1
|
Zhang CX, Mao YY, Tan YP, Zhang MY, Shao K, Wang SJ, Ji P, Wang JY, Yin L, Wang Y. Enhancement of CD8 +T cell cytotoxicity activity by IFN-α implies alternative pathologic role in systemic lupus erythematosus. J Transl Autoimmun 2025; 10:100276. [PMID: 39995790 PMCID: PMC11849641 DOI: 10.1016/j.jtauto.2025.100276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/15/2025] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
Objective Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease which is affected by the environmental, genetic factors as well as the immune system. Previous reports have implicated IFN-α in the pathogenesis of SLE. Up to date, however, no research has ever investigated the effect of IFN-α on CD8+T cells, which might be implicated in the pathogenesis of SLE. In the present study, we aimed to explore the pathologic role of IFN-α in regard to dysfunction of CD8+T cells in SLE. Methods Serum level of IFN-α was detected in SLE and healthy controls (HC). Surface expression of lysosome-associated membrane protein 1 (LAMP-1; CD107a) and secretion of granzyme B of CD8+T cells was measured in SLE and HC with or without IFN-α co-stimulation/PI3K inhibitor. Results Our results demonstrated that there was increased surface expression of CD107a of CD8+T cells in SLE patients compared with healthy controls (HC), indicating enhanced cytotoxicity of CD8+T cells in SLE patients. Meanwhile, increased secretion of granzyme B was also detected in CD8+T cells of SLE compared with HC, which correlated with the disease activity (SLEDAI). Furthermore, elevated serum level of IFN-α in SLE was confirmed in our study. In vitro study, granzyme B secretion by CD8+T cells was upregulated upon IFN-α costimulation, which was consistent with enhanced cytotoxicity of CD8+T cells upon IFN-α costimulation, as reflected by elevated surface expression of CD107a. PI3K inhibitor reversed increased granzyme B synthesis upon IFN-α costimulation in a dose-dependent manner. Conclusion In summary, elevated serum level of IFN-α was responsible for increased secretion of granzyme B and enhanced cytotoxicity of CD8+T cells in SLE and this process may be related to PI3K pathway. Relevant molecules and mechanism remains to be explored in the future.
Collapse
Affiliation(s)
- Chen-xing Zhang
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - You-ying Mao
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
- Department of Pediatrics, Songjiang Hospital Affiliated to the Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu-pin Tan
- Department of Pediatrics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, China
| | - Mei-yu Zhang
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Kang Shao
- Department of Laboratory, Renji Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Shu-jun Wang
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Ping Ji
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Jia-yuan Wang
- Department of Laboratory, Renji Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Lei Yin
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Ying Wang
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| |
Collapse
|
2
|
Hugaboom MB, Wirth LV, Street K, Ruthen N, Jegede OA, Schindler NR, Shah V, Zaemes JP, El Ahmar N, Matar S, Savla V, Choueiri TK, Denize T, West DJ, McDermott DF, Plimack ER, Sosman JA, Haas NB, Stein MN, Alter R, Bilen MA, Hurwitz ME, Hammers H, Signoretti S, Atkins MB, Wu CJ, Braun DA. Presence of Tertiary Lymphoid Structures and Exhausted Tissue-Resident T Cells Determines Clinical Response to PD-1 Blockade in Renal Cell Carcinoma. Cancer Discov 2025; 15:948-968. [PMID: 39992403 DOI: 10.1158/2159-8290.cd-24-0991] [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: 07/12/2024] [Revised: 01/08/2025] [Accepted: 02/21/2025] [Indexed: 02/25/2025]
Abstract
SIGNIFICANCE We describe a paradigm wherein combined high TLS and low tissue-resident exhausted CD8+ T cells are required for optimal response to PD-1 blockade in RCC. This analysis identifies key determinants of response to PD-1 blockade in advanced RCC and suggests avenues for future immune modulation through rational combination therapy strategies.
Collapse
Affiliation(s)
- Miya B Hugaboom
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Lena V Wirth
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Kelly Street
- Division of Biostatistics, Keck School of Medicine of USC, Los Angeles, California
| | - Neil Ruthen
- Weill Cornell Graduate School of Medical Sciences, New York, New York
| | - Opeyemi A Jegede
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Valisha Shah
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jacob P Zaemes
- Georgetown Lombardi Comprehensive Cancer Center, Washington, District of Columbia
| | - Nourhan El Ahmar
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sayed Matar
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Varunika Savla
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Thomas Denize
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Destiny J West
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - David F McDermott
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | - Jeffrey A Sosman
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois
| | - Naomi B Haas
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mark N Stein
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
| | - Robert Alter
- Hackensack University Medical Center, Hackensack, New Jersey
| | - Mehmet A Bilen
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Michael E Hurwitz
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Hans Hammers
- Division of Hematology and Oncology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Michael B Atkins
- Georgetown Lombardi Comprehensive Cancer Center, Washington, District of Columbia
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David A Braun
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
3
|
Harirah HAA, Mohammed MH, Basha SAZ, Uthirapathy S, Ganesan S, Shankhyan A, Sharma GC, Devi A, Kadhim AJ, S NH. Targeting EZH2 in autoimmune diseases: unraveling epigenetic regulation and therapeutic potential. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04127-6. [PMID: 40198399 DOI: 10.1007/s00210-025-04127-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/29/2025] [Indexed: 04/10/2025]
Abstract
Approximately 8-10% of the global population is affected by autoimmune diseases (ADs), which encompass a wide array of idiopathic conditions resulting from dysregulated immune responses. The enzymatic component of the polycomb-repressive complex 2 (PRC2), enhancer of zeste homolog 2 (EZH2, also referred to as KMT6), functions as a methyltransferase possessing a SET domain that plays crucial roles in epigenetic regulation, explicitly facilitating the methylation of histone H3 at lysine 27. Notably, EZH2 is catalytically inactive and requires association with EED and SUZ12 to form an active PRC2 complex. Hyperactivation of EZH2 has been implicated in various malignancies, prompting the development of EZH2 inhibitors as therapeutic agents for several cancers, including lymphoma, prostate, breast, and colon cancer. The application of EZH2-targeting therapies has also been explored in the context of autoimmune diseases. While there have been advancements in certain ADs, responses can vary significantly, as evidenced by mixed outcomes in cases such as inflammatory bowel disease. Consequently, the dual role of EZH2 and the therapeutic potential of its inhibitors in the treatment of ADs remain nascent fields of study. This review will elucidate the interplay between EZH2 and autoimmune diseases, highlighting emerging insights and therapeutic avenues.
Collapse
Affiliation(s)
- Hashem Ahmed Abu Harirah
- Medical Laboratory Department, Faculty of Allied Medical Sciences, Zarqa University, Zarqa, Jordan.
| | - Mohammed Hashim Mohammed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, Al-Maarif University, Anbar, Iraq.
| | - Sami Ahmed Zaher Basha
- Physical Therapy Department, Faculty of Allied Medical Sciences, Zarqa University, Zarqa, Jordan
- Department of Cardiovascular Pulmonary and Geriatrics, Faculty of Physical Therapy, Pharos University, Alexandria, Egypt
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Kurdistan Region, Erbil, Iraq
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Aman Shankhyan
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges Jhanjeri, Mohali, 140307, Punjab, India
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Naher H S
- Laboratories Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| |
Collapse
|
4
|
Lingel H, Fischer L, Remstedt S, Kuropka B, Philipsen L, Han I, Sander JE, Freund C, Arra A, Brunner-Weinzierl MC. SLAMF7 (CD319) on activated CD8 + T cells transduces environmental cues to initiate cytotoxic effector cell responses. Cell Death Differ 2025; 32:561-572. [PMID: 39390117 PMCID: PMC11893764 DOI: 10.1038/s41418-024-01399-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 09/10/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024] Open
Abstract
CD8+ T-cell responses are meticulously orchestrated processes regulated by intercellular receptor:ligand interactions. These interactions critically control the dynamics of CD8+ T-cell populations that is crucial to overcome threats such as viral infections or cancer. Yet, the mechanisms governing these dynamics remain incompletely elucidated. Here, we identified a hitherto unknown T-cell referred function of the self-ligating surface receptor SLAMF7 (CD319) on CD8+ T cells during initiation of cytotoxic T-cell responses. According to its cytotoxicity related expression on T effector cells, we found that CD8+ T cells could utilize SLAMF7 to transduce environmental cues into cellular interactions and information exchange. Indeed, SLAMF7 facilitated a dose-dependent formation of stable homotypic contacts that ultimately resulted in stable cell-contacts, quorum populations and commitment to expansion and differentiation. Using pull-down assays and network analyses, we identified novel SLAMF7-binding intracellular signaling molecules including the CRK, CRKL, and Nck adaptors, which are involved in T-cell contact formation and may mediate SLAMF7 functions in sensing and adhesion. Hence, providing SLAMF7 signals during antigen recognition of CD8+ T cells enhanced their overall magnitude, particularly in responses towards low-affinity antigens, resulting in a significant boost in their proliferation and cytotoxic capacity. Overall, we have identified and characterized a potent initiator of the cytotoxic T lymphocyte response program and revealed advanced mechanisms to improve CD8+ T-cell response decisions against weak viral or tumor-associated antigens, thereby strengthening our defense against such adversaries.
Collapse
Affiliation(s)
- Holger Lingel
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Laura Fischer
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Sven Remstedt
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Benno Kuropka
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Lars Philipsen
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
- Multi-parametric bioimaging and cytometry (MPBIC) core facility, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Institute of Cellular and Molecular Immunology, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Irina Han
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Jan-Erik Sander
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Christian Freund
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Aditya Arra
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany
| | - Monika C Brunner-Weinzierl
- Department of Experimental Paediatrics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany.
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke-University, Magdeburg, Germany.
| |
Collapse
|
5
|
Lupu A, Stoleriu G, Nedelcu AH, Perju SN, Gavrilovici C, Baciu G, Mihai CM, Chisnoiu T, Morariu ID, Grigore E, Shawais SK, Salaru DL, Revenco N, Lupu VV. Overview of Oxidative Stress in Systemic Lupus Erythematosus. Antioxidants (Basel) 2025; 14:303. [PMID: 40227251 PMCID: PMC11939823 DOI: 10.3390/antiox14030303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/16/2025] [Accepted: 02/27/2025] [Indexed: 04/15/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that is frequently diagnosed in female patients, caused by multiple interacting factors. It has a complex pathogenesis which can affect almost any organ, from the kidneys to the cardiovascular, pulmonary, neurological, osteoarticular, and hematological systems. The present narrative review seeks to elucidate the role of reactive oxygen species (ROS) in the pathogenesis of SLE. The central question guiding this study is to what extent these serum protein modifications correlate with disease activity and organ damage in SLE. It is characterized by the decreased apoptosis and increased necrosis of T cells and the NETosis of granulocytes. Given the impact of an SLE diagnosis on one's life, this narrative review aims to evaluate the intricacies of oxidative stress and its relevance to the pathogenesis and treatment of the disease. Topics such as understanding processes of oxidative stress, their damaging pathways, oxidative stress biomarkers, and their role in the future assistance of clinical decisions will be discussed in the article. The accurate determination of biomarkers is taught to improve both the diagnosis and the management of the disease, while antioxidant therapy may open a new door for the treatment.
Collapse
Affiliation(s)
- Ancuta Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (C.G.); (V.V.L.)
| | - Gabriela Stoleriu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.S.); (G.B.)
| | - Alin Horatiu Nedelcu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (S.K.S.); (D.L.S.)
| | - Sara Nadeea Perju
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (C.G.); (V.V.L.)
| | - Cristina Gavrilovici
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (C.G.); (V.V.L.)
| | - Ginel Baciu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (G.S.); (G.B.)
| | - Cristina Maria Mihai
- Pediatrics, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania; (C.M.M.); (T.C.)
| | - Tatiana Chisnoiu
- Pediatrics, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania; (C.M.M.); (T.C.)
| | - Ionela Daniela Morariu
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Ecaterina Grigore
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (S.K.S.); (D.L.S.)
| | - Shwan Karwan Shawais
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (S.K.S.); (D.L.S.)
| | - Delia Lidia Salaru
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (S.K.S.); (D.L.S.)
| | - Ninel Revenco
- Pediatrics, “Nicolae Testemitanu” State University of Medicine and Pharmacy, 2004 Chisinau, Moldova;
| | - Vasile Valeriu Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (C.G.); (V.V.L.)
| |
Collapse
|
6
|
Radziszewska A, Peckham H, Restuadi R, Kartawinata M, Moulding D, de Gruijter NM, Robinson GA, Butt M, Deakin CT, Wilkinson MGL, Wedderburn LR, Jury EC, Rosser EC, Ciurtin C. Type I interferon and mitochondrial dysfunction are associated with dysregulated cytotoxic CD8+ T cell responses in juvenile systemic lupus erythematosus. Clin Exp Immunol 2025; 219:uxae127. [PMID: 39719886 PMCID: PMC11748002 DOI: 10.1093/cei/uxae127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/03/2024] [Accepted: 12/22/2024] [Indexed: 12/26/2024] Open
Abstract
Juvenile systemic lupus erythematosus (JSLE) is an autoimmune condition which causes significant morbidity in children and young adults and is more severe in its presentation than adult-onset SLE. While many aspects of immune dysfunction have been studied extensively in adult-onset SLE, there is limited and contradictory evidence of how cytotoxic CD8+ T cells contribute to disease pathogenesis and studies exploring cytotoxicity in JSLE are virtually non-existent. Here, we report that CD8+ T cell cytotoxic capacity is reduced in JSLE versus healthy controls, irrespective of treatment or disease activity. Transcriptomic and serum metabolomic analysis identified that this reduction in cytotoxic CD8+ T cells in JSLE was associated with upregulated type I interferon (IFN) signalling, mitochondrial dysfunction, and metabolic disturbances when compared to controls. Greater interrogation of the influence of these pathways on altered cytotoxic CD8+ T cell function demonstrated that JSLE CD8+ T cells had enlarged mitochondria and enhanced sensitivity to IFN-α leading to selective apoptosis of effector memory (EM) CD8+ T cells, which are enriched for cytotoxic mediator-expressing cells. This process ultimately contributes to the observed reduction in CD8+ T cell cytotoxicity in JSLE, reinforcing the growing evidence that mitochondrial dysfunction is a key pathogenic factor affecting multiple immune cell populations in type I IFN-driven rheumatic diseases.
Collapse
Affiliation(s)
- Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Restuadi Restuadi
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Melissa Kartawinata
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Dale Moulding
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
- Developmental Biology and Cancer Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Nina M de Gruijter
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - George A Robinson
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Maryam Butt
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Claire T Deakin
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
| | - Meredyth G Ll Wilkinson
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
| | - Lucy R Wedderburn
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
| | - Elizabeth C Jury
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Elizabeth C Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| |
Collapse
|
7
|
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] [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
|
8
|
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
|
9
|
Long H, Espinosa L, Sawalha AH. Unraveling the immunomodulatory impact of hydroxychloroquine on peripheral T cells using single-cell RNA sequencing. J Autoimmun 2024; 149:103324. [PMID: 39405653 DOI: 10.1016/j.jaut.2024.103324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/28/2024] [Accepted: 10/05/2024] [Indexed: 12/15/2024]
Abstract
Hydroxychloroquine (HCQ) is widely used in the treatment of a variety of autoimmune diseases. However, the mechanisms responsible for the immunomodulatory properties of HCQ in T cells remain unclear. Here we used single-cell RNA-sequencing to examine the effect of HCQ on T cells following in vitro stimulation. HCQ treatment led to a reduction in effector CD4+ T cells and upregulation of inhibitory genes including CTLA4 and TNFAIP3 in effector and naive CD4+ T cells, respectively. HCQ induced a significant expansion of effector CD8+ T cells, and significantly upregulated key cytotoxicity genes including GZMA, GZMB, GZMH, KLRD1, NKG7, and PRF1, as well as IFNG expression. Furthermore, HCQ treatment led to a reduction in the CD38+ CD8+ T cell subset, which is characterized by defective cytotoxicity and thought to both play a pathogenic role and increase susceptibility to infections in autoimmunity. We analyzed single-cell RNA-sequencing data in effector CD8+ T cells from lupus patients with or without HCQ treatment and confirmed upregulation of key cytotoxicity genes in patients receiving HCQ. In conclusion, this work provides additional insights into the immunomodulatory effects of HCQ and indicates that HCQ improves T cell cytotoxicity, which could explain a previously suggested protective effect of HCQ against infections in patients with autoimmune diseases.
Collapse
Affiliation(s)
- Huizhong Long
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China; Division of Rheumatology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Luis Espinosa
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA; Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Lupus Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Su X, Yu H, Lei Q, Chen X, Tong Y, Zhang Z, Yang W, Guo Y, Lin L. Systemic lupus erythematosus: pathogenesis and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:54. [PMID: 39472388 PMCID: PMC11522254 DOI: 10.1186/s43556-024-00217-8] [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: 07/14/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifaceted autoimmune disorder characterized by dysregulated immune responses and autoantibody production, which affects multiple organs and varies in clinical presentation and disease severity. The development of SLE is intricate, encompassing dysregulation within the immune system, a collapse of immunological tolerance, genetic susceptibilities to the disease, and a variety of environmental factors that can act as triggers. This review provides a comprehensive discussion of the pathogenesis and treatment strategies of SLE and focuses on the progress and status of traditional and emerging treatment strategies for SLE. Traditional treatment strategies for SLE have mainly employed non-specific approaches, including cytotoxic and immunosuppressive drugs, antimalarials, glucocorticoids, and NSAIDs. These strategies are effective in mitigating the effects of the disease, but they are not a complete cure and are often accompanied by adverse reactions. Emerging targeted therapeutic drugs, on the other hand, aim to control and treat SLE by targeting B and T cells, inhibiting their activation and function, as well as the abnormal activation of the immune system. A deeper understanding of the pathogenesis of SLE and the exploration of new targeted treatment strategies are essential to advance the treatment of this complex autoimmune disease.
Collapse
Affiliation(s)
- Xu Su
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Hui Yu
- Department of Urology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610014, China
| | - Qingqiang Lei
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400000, China
| | - Xuerui Chen
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Yanli Tong
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades, Paris, F-75015, France
| | - Zhongyang Zhang
- Department of Health Technology, The Danish National Research Foundation and Villum Foundation's Center IDUN, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Wenyong Yang
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
- Department of Neurosurgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610014, China.
| | - Yuanbiao Guo
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
| | - Liangbin Lin
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
11
|
Zhu M, Wu Y, Zhu T, Chen J, Chen Z, Ding H, Tan S, He J, Zeng Q, Huang X. Multifunctional Bispecific Nanovesicles Targeting SLAMF7 Trigger Potent Antitumor Immunity. Cancer Immunol Res 2024; 12:1007-1021. [PMID: 38819238 DOI: 10.1158/2326-6066.cir-23-1102] [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: 12/27/2023] [Revised: 04/03/2024] [Accepted: 05/29/2024] [Indexed: 06/01/2024]
Abstract
The effectiveness of immune checkpoint inhibitor (ICI) therapy is hindered by the ineffective infiltration and functioning of cytotoxic T cells and the immunosuppressive tumor microenvironment (TME). Signaling lymphocytic activation molecule family member 7 (SLAMF7) is a pivotal co-stimulatory receptor thought to simultaneously trigger NK-cell, T-cell, and macrophage antitumor cytotoxicity. However, the potential of this collaborative immune stimulation in antitumor immunity for solid tumors is underexplored due to the exclusive expression of SLAMF7 by hematopoietic cells. Here, we report the development and characterization of multifunctional bispecific nanovesicles (NVs) targeting SLAMF7 and glypican-3-a hepatocellular carcinoma (HCC)-specific tumor antigen. We found that by effectively "decorating" the surfaces of solid tumors with SLAMF7, these NVs directly induced potent and specific antitumor immunity and remodeled the immunosuppressive TME, sensitizing the tumors to programmed cell death protein 1 (PD1) blockade. Our findings highlight the potential of SLAMF7-targeted multifunctional bispecific NVs as an anticancer strategy with implications for designing next-generation targeted cancer therapies.
Collapse
Affiliation(s)
- Manman Zhu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Tianchuan Zhu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jian Chen
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Zhenxing Chen
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Kingcell Regenerative Medicine (Guangdong) Co., Zhuhai, China
| | - Hanxi Ding
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Siyi Tan
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jianzhong He
- Department of Pathology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Qi Zeng
- Cancer Center, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
12
|
Balog JÁ, Zvara Á, Bukovinszki V, Puskás LG, Balog A, Szebeni GJ. Comparative single-cell multiplex immunophenotyping of therapy-naive patients with rheumatoid arthritis, systemic sclerosis, and systemic lupus erythematosus shed light on disease-specific composition of the peripheral immune system. Front Immunol 2024; 15:1376933. [PMID: 38726007 PMCID: PMC11079270 DOI: 10.3389/fimmu.2024.1376933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/03/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Systemic autoimmune diseases (SADs) are a significant burden on the healthcare system. Understanding the complexity of the peripheral immunophenotype in SADs may facilitate the differential diagnosis and identification of potential therapeutic targets. Methods Single-cell mass cytometric immunophenotyping was performed on peripheral blood mononuclear cells (PBMCs) from healthy controls (HCs) and therapy-naive patients with rheumatoid arthritis (RA), progressive systemic sclerosis (SSc), and systemic lupus erythematosus (SLE). Immunophenotyping was performed on 15,387,165 CD45+ live single cells from 52 participants (13 cases/group), using an antibody panel to detect 34 markers. Results Using the t-SNE (t-distributed stochastic neighbor embedding) algorithm, the following 17 main immune cell types were determined: CD4+/CD57- T cells, CD4+/CD57+ T cells, CD8+/CD161- T cells, CD8+/CD161+/CD28+ T cells, CD8dim T cells, CD3+/CD4-/CD8- T cells, TCRγ/δ T cells, CD4+ NKT cells, CD8+ NKT cells, classic NK cells, CD56dim/CD98dim cells, B cells, plasmablasts, monocytes, CD11cdim/CD172dim cells, myeloid dendritic cells (mDCs), and plasmacytoid dendritic cells (pDCs). Seven of the 17 main cell types exhibited statistically significant frequencies in the investigated groups. The expression levels of the 34 markers in the main populations were compared between HCs and SADs. In summary, 59 scatter plots showed significant differences in the expression intensities between at least two groups. Next, each immune cell population was divided into subpopulations (metaclusters) using the FlowSOM (self-organizing map) algorithm. Finally, 121 metaclusters (MCs) of the 10 main immune cell populations were found to have significant differences to classify diseases. The single-cell T-cell heterogeneity represented 64MCs based on the expression of 34 markers, and the frequency of 23 MCs differed significantly between at least twoconditions. The CD3- non-T-cell compartment contained 57 MCs with 17 MCs differentiating at least two investigated groups. In summary, we are the first to demonstrate the complexity of the immunophenotype of 34 markers over 15 million single cells in HCs vs. therapy-naive patients with RA, SSc, and SLE. Disease specific population frequencies or expression patterns of peripheral immune cells provide a single-cell data resource to the scientific community.
Collapse
Affiliation(s)
- József Á. Balog
- Laboratory of Functional Genomics, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
- Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Ágnes Zvara
- Laboratory of Functional Genomics, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
- Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Vivien Bukovinszki
- Department of Rheumatology and Immunology, Faculty of Medicine, Albert Szent-Gyorgyi Health Centre, University of Szeged, Szeged, Hungary
| | - László G. Puskás
- Laboratory of Functional Genomics, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
- Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Attila Balog
- Department of Rheumatology and Immunology, Faculty of Medicine, Albert Szent-Gyorgyi Health Centre, University of Szeged, Szeged, Hungary
| | - Gábor J. Szebeni
- Laboratory of Functional Genomics, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
- Core Facility, HUN-REN Biological Research Centre, Szeged, Hungary
- Department of Internal Medicine, Hematology Centre, Faculty of Medicine University of Szeged, Szeged, Hungary
- Astridbio Technologies Ltd., Szeged, Hungary
| |
Collapse
|
13
|
Wang CM, Jan Wu YJ, Zheng JW, Huang LY, Tan KP, Chen JY. T cell expressions of aberrant gene signatures and Co-inhibitory receptors (Co-IRs) as predictors of renal damage and lupus disease activity. J Biomed Sci 2024; 31:41. [PMID: 38650001 PMCID: PMC11034032 DOI: 10.1186/s12929-024-01024-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 04/01/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is distinguished by an extensive range of clinical heterogeneity with unpredictable disease flares and organ damage. This research investigates the potential of aberrant signatures on T cell genes, soluble Co-IRs/ligands, and Co-IRs expression on T cells as biomarkers for lupus disease parameters. METHODS Comparative transcriptome profiling analysis of non-renal and end-stage renal disease (ESRD) phenotypes of SLE was performed using CD4 + and CD8 + cDNA microarrays of sorted T cells. Comparing the expression of Co-IRs on T cells and serum soluble mediators among healthy and SLE phenotypes. RESULTS SLE patients with ESRD were downregulated CD38, PLEK, interferon-γ, CX3CR1, FGFBP2, and SLCO4C1 transcripts on CD4 + and CD8 + T cells simultaneously and NKG7, FCRL6, GZMB/H, FcγRIII, ITGAM, Fas ligand, TBX21, LYN, granulysin, CCL4L1, CMKLR1, HLA-DRβ, KIR2DL3, and KLRD1 in CD8 T cells. Pathway enrichment and PPI network analyses revealed that the overwhelming majority of Differentially Expressed Genes (DEGs) have been affiliated with novel cytotoxic, antigen presentation, and chemokine-cell migration signature pathways. CD8 + GZMK + T cells that are varied in nature, including CD161 + Mucosal-associated invariant T (MAIT) cells and CD161- aged-associated T (Taa) cells and CD161-GZMK + GZMB + T cells might account for a higher level of GZMK in CD8 + T cells associated with ESRD. SLE patients have higher TIGIT + , PD1 + , and lower CD127 + cell percentages on CD4 + T cells, higher TIM3 + , TIGIT + , HLA-DR + cell frequency, and lower MFI expression of CD127, CD160 in CD8 T cells. Co-IRs expression in T cells was correlated with soluble PD-1, PDL-2, and TIM3 levels, as well as SLE disease activity, clinical phenotypes, and immune-therapy responses. CONCLUSION The signature of dysfunctional pathways defines a distinct immunity pattern in LN ESRD patients. Expression levels of Co-IRs in peripheral blood T cells and serum levels of soluble PD1/PDL-2/TIM3 can serve as biomarkers for evaluating clinical parameters and therapeutic responses.
Collapse
Affiliation(s)
- Chin-Man Wang
- Department of Rehabilitation, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, No. 5, Fu-Shin St. Kwei-Shan, Taoyuan, Republic of China
| | - Yeong-Jian Jan Wu
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan, No. 5, Fu-Shin St. Kwei-Shan, Republic of China
| | - Jian-Wen Zheng
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan, No. 5, Fu-Shin St. Kwei-Shan, Republic of China
| | - Li Yu Huang
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan, No. 5, Fu-Shin St. Kwei-Shan, Republic of China
| | - Keng Poo Tan
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan, No. 5, Fu-Shin St. Kwei-Shan, Republic of China
| | - Ji-Yih Chen
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan, No. 5, Fu-Shin St. Kwei-Shan, Republic of China.
| |
Collapse
|
14
|
Yuan F, Wei J, Cheng Y, Wang F, Gu M, Li Y, Zhao X, Sun H, Ban R, Zhou J, Xia Z. SLAMF7 Promotes Foam Cell Formation of Macrophage by Suppressing NR4A1 Expression During Carotid Atherosclerosis. Inflammation 2024; 47:530-542. [PMID: 37971565 DOI: 10.1007/s10753-023-01926-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/25/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Abstract
Macrophage-derived lipid-laden foam cells from the subendothelium play a crucial role in the initiation and progression of atherosclerosis. However, the molecule mechanism that regulates the formation of foam cells is not completely understood. Here, we found that SLAMF7 was upregulated in mice bone marrow-derived macrophages and RAW264.7 cells stimulated with oxidized low-density lipoprotein (ox-LDL). SLAMF7 promoted ox-LDL-mediated macrophage lipid accumulation and M1-type polarization. SLAMF7 deficiency reduced serum lipid levels and improved the lesions area of carotid plaque and aortic arch in high-fat diet-fed ApoE-/- mice. In response to ox-LDL, SLAMF7 downregulated NR4A1 and upregulated RUNX3 through transcriptome sequencing analysis. Overexpression NR4A1 reversed SLAMF7-induced lipid uptake and M1 polarization via inhibiting RUNX3 expression. Furthermore, RUNX3 enhanced foam cell formation and M1-type polarization. Taken together, the study suggested that SLAMF7 play contributing roles in the pro-atherogenic effects by regulating NR4A1-RUNX3.
Collapse
Affiliation(s)
- Fengjiao Yuan
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, People's Republic of China
- Department of Neurology, Liaocheng People's Hospital, Shandong University, Jinan, Shandong, 250012, People's Republic of China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Jianmei Wei
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, People's Republic of China
| | - Yan Cheng
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, People's Republic of China
| | - Feifei Wang
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, People's Republic of China
| | - Mingliang Gu
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, People's Republic of China
| | - Yanhui Li
- Department of Rehabilitation Medicine, Liaocheng Chinese Medicine Hospital, Liaocheng, Shandong, 252000, People's Republic of China
| | - Xin Zhao
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong, 252000, People's Republic of China
| | - Hao Sun
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong, 252000, People's Republic of China
| | - Ru Ban
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong, 252000, People's Republic of China
| | - Jing Zhou
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250014, People's Republic of China.
| | - Zhangyong Xia
- Department of Neurology, Liaocheng People's Hospital, Shandong University, Jinan, Shandong, 250012, People's Republic of China.
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong, 252000, People's Republic of China.
| |
Collapse
|
15
|
Velayutham B, Padhi S, Devi S, Patra S, Panigrahi C, Ramasubbu MK, Kumar R, Raheman S. Immunohistochemical expression of perforin in adult systemic lupus erythematosus associated macrophage activation syndrome: Clinicohematological correlation and literature review. Lupus 2024; 33:26-39. [PMID: 38069452 DOI: 10.1177/09612033231221414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
OBJECTIVE To study the bone marrow (BM) immunohistomorphological characteristics in adult systemic lupus erythematosus (SLE) associated macrophage activation syndrome (SLE-MAS). MATERIALS AND METHODS Immunohistochemical (IHC) expression of CD3, CD8, perforin (PFN), and CD163 was studied on BM trephine biopsies from 30 cytopenic adult SLE cases (male: female = 1:5, age; 24 years, range; 19-32) and compared them with ten age matched controls. Clinicopathological parameters were compared among the cases likely (L) or unlikely (U) to have MAS using probability scoring criteria. The best cut off laboratory parameters to discriminate between the two were obtained through receiver operator curve (ROC) analysis. RESULTS MAS occurred in 12/30 (40%) cases and was more commonly associated with prior immunosuppressive therapy (p = .07), ≥ 3 system involvement (p = .09), lower fibrinogen (p < .01), increased triglyceride (p = .002), increased BM hemophagocytosis (p = .002), and higher MAS score [185 (176-203) vs. 105 (77-119), p < .01] than MAS-U subgroup. Although PFN+CD8+ T lymphocytes significantly decreased among cases than controls (p < .05), it was comparable between MAS-L and MAS-U subgroups. Fibrinogen (< 2.4 g/L, AUC; 0.93, p < .01), hemophagocytosis score (> 1.5, AUC; 0.71, p = .03), and an MAS probability score of ≥ 164 (AUC; 1, p < .01) discriminated MAS from those without MAS. CONCLUSION We noted a decrease in perforin mediated CD8 + T cell cytotoxicity in SLE. Immunohistochemical demonstration of the same along with histiocytic hemophagocytosis on BM biopsy may be useful adjunct in early diagnosis and management of MAS in SLE.
Collapse
Affiliation(s)
- Bakialakshmi Velayutham
- Department of Pathology with Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Somanath Padhi
- Department of Pathology with Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Sujata Devi
- Department of General Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Susama Patra
- Department of Pathology with Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Chinmayee Panigrahi
- Department of Pathology with Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Mathan Kumar Ramasubbu
- Department of Pharmacology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Rajesh Kumar
- Department of General Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
- Department of General Medicine, All India Institute of Medical Sciences, Deoghar, Jharkhand, India
| | - Samiur Raheman
- Department of Pathology with Laboratory Medicine, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| |
Collapse
|
16
|
Turner CN, Camilo Sanchez Arcila J, Huerta N, Quiguoe AR, Jensen KDC, Hoyer KK. T cell exhaustion dynamics in systemic autoimmune disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.23.573167. [PMID: 38187518 PMCID: PMC10769367 DOI: 10.1101/2023.12.23.573167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Unlike in infection and cancer, T cell exhaustion in autoimmune disease has not been clearly defined. Here we set out to understand inhibitory protein (PD-1, Tim3, CTLA4, Lag3) expression in CXCR5- and CXCR5+ CD8 and CD4 T cells in systemic lupus erythematosus. CXCR5+ CD8 and CD4 T cells express PD-1 and engage B cells in germinal center reactions, leading to autoantibody formation in autoimmunity. We hypothesized that CXCR5+ CD8 T cells develop an exhausted phenotype as SLE autoimmunity expands from initial to chronic, self-perpetuating disease due to chronic self-antigen exposure. Our results indicate that there is no exhaustion frequency differences between sexes, although disease kinetics vary by sex. CXCR5+ CD8 T cells express primarily IFNγ, known to promote autoimmune disease development, whereas CXCR5-CD8 T cells express TNFα and IFNγ as disease progresses from 2-6 months. Tim3 is the highest expressed inhibitory marker for all CD4 and CD8 T cell populations demonstrating potential for terminally exhausted populations. CTLA4 expression on CD4 T cells suggests potential tolerance induction in these cells. We identified exhaustion phenotypes within autoimmune disease that progress with increasing lupus erythematosus severity and possibly provide a feedback mechanism for immunological tolerance. Highlights CXCR5- and CXCR5+ CD8 T cells expand with rate of disease in SLE mouse model.CXCR5+ CD8 T cells are low contributors to TNFα disease progression unlike CXCR5-CD8 T cells but may increase disease mechanisms through high IFNγ production.Inhibitory markers upregulate in frequency with the highest amounts seen in Tim3+ populations. Tim3+Lag3+ expression may be an indicator of terminal differentiation for all populations.Inhibitory marker expression frequency was unrelated to sex.
Collapse
|
17
|
Deng Y, Ou YY, Mo CJ, Huang L, Qin X. Characteristics and clustering analysis of peripheral blood lymphocyte subsets in children with systemic lupus erythematosus complicated with clinical infection. Clin Rheumatol 2023; 42:3299-3309. [PMID: 37537315 DOI: 10.1007/s10067-023-06716-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/14/2023] [Accepted: 07/22/2023] [Indexed: 08/05/2023]
Abstract
OBJECTIVES Clinical infection is a common complication in children with systemic lupus erythematosus (SLE). However, few studies have investigated immune alterations in children with SLE complicated with clinical infection. We assessed lymphocyte subsets in children with SLE to explore the possibility of clinical infection. METHODS We retrospectively analyzed the proportion of peripheral lymphocyte subsets in 140 children with SLE. Children with SLE were classified into different clusters according to the proportion of peripheral blood lymphocyte subsets: (CD3 + /CD4 + T cell, CD3 + /CD8 + T cell, CD3 + /CD4 + /CD8 + T cell, CD3 + /CD4-/CD8- T cell, CD19 + B cell, and CD3-/CD16 + /CD56 + NK cell). Differences in the proportion of lymphoid subsets, infection rates, and systemic lupus erythematosus disease activity index (SLEDAI) scores were compared between clusters. In addition, we grouped the subjects according to the presence or absence of infection. Proportions of lymphoid subsets, demographic variables, clinical presentation, and other laboratory variables were compared between the infected and uninfected groups. Finally, the diagnostic ability of lymphocyte subset ratios to distinguish secondary infection in children with SLE was predicted using an ROC curve. RESULTS Cluster C2 had a higher proportion of B cells than Cluster C1, while Cluster C1 had a lower proportion of NK cells, CD3 + T cells, CD3 + /CD4 + T cells, CD3 + /CD8 + T cells, and CD3 + /CD4-/CD8- T cells. Infection rates and SLEDAI scores were higher in Cluster C2 than in Cluster C1. The infected children had a higher proportion of B cells and a lower proportion of CD3 + T cells, CD3 + /CD4 + T cells, CD3 + /CD8 + T cells, and CD3 + /CD4-/CD8- T cells. There were no significant differences in lymphoid subsets between children in Cluster C2 and the infected groups. The area under the ROC curve of B lymphocytes in predicting SLE children with infection was 0.842. The area under the ROC curve was 0.855 when a combination of B cells, NK cells, CD4 + T cells, and CD8 + T cells was used to predict the outcome of coinfection. CONCLUSIONS A high percentage of B cells and a low percentage of CD3 + T cells, CD3 + /CD4 + T cells, CD3 + /CD8 + T cells, CD3 + /CD4 + /CD8 + T cells, and CD3 + /CD4-/CD8- T cells may be associated with infection in children with SLE. B cells was used to predict the outcome of coinfection in children with SLE. Key Points • A high percentage of B cells and a low percentage of CD3 + T cells, CD3 + /CD4 + T cells, CