 nonsmokers) and RNA-seq on 75 samples (38 smokers and 37 nonsmokers), and cytokine immunoassay on plasma from 22 males (9 smokers and 13 nonsmokers) who were recruited from the city of Jincheng in China. By comparing the data of the two groups, we discovered a genome-wide methylation landscape of differentially methylated regions (DMRs) associated with smoking. Functional enrichment analyses revealed that both smoking-related hyper-DMR genes (DMGs) and hypo-DMGs were related to synapse-related pathways, whereas the hypo-DMGs were specifically related to cancer and addiction. The differentially expressed genes (DEGs) revealed by RNA-seq analysis were significantly enriched in the "immunosuppression" pathway. Correlation analysis of DMRs with their corresponding gene expression showed that genes affected by tobacco smoking were mostly related to immune system diseases. Finally, by comparing cytokine concentrations between smokers and nonsmokers, we found that vascular endothelial growth factor (VEGF) was significantly upregulated in smokers. CONCLUSIONS In sum, we found that smoking-induced DMRs have different distribution patterns in hypermethylated and hypomethylated areas between smokers and nonsmokers. We further identified and verified smoking-related DMGs and DEGs through multi-omics integration analysis of DNA methylome and transcriptome data. These findings provide us a comprehensive genomic map of the molecular changes induced by smoking which would enhance our understanding of the harms of smoking and its relationship with diseases.
Collapse
Affiliation(s)
- Ying Mao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Maiqiu Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming D Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Research Center for Air Pollution and Health, Zhejiang University, Hangzhou, China.
| | - Zhongli Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
20
|
Oommen AM, Cunningham S, O'Súilleabháin PS, Hughes BM, Joshi L. An integrative network analysis framework for identifying molecular functions in complex disorders examining major depressive disorder as a test case. Sci Rep 2021; 11:9645. [PMID: 33958659 PMCID: PMC8102631 DOI: 10.1038/s41598-021-89040-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/14/2021] [Indexed: 12/02/2022] Open
Abstract
In addition to the psychological depressive phenotype, major depressive disorder (MDD) patients are also associated with underlying immune dysregulation that correlates with metabolic syndrome prevalent in depressive patients. A robust integrative analysis of biological pathways underlying the dysregulated neural connectivity and systemic inflammatory response will provide implications in the development of effective strategies for the diagnosis, management and the alleviation of associated comorbidities. In the current study, focusing on MDD, we explored an integrative network analysis methodology to analyze transcriptomic data combined with the meta-analysis of biomarker data available throughout public databases and published scientific peer-reviewed articles. Detailed gene set enrichment analysis and complex protein–protein, gene regulatory and biochemical pathway analysis has been undertaken to identify the functional significance and potential biomarker utility of differentially regulated genes, proteins and metabolite markers. This integrative analysis method provides insights into the molecular mechanisms along with key glycosylation dysregulation underlying altered neutrophil-platelet activation and dysregulated neuronal survival maintenance and synaptic functioning. Highlighting the significant gap that exists in the current literature, the network analysis framework proposed reduces the impact of data gaps and permits the identification of key molecular signatures underlying complex disorders with multiple etiologies such as within MDD and presents multiple treatment options to address their molecular dysfunction.
Collapse
Affiliation(s)
- Anup Mammen Oommen
- Advanced Glycoscience Research Cluster (AGRC), National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - Stephen Cunningham
- Advanced Glycoscience Research Cluster (AGRC), National University of Ireland Galway, Galway, Ireland. .,Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland.
| | - Páraic S O'Súilleabháin
- Department of Psychology, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - Brian M Hughes
- School of Psychology, National University of Ireland Galway, Galway, Ireland
| | - Lokesh Joshi
- Advanced Glycoscience Research Cluster (AGRC), National University of Ireland Galway, Galway, Ireland. .,Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
21
|
Loughland JR, Woodberry T, Field M, Andrew DW, SheelaNair A, Dooley NL, Piera KA, Amante FH, Kenangalem E, Price RN, Engwerda CR, Anstey NM, McCarthy JS, Boyle MJ, Minigo G. Transcriptional profiling and immunophenotyping show sustained activation of blood monocytes in subpatent Plasmodium falciparum infection. Clin Transl Immunology 2020; 9:e1144. [PMID: 32566226 PMCID: PMC7302943 DOI: 10.1002/cti2.1144] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVES Malaria, caused by Plasmodium infection, remains a major global health problem. Monocytes are integral to the immune response, yet their transcriptional and functional responses in primary Plasmodium falciparum infection and in clinical malaria are poorly understood. METHODS The transcriptional and functional profiles of monocytes were examined in controlled human malaria infection with P. falciparum blood stages and in children and adults with acute malaria. Monocyte gene expression and functional phenotypes were examined by RNA sequencing and flow cytometry at peak infection and compared to pre-infection or at convalescence in acute malaria. RESULTS In subpatent primary infection, the monocyte transcriptional profile was dominated by an interferon (IFN) molecular signature. Pathways enriched included type I IFN signalling, innate immune response and cytokine-mediated signalling. Monocytes increased TNF and IL-12 production upon in vitro toll-like receptor stimulation and increased IL-10 production upon in vitro parasite restimulation. Longitudinal phenotypic analyses revealed sustained significant changes in the composition of monocytes following infection, with increased CD14+CD16- and decreased CD14-CD16+ subsets. In acute malaria, monocyte CD64/FcγRI expression was significantly increased in children and adults, while HLA-DR remained stable. Although children and adults showed a similar pattern of differentially expressed genes, the number and magnitude of gene expression change were greater in children. CONCLUSIONS Monocyte activation during subpatent malaria is driven by an IFN molecular signature with robust activation of genes enriched in pathogen detection, phagocytosis, antimicrobial activity and antigen presentation. The greater magnitude of transcriptional changes in children with acute malaria suggests monocyte phenotypes may change with age or exposure.
Collapse
Affiliation(s)
- Jessica R Loughland
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia,Menzies School of Health ResearchDarwinNTAustralia
| | - Tonia Woodberry
- Menzies School of Health ResearchDarwinNTAustralia,Charles Darwin UniversityDarwinNTAustralia,Present address:
The University of NewcastleCallaghanNSWAustralia
| | - Matt Field
- Australian Institute of Tropical Health and Medicine and Centre for Tropical Bioinformatics and Molecular BiologyJames Cook UniversityCairnsQLDAustralia,John Curtin School of Medical ResearchAustralian National UniversityCanberraACTAustralia
| | - Dean W Andrew
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Arya SheelaNair
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | | | - Kim A Piera
- Menzies School of Health ResearchDarwinNTAustralia,Charles Darwin UniversityDarwinNTAustralia
| | - Fiona H Amante
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Enny Kenangalem
- Timika Malaria Research ProgramPapuan Health and Community Development FoundationTimikaIndonesia,District Health AuthorityTimikaIndonesia
| | - Ric N Price
- Menzies School of Health ResearchDarwinNTAustralia,Charles Darwin UniversityDarwinNTAustralia,Centre for Tropical Medicine and Global HealthNuffield Department of Clinical MedicineUniversity of OxfordOxfordUK,Mahidol‐Oxford Tropical Medicine Research UnitFaculty of Tropical MedicineMahidol UniversityBangkokThailand
| | | | - Nicholas M Anstey
- Menzies School of Health ResearchDarwinNTAustralia,Charles Darwin UniversityDarwinNTAustralia
| | | | - Michelle J Boyle
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia,Menzies School of Health ResearchDarwinNTAustralia
| | - Gabriela Minigo
- Menzies School of Health ResearchDarwinNTAustralia,Charles Darwin UniversityDarwinNTAustralia,College of Health and Human SciencesCharles Darwin UniversityDarwinNTAustralia
| |
Collapse
|
22
|
Elmaagacli AH, Jehn C, Shikova Y, Huber M, Salwender H, Dahmash F, Singh A, Niggemann C, Vierbuchen M. Advanced systemic mastocytosis with strong expression of signaling lymphocyte activation marker family member 7 (SLAMF7) responsive to therapy with elotuzumab and lenalidomide. Leuk Lymphoma 2019; 61:485-487. [PMID: 31566043 DOI: 10.1080/10428194.2019.1668939] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Ahmet H Elmaagacli
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St. Georg, Hamburg, Germany
| | - Christian Jehn
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St. Georg, Hamburg, Germany
| | - Yana Shikova
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St. Georg, Hamburg, Germany
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Hans Salwender
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St. Georg, Hamburg, Germany
| | - Farouk Dahmash
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St. Georg, Hamburg, Germany
| | - Anju Singh
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St. Georg, Hamburg, Germany
| | | | | |
Collapse
|
23
|
Khalaf WS, Garg M, Mohamed YS, Stover CM, Browning MJ. In vitro Generation of Cytotoxic T Cells With Potential for Adoptive Tumor Immunotherapy of Multiple Myeloma. Front Immunol 2019; 10:1792. [PMID: 31428094 PMCID: PMC6687956 DOI: 10.3389/fimmu.2019.01792] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 07/16/2019] [Indexed: 12/25/2022] Open
Abstract
Multiple myeloma is a life-threatening hematological malignancy, which is rarely curable by conventional therapies. Immunotherapy, using tumor antigen-specific, cytotoxic T-lymphocytes, may represent an alternative or additional treatment for multiple myeloma. In this study, we used hybrid cell lines, generated by fusion of an EBV B-lymphoblastoid cell line (B-LCL) and myeloma cells, to stimulate in vitro peripheral blood lymphocytes (PBLs) from patients with multiple myeloma. We investigated induction of antigen-specific, cytotoxic T-lymphocytes to the well-defined tumor associated antigens (TAAs) hTERT, MUC1, MAGE-C1 and CS1, which have been shown to be expressed in a high proportion of cases of multiple myeloma. HLA-A2-peptide pentamer staining, interferon-γ and perforin ELISpot assays, as well as cytotoxicity assays were used. Following several rounds of in vitro stimulation, the hybrid cell lines induced antigen-specific, cytotoxic T-lymphocytes to four candidate TAAs in PBLs from HLA-A2+ multiple myeloma patients, using known HLA-A2 restricted peptide epitopes of the TAAs. In contrast, the HLA-A2+ myeloma cell line U266 failed to induce antigen-specific, cytotoxic T-lymphocytes in vitro. Our data indicate that B-LCL/myeloma hybrid cell lines induce antigen-specific, cytotoxic T-lymphocytes in PBLs isolated from multiple myeloma patients in vitro and may represent a novel strategy for use in adoptive immunotherapy of multiple myeloma.
Collapse
Affiliation(s)
- Wafaa S Khalaf
- Department of Infection, Immunity and Inflammation, Leicester University, Leicester, United Kingdom.,Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mamta Garg
- Department of Haematology, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| | - Yehia S Mohamed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt.,Department of Microbiology, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Cordula M Stover
- Department of Infection, Immunity and Inflammation, Leicester University, Leicester, United Kingdom
| | - Michael J Browning
- Department of Infection, Immunity and Inflammation, Leicester University, Leicester, United Kingdom.,Department of Immunology, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| |
Collapse
|
24
|
Elmaagacli AH, Salwender H, Jehn C, Dahmash F, Singh A, Wilson O, Pannenbeckers M, Niggemann C, Vierbuchen M. Strong expression of SLAMF7 in natural killer/T-cell lymphoma and large granular lymphocyte leukemia - a prominent biomarker and potential target for anti-SLAMF7 antibody therapy. Leuk Lymphoma 2019; 60:3335-3338. [PMID: 31164030 DOI: 10.1080/10428194.2019.1623887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Ahmet H Elmaagacli
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | - Hans Salwender
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | - Christian Jehn
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | - Farouk Dahmash
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | - Anju Singh
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | - Oliver Wilson
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | - Marc Pannenbeckers
- Department of Hematology/Oncology and Stem Cell Transplantation, Asklepios Klinik St.Georg, Hamburg, Germany
| | | | | |
Collapse
|
25
|
O'Connell P, Pepelyayeva Y, Blake MK, Hyslop S, Crawford RB, Rizzo MD, Pereira-Hicks C, Godbehere S, Dale L, Gulick P, Kaminski NE, Amalfitano A, Aldhamen YA. SLAMF7 Is a Critical Negative Regulator of IFN-α-Mediated CXCL10 Production in Chronic HIV Infection. THE JOURNAL OF IMMUNOLOGY 2018; 202:228-238. [PMID: 30530590 DOI: 10.4049/jimmunol.1800847] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/30/2018] [Indexed: 01/17/2023]
Abstract
Current advances in combined antiretroviral therapy have rendered HIV infection a chronic, manageable disease; however, the problem of persistent immune activation still remains despite treatment. The immune cell receptor SLAMF7 has been shown to be upregulated in diseases characterized by chronic immune activation. In this study, we studied the function of the SLAMF7 receptor in immune cells of HIV patients and the impacts of SLAMF7 signaling on peripheral immune activation. We observed increased frequencies of SLAMF7+ PBMCs in HIV+ individuals in a clinical phenotype-dependent manner, with discordant and long-term nonprogressor patients showing elevated SLAMF7 levels, and elite controllers showing levels comparable to healthy controls. We also noted that SLAMF7 was sensitive to IFN-⍺ stimulation, a factor elevated during HIV infection. Further studies revealed SLAMF7 to be a potent inhibitor of the monocyte-derived proinflammatory chemokine CXCL10 (IP-10) and other CXCR3 ligands, except in a subset of HIV+ patients termed SLAMF7 silent (SF7S). Studies utilizing small molecule inhibitors revealed that the mechanism of CXCL10 inhibition is independent of known SLAMF7 binding partners. Furthermore, we determined that SLAMF7 activation on monocytes is able to decrease their susceptibility to HIV-1 infection in vitro via downregulation of CCR5 and upregulation of the CCL3L1 chemokine. Finally, we discovered that neutrophils do not express SLAMF7, are CXCL10+ at baseline, are able to secrete CXCL10 in response to IFN-⍺ and LPS, and are nonresponsive to SLAMF7 signaling. These findings implicate the SLAMF7 receptor as an important regulator of IFN-⍺-driven innate immune responses during HIV infection.
Collapse
Affiliation(s)
- Patrick O'Connell
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824
| | - Yuliya Pepelyayeva
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824
| | - Maja K Blake
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824
| | - Sean Hyslop
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824
| | - Robert B Crawford
- Center for Integrative Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824
| | - Michael D Rizzo
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824
| | - Cristiane Pereira-Hicks
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824
| | - Sarah Godbehere
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824
| | - Linda Dale
- Department of Osteopathic Medical Specialties, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824
| | - Peter Gulick
- Department of Osteopathic Medical Specialties, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824
| | - Norbert E Kaminski
- Center for Integrative Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824.,Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824
| | - Andrea Amalfitano
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824.,Department of Pediatrics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824; and
| | - Yasser A Aldhamen
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824; .,College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824
| |
Collapse
|
26
|
Identification of Differentially Expressed Non-coding RNA in Porcine Alveolar Macrophages from Tongcheng and Large White Pigs Responded to PRRSV. Sci Rep 2018; 8:15621. [PMID: 30353051 PMCID: PMC6199292 DOI: 10.1038/s41598-018-33891-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 10/04/2018] [Indexed: 01/02/2023] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the most ruinous diseases in pig production. Our previous work showed that Tongcheng pigs (TC) were less susceptible to PRRS virus (PRRSV) than Large White (LW) pigs. To elucidate the difference in PRRSV resistance between the two breeds, small RNA-seq and ribo-zero RNA-seq were used to identify differentially expressed non-coding RNAs (including miRNAs and lincRNAs) responded to PRRSV in porcine alveolar macrophages (PAMs) from TC and LW pigs. Totally, 250 known mature miRNAs were detected. For LW pigs, there were 44 down-regulated and 67 up-regulated miRNAs in infection group; while for TC pigs, 12 down-regulated and 23 up-regulated miRNAs in TC infection group were identified. The target genes of the common differentially expressed miRNAs (DEmiRNAs) in these two breeds were enriched in immune-related processes, including apoptosis process, inflammatory response, T cell receptor signaling pathway and so on. In addition, 5 shared DEmiRNAs (miR-181, miR-1343, miR-296-3p, miR-199a-3p and miR-34c) were predicted to target PRRSV receptors, of which miR-199a-3p was validated to inhibit the expression of CD151. Interestingly, miR-378 and miR-10a-5p, which could inhibit PRRSV replication, displayed higher expression level in TC control group than that in LW control group. Contrarily, miR-145-5p and miR-328, which were specifically down-regulated in LW pigs, could target inhibitory immunoreceptors and may involve in immunosuppression caused by PRRSV. This indicates that DEmiRNAs are involved in the regulation of the immunosuppression and immune escape of the two breeds. Furthermore, we identified 616 lincRNA transcripts, of which 48 and 30 lincRNAs were differentially expressed in LW and TC pigs, respectively. LincRNA TCONS_00125566 may play an important role in the entire regulatory network, and was predicted to regulate the expression of immune-related genes through binding with miR-1343 competitively. In conclusion, this study provides an important resource for further revealing the interaction between host and virus, which will specify a new direction for anti-PRRSV research.
Collapse
|
27
|
Malaer JD, Marrufo AM, Mathew PA. 2B4 (CD244, SLAMF4) and CS1 (CD319, SLAMF7) in systemic lupus erythematosus and cancer. Clin Immunol 2018; 204:50-56. [PMID: 30347240 DOI: 10.1016/j.clim.2018.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 10/17/2018] [Accepted: 10/18/2018] [Indexed: 02/08/2023]
Abstract
Signaling Lymphocyte Activation Molecule (SLAM) family receptors are expressed on different types of hematopoietic cells and play important role in immune regulation in health and disease. 2B4 (CD244, SLAMF4) and CS1 (CD319, CRACC, SLAMF7) were originally identified as NK cell receptors regulating NK cell cytolytic activity. 2B4 is expressed on all NK cells, a subpopulation of T cells, monocytes and basophils. Unlike other activating and inhibitory receptors, 2B4 (CD244) interaction with its ligand CD48 has been shown to mediate both activating and inhibitory functions. Defective signaling via 2B4 due to mutations in signaling adaptor SAP contributes to X-linked lymphoproliferative Disease (XLP). Expression of 2B4 and CS1 are altered in systemic lupus erythematosus (SLE). CS1 is overexpressed in multiple myeloma (MM) and anti-CS1 mab (Elotuzumab/Empliciti) has been approved by FDA as a breakthrough drug for treatment for MM patients. CAR -T cells or CAR- NK cells containing full length CS1 or the signaling domain of 2B4 with TCR-ζ have shown promising results to treat cancer and autoimmune diseases.
Collapse
Affiliation(s)
- Joseph D Malaer
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Armando M Marrufo
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Porunelloor A Mathew
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| |
Collapse
|
28
|
Jacometo C, Alharthi A, Zhou Z, Luchini D, Loor J. Maternal supply of methionine during late pregnancy is associated with changes in immune function and abundance of microRNA and mRNA in Holstein calf polymorphonuclear leukocytes. J Dairy Sci 2018; 101:8146-8158. [DOI: 10.3168/jds.2018-14428] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/02/2018] [Indexed: 12/12/2022]
|
29
|
SLAMF7-CAR T cells eliminate myeloma and confer selective fratricide of SLAMF7 + normal lymphocytes. Blood 2017; 130:2838-2847. [PMID: 29089311 DOI: 10.1182/blood-2017-04-778423] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 10/24/2017] [Indexed: 12/19/2022] Open
Abstract
SLAMF7 is under intense investigation as a target for immunotherapy in multiple myeloma. In this study, we redirected the specificity of T cells to SLAMF7 through expression of a chimeric antigen receptor (CAR) derived from the huLuc63 antibody (elotuzumab) and demonstrate that SLAMF7-CAR T cells prepared from patients and healthy donors confer potent antimyeloma reactivity. We confirmed uniform, high-level expression of SLAMF7 on malignant plasma cells in previously untreated and in relapsed/refractory (R/R) myeloma patients who had received previous treatment with proteasome inhibitors and immunomodulatory drugs. Consequently, SLAMF7-CAR T cells conferred rapid cytolysis of previously untreated and R/R primary myeloma cells in vitro. In addition, a single administration of SLAMF7-CAR T cells led to resolution of medullary and extramedullary myeloma manifestations in a murine xenograft model in vivo. SLAMF7 is expressed on a fraction of normal lymphocytes, including subsets of natural killer (NK) cells, T cells, and B cells. After modification with the SLAMF7-CAR, both CD8+ and CD4+ T cells rapidly acquired and maintained a SLAMF7- phenotype and could be readily expanded to therapeutically relevant cell doses. We analyzed the recognition of normal lymphocytes by SLAMF7-CAR T cells and show that they induce selective fratricide of SLAMF7+/high NK cells, CD4+ and CD8+ T cells, and B cells. Importantly, however, the fratricide conferred by SLAMF7-CAR T cells spares the SLAMF7-/low fraction in each cell subset and preserves functional lymphocytes, including virus-specific T cells. In aggregate, our data illustrate the potential use of SLAMF7-CAR T-cell therapy as an effective treatment against multiple myeloma and provide novel insights into the consequences of targeting SLAMF7 for the normal lymphocyte compartment.
Collapse
|
30
|
Costa ML, Robinette ML, Bugatti M, Longtine MS, Colvin BN, Lantelme E, Vermi W, Colonna M, Nelson DM, Cella M. Two Distinct Myeloid Subsets at the Term Human Fetal-Maternal Interface. Front Immunol 2017; 8:1357. [PMID: 29123519 PMCID: PMC5662895 DOI: 10.3389/fimmu.2017.01357] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 10/03/2017] [Indexed: 12/13/2022] Open
Abstract
During pregnancy, immune cells infiltrate the placenta at different stages of fetal development. NK cells and macrophages are the most predominant cell types. These immune cells play pleiotropic roles, as they control spiral artery remodeling to ensure appropriate blood supply and maintain long-term tolerance to a true allograft; yet, they must be able to mount appropriate immune defenses to pathogens that may threaten the fetus. Whether the same cell type accomplishes all these tasks or if there are dedicated subsets remains controversial. Here, we identify and characterize two distinct subsets of myeloid cells that differ in their pro-inflammatory/regulatory capacity. While one subset predominantly produces the immune-modulating cytokine IL-10, the second subset has superior capacity to secrete pro-inflammatory mediators, such as IL-1β and IL-6. The putative regulatory myeloid cells also express high levels of inhibitory receptors and their ligands, including programmed cell death 1 (PD1) ligands. Importantly, a large fraction of CD8 and CD4 cells in normal term human placenta are PD1 positive, suggesting that the PD1/PD1 ligands axis might be critical to maintain tolerance during pregnancy.
Collapse
Affiliation(s)
- Maria Laura Costa
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO, United States.,Department of Obstetrics and Gynecology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Michelle L Robinette
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, United States
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy
| | - Mark S Longtine
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO, United States
| | - Bryanne N Colvin
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO, United States.,Department of Pediatrics, Washington University School of Medicine, St Louis, MO, United States
| | - Erica Lantelme
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, United States
| | - William Vermi
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, United States.,Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, United States
| | - D Michael Nelson
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO, United States
| | - Marina Cella
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, United States
| |
Collapse
|
31
|
Radhakrishnan SV, Bhardwaj N, Steinbach M, Weidner J, Luetkens T, Atanackovic D. Elotuzumab as a novel anti-myeloma immunotherapy. Hum Vaccin Immunother 2017; 13:1751-1757. [PMID: 28604269 DOI: 10.1080/21645515.2017.1327487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Treatment of multiple myeloma has undergone significant change in the last decade with the introduction of new immunomodulatory agents, proteasome inhibitors, and immunotherapeutic approaches. Elotuzumab is a humanized monoclonal antibody targeting CS1, which is a member of the SLAM (Signaling Lymphocyte Activation Molecule) family of proteins, expressed on the surface of myeloma plasma cells. Here we review the preclinical investigations that led to the development of elotuzumab and the clinical studies that resulted in its approval for the treatment of relapsed/refractory multiple myeloma. Although preclinical data looked very promising, elotuzumab monotherapy did not result in objective clinical responses in patients with relapsed/refractory multiple myeloma. However, combination treatment with immunomodulators and proteasome inhibitors resulted in substantial clinical activity in relapsed/refractory MM. Currently, there are several clinical trials ongoing investigating the role of elotuzumab in newly diagnosed myeloma patients and in patients receiving maintenance therapy.
Collapse
Affiliation(s)
- Sabarinath Venniyil Radhakrishnan
- a Multiple Myeloma Program & Cancer Immunotherapy , Hematology and Hematologic Malignancies, University of Utah / Huntsman Cancer Institute , Salt Lake City , UT , USA
| | - Neelam Bhardwaj
- a Multiple Myeloma Program & Cancer Immunotherapy , Hematology and Hematologic Malignancies, University of Utah / Huntsman Cancer Institute , Salt Lake City , UT , USA
| | - Mary Steinbach
- a Multiple Myeloma Program & Cancer Immunotherapy , Hematology and Hematologic Malignancies, University of Utah / Huntsman Cancer Institute , Salt Lake City , UT , USA
| | - Janet Weidner
- a Multiple Myeloma Program & Cancer Immunotherapy , Hematology and Hematologic Malignancies, University of Utah / Huntsman Cancer Institute , Salt Lake City , UT , USA
| | - Tim Luetkens
- a Multiple Myeloma Program & Cancer Immunotherapy , Hematology and Hematologic Malignancies, University of Utah / Huntsman Cancer Institute , Salt Lake City , UT , USA
| | - Djordje Atanackovic
- a Multiple Myeloma Program & Cancer Immunotherapy , Hematology and Hematologic Malignancies, University of Utah / Huntsman Cancer Institute , Salt Lake City , UT , USA
| |
Collapse
|
32
|
Cai Q, Tu M, Xu-Monette ZY, Sun R, Manyam GC, Xu X, Tzankov A, Hsi ED, Møller MB, Medeiros LJ, Ok CY, Young KH. NF-κB p50 activation associated with immune dysregulation confers poorer survival for diffuse large B-cell lymphoma patients with wild-type p53. Mod Pathol 2017; 30:854-876. [PMID: 28281555 DOI: 10.1038/modpathol.2017.5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/29/2016] [Accepted: 12/29/2016] [Indexed: 12/12/2022]
Abstract
Dysregulated NF-κB signaling is critical for lymphomagenesis, however, the expression and clinical relevance of NF-κB subunit p50 in diffuse large B-cell lymphoma have not been evaluated. In this study, we analyzed the prognostic significance and gene expression signatures of p50 nuclear expression as a surrogate for p50 activation in 465 patients with de novo diffuse large B-cell lymphoma. We found that p50+ nuclear expression, observed in 34.6% of diffuse large B-cell lymphoma, predominantly composed of activated B-cell-like subtype, was an independent adverse prognostic factor in patients with activated B-cell-like diffuse large B-cell lymphoma. It was also an adverse prognostic factor in patients with wild-type TP53 independent of the activated B-cell-like and germinal center B-cell-like subtypes, even though p50 activation correlated with significantly lower levels of Myc, PI3K, phospho-AKT, and CXCR4 expression and less frequent BCL2 translocations. In contrast, in germinal center B-cell-like diffuse large B-cell lymphoma patients with TP53 mutations, p50+ nuclear expression correlated with significantly better clinical outcomes, and decreased p53, Bcl-2, and Myc expression. Gene expression profiling revealed multiple signaling pathways potentially upstream the p50 activation through either canonical or noncanonical NF-κB pathways, and suggested that immune suppression, including that by the immune checkpoint TIM-3 and that through leukocyte immunoglobulin-like receptors, but not antiapoptosis and proliferation, may underlie the observed poorer survival rates associated with p50+ nuclear expression in diffuse large B-cell lymphoma. In conclusion, these data show that p50 is important as a unique mechanism of R-CHOP-resistance in activated B-cell-like diffuse large B-cell lymphoma and in patients without TP53 mutations. The results also provide insights into the regulation and function of p50 in diffuse large B-cell lymphoma and its cross talk with the p53 pathway with important therapeutic implications.
Collapse
MESH Headings
- Antibodies, Monoclonal, Murine-Derived/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Cell Nucleus/chemistry
- Cyclophosphamide/therapeutic use
- Doxorubicin/therapeutic use
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Kaplan-Meier Estimate
- Lymphoma, Large B-Cell, Diffuse/chemistry
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/immunology
- Male
- Middle Aged
- Multivariate Analysis
- Mutation
- NF-kappa B p50 Subunit/analysis
- NF-kappa B p50 Subunit/genetics
- Prednisone/therapeutic use
- Rituximab
- Time Factors
- Transcriptome
- Treatment Outcome
- Tumor Suppressor Protein p53/genetics
- Vincristine/therapeutic use
Collapse
Affiliation(s)
- Qingqing Cai
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meifeng Tu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital &Institute, Beijing, China
| | - Zijun Y Xu-Monette
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ruifang Sun
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ganiraju C Manyam
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaolu Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | | | - Eric D Hsi
- Department of Pathology, Cleveland Clinic, Cleveland, OH, USA
| | - Michael B Møller
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chi Young Ok
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ken H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas School of Medicine, Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
33
|
Acute effects of focused ultrasound-induced increases in blood-brain barrier permeability on rat microvascular transcriptome. Sci Rep 2017; 7:45657. [PMID: 28374753 PMCID: PMC5379491 DOI: 10.1038/srep45657] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/02/2017] [Indexed: 02/07/2023] Open
Abstract
Therapeutic treatment options for central nervous system diseases are greatly limited by the blood-brain barrier (BBB). Focused ultrasound (FUS), in conjunction with circulating microbubbles, can be used to induce a targeted and transient increase in BBB permeability, providing a unique approach for the delivery of drugs from the systemic circulation into the brain. While preclinical research has demonstrated the utility of FUS, there remains a large gap in our knowledge regarding the impact of sonication on BBB gene expression. This work is focused on investigating the transcriptional changes in dorsal hippocampal rat microvessels in the acute stages following sonication. Microarray analysis of microvessels was performed at 6 and 24 hrs post-FUS. Expression changes in individual genes and bioinformatic analysis suggests that FUS may induce a transient inflammatory response in microvessels. Increased transcription of proinflammatory cytokine genes appears to be short-lived, largely returning to baseline by 24 hrs. This observation may help to explain some previously observed bioeffects of FUS and may also be a driving force for the angiogenic processes and reduced drug efflux suggested by this work. While further studies are necessary, these results open up intriguing possibilities for novel FUS applications and suggest possible routes for pharmacologically modifying the technique.
Collapse
|
34
|
CRACC-targeting Fc-fusion protein induces activation of NK cells and DCs and improves T cell immune responses to antigenic targets. Vaccine 2016; 34:3109-3118. [PMID: 27151882 DOI: 10.1016/j.vaccine.2016.04.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 04/13/2016] [Accepted: 04/23/2016] [Indexed: 11/20/2022]
Abstract
The CD2-like receptor activating cytotoxic cell (CRACC) receptor is a member of the SLAM family of receptors that are found on several types of immune cells. We previously demonstrated that increasing the abundance of the adaptor protein EAT-2 during vaccination enhanced innate and adaptive immune responses to vaccine antigens. Engagement of the CRACC receptor in the presence of the EAT-2 adaptor generally results in immune cell activation, while activating CRACC signaling in cells that lack EAT-2 adaptor inhibits their effector and regulatory functions. As EAT-2 is the only SAP adaptor that interacts with the CRACC receptor, we hypothesized that technologies that specifically modulate CRACC signaling during vaccination may also improve antigen specific adaptive immune responses. To test this hypothesis, we constructed a CRACC-targeting Fc fusion protein and included it in vaccination attempts. Indeed, mice co-vaccinated with the CRACC-Fc fusion protein and an adenovirus vaccine expressing the HIV-Gag protein had improved Gag-specific T cell responses, as compared to control mice. These responses are characterized by increased numbers of Gag-specific tetramer+ CD8+ T cells and increases in production of IFNγ, TNFα, and IL2, by Gag-specific CD8+ T cells. Moreover, our results revealed that use of the CRACC-Fc fusion protein enhances vaccine-elicited innate immune responses, as characterized by increased dendritic cells (DCs) maturation and IFNγ production from NK cells. This study highlights the importance of CRACC signaling during the induction of an immune response generally, and during vaccinations specifically, and also lends insight into the mechanisms underlying our prior results noting EAT-2-dependent improvements in vaccine efficacy.
Collapse
|
35
|
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: 23] [Impact Index Per Article: 2.3] [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
|
36
|
Transcriptome analysis highlights the conserved difference between embryonic and postnatal-derived alveolar macrophages. Blood 2015; 126:1357-66. [PMID: 26232173 DOI: 10.1182/blood-2015-01-624809] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 07/27/2015] [Indexed: 12/22/2022] Open
Abstract
Alveolar macrophages (AMs) reside on the luminal surfaces of the airways and alveoli where they maintain host defense and promote alveolar homeostasis by ingesting inhaled particulates and regulating inflammatory responses. Recent studies have demonstrated that AMs populate the lungs during embryogenesis and self-renew throughout life with minimal replacement by circulating monocytes, except under extreme conditions of depletion or radiation injury. Here we demonstrate that on a global scale, environment appears to dictate AM development and function. Indeed, transcriptome analysis of embryonic host-derived and postnatal donor-derived AMs coexisting within the same mouse demonstrated >98% correlation and overall functional analyses were similar. However, we also identified several genes whose expression was dictated by origin rather than environment. The most differentially expressed gene not altered by environment was Marco, a gene recently demonstrated to have enhancer activity in embryonic-derived but not postnatal-derived tissue macrophages. Overall, we show that under homeostatic conditions, the environment largely dictates the programming and function of AMs, whereas the expression of a small number of genes remains linked to the origin of the cell.
Collapse
|
37
|
Filsoof DM, Safford RE, Newby K, Rosenberg S, Kontras DG, Baker A, Odunukan OW, Fletcher G. Impact of exercise stress testing on diagnostic gene expression in patients with obstructive and nonobstructive coronary artery disease. Am J Cardiol 2015; 115:1346-50. [PMID: 25776454 DOI: 10.1016/j.amjcard.2015.02.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 02/10/2015] [Accepted: 02/10/2015] [Indexed: 11/30/2022]
Abstract
A blood-based gene expression test can diagnose obstructive coronary artery disease (CAD). The test is sensitive to inflammatory and immune processes associated with atherosclerosis. Acute exercise engages short-term inflammatory pathways, and exercise stress testing may affect results of gene expression testing during the same diagnostic workup. The objective of this study was to evaluate the effect of exercise on diagnostic gene expression testing. Ten patients with obstructive CAD (≥50% stenosis) and 10 with no/minimal CAD (≤20% stenosis) were identified by angiography. Blood samples for gene expression were obtained at baseline, peak exercise, 30 to 60 minutes after testing, and 24 to 36 hours after testing. Core-lab gene expression analysis yielded raw gene expression scores (GES) for each time point. Linear models were used to estimate changes in GES, adjusting for CAD status and other covariates. GES increased during peak exercise across both genders, with no significant differences as a function of CAD status. The overall adjusted mean GES increase at peak exercise was 0.29 (95% confidence interval 0.22 to 0.36; p <0.001). GES after exercise were not significantly different from baseline. The change in gene expression levels during peak exercise may reflect a transient inflammatory response to acute exercise that may be independent of patient gender or CAD status. In conclusion, CAD GES increase at peak exercise testing and rapidly return to baseline. Such may reflect a transient inflammatory response to acute exercise independent of gender or extent of CAD.
Collapse
Affiliation(s)
- David M Filsoof
- Division of Cardiovascular Diseases at Mayo Clinic, Jacksonville, Florida
| | - Robert E Safford
- Division of Cardiovascular Diseases at Mayo Clinic, Jacksonville, Florida
| | - Kristin Newby
- Division of Cardiology, Duke University, Durham, North Carolina
| | | | - Dana G Kontras
- Division of Cardiovascular Diseases at Mayo Clinic, Jacksonville, Florida
| | | | | | - Gerald Fletcher
- Division of Cardiovascular Diseases at Mayo Clinic, Jacksonville, Florida.
| |
Collapse
|
38
|
Christensen LB, Woods TA, Carmody AB, Caughey B, Peterson KE. Age-related differences in neuroinflammatory responses associated with a distinct profile of regulatory markers on neonatal microglia. J Neuroinflammation 2014; 11:70. [PMID: 24708744 PMCID: PMC4234188 DOI: 10.1186/1742-2094-11-70] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 03/21/2014] [Indexed: 02/04/2023] Open
Abstract
Background The perinatal period is one in which the mammalian brain is particularly vulnerable to immune-mediated damage. Early inflammation in the central nervous system (CNS) is linked with long-term impairment in learning and behavior, necessitating a better understanding of mediators of neuroinflammation. We therefore directly examined how age affected neuroinflammatory responses to pathogenic stimuli. Methods In mice, susceptibility to neurological damage changes dramatically during the first few weeks of life. Accordingly, we compared neuroinflammatory responses to pathogen associated molecular patterns (PAMPs) of neonatal (two day-old) and weanling (21 day-old) mice. Mice were inoculated intracerebrally with PAMPs and the cellular and molecular changes in the neuroinflammatory response were examined. Results Of the 12 cytokines detected in the CNS following toll-like receptor 4 (TLR4) stimulation, ten were significantly higher in neonates compared with weanling mice. A similar pattern of increased cytokines in neonates was also observed with TLR9 stimulation. Analysis of cellular responses indicated a difference in microglial activation markers in the CNS of neonatal mice and increased expression of proteins known to modulate cellular activation including CD11a, F4/80 and CD172a. We also identified a new marker on microglia, SLAMF7, which was expressed at higher levels in neonates compared with weanlings. Conclusions A unique neuroinflammatory profile, including higher expression of several proinflammatory cytokines and differential expression of microglial markers, was observed in brain tissue from neonates following TLR stimulation. This increased neuroinflammatory response to PAMPs may explain why the developing brain is particularly sensitive to infection and why infection or stress during this time can lead to long-term damage in the CNS.
Collapse
Affiliation(s)
| | | | | | | | - Karin E Peterson
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, Canada.
| |
Collapse
|
39
|
Aldhamen YA, Seregin SS, Aylsworth CF, Godbehere S, Amalfitano A. Manipulation of EAT-2 expression promotes induction of multiple beneficial regulatory and effector functions of the human innate immune system as a novel immunomodulatory strategy. Int Immunol 2013; 26:291-303. [PMID: 24374770 DOI: 10.1093/intimm/dxt061] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The signaling lymphocytic activation molecule (SLAM) receptor-associated adaptor Ewing's sarcoma-associated transcript-2 (EAT-2) is primarily expressed in innate immune cells including dendritic cells (DCs), macrophages and NK cells. A recent human HIV vaccine study confirmed that EAT-2 expression was associated with the enhanced immunogenicity induced by the MRKAd5/HIV vaccine. We previously harnessed the capability of EAT-2 to modulate signaling mediated by SLAM receptors and demonstrated that by incorporating EAT-2 expression into vaccines, one could enhance innate and adaptive immune responses in mice, even in the face of pre-existing immunity to the vaccine vectors. Herein, we investigated the innate immune responses of human cells exposed to EAT-2-over-expressing vaccines. Our results demonstrate that EAT-2 over-expression can significantly alter the kinetics of critical pro-inflammatory cytokine and chemokine responses elaborated by human PBMCs. In addition, enhanced DC maturation and increased monocyte phagocytosis were observed in EAT-2-transduced human cells. We also found that EAT-2 over-expression improved antigen presentation by human cells. Moreover, EAT-2 over-expression increased the anti-tumor activity of human NK cells against K562 tumor cell targets. Many of these responses were extinguished with use of an EAT-2 variant carrying a mutant SH2 domain (R31Q), suggesting a critical role for the interaction between EAT-2 and SLAM receptors in mediating these responses. In conclusion, these results provide evidence that EAT-2 interacts with key components of multiple arms of the human innate immune system, and that this role highlights the potential for targeting EAT-2 functions so as to improve a number of human immunotherapeutic approaches, including vaccine development.
Collapse
Affiliation(s)
- Yasser A Aldhamen
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | | | | | | | | |
Collapse
|
40
|
Hagberg N, Theorell J, Schlums H, Eloranta ML, Bryceson YT, Rönnblom L. Systemic lupus erythematosus immune complexes increase the expression of SLAM family members CD319 (CRACC) and CD229 (LY-9) on plasmacytoid dendritic cells and CD319 on CD56(dim) NK cells. THE JOURNAL OF IMMUNOLOGY 2013; 191:2989-98. [PMID: 23956418 DOI: 10.4049/jimmunol.1301022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Patients with systemic lupus erythematosus (SLE) display an activated type I IFN system due to unceasing IFN-α release from plasmacytoid dendritic cells (pDCs) stimulated by nucleic acid-containing immune complexes (ICs). NK cells strongly promote the IFN-α production by pDCs; therefore, we investigated surface molecules that could be involved in the pDC-NK cell cross-talk. In human PBMCs stimulated with RNA-containing ICs (RNA-ICs), the expression of the signaling lymphocyte activation molecule (SLAM) family receptors CD319 and CD229 on pDCs and CD319 on CD56(dim) NK cells was selectively increased. Upregulation of CD319 and CD229 on RNA-IC-stimulated pDCs was induced by NK cells or cytokines (e.g., GM-CSF, IL-3). IFN-α-producing pDCs displayed a higher expression of SLAM molecules compared with IFN-α⁻ pDCs. With regard to signaling downstream of SLAM receptors, pDCs expressed SHIP-1, SHP-1, SHP-2, and CSK but lacked SLAM-associated protein (SAP) and Ewing's sarcoma-activated transcript 2 (EAT2), indicating that these receptors may act as inhibitory receptors on pDCs. Furthermore, pDCs from patients with SLE had decreased expression of CD319 on pDCs and CD229 on CD56(dim) NK cells, but RNA-IC stimulation increased CD319 and CD229 expression. In conclusion, this study reveals that the expression of the SLAM receptors CD319 and CD229 is regulated on pDCs and NK cells by lupus ICs and that the expression of these receptors is specifically altered in SLE. These results, together with the observed genetic association between the SLAM locus and SLE, suggest a role for CD319 and CD229 in the SLE disease process.
Collapse
Affiliation(s)
- Niklas Hagberg
- Section of Rheumatology, Department of Medical Sciences, Uppsala University, S-751 85 Uppsala, Sweden.
| | | | | | | | | | | |
Collapse
|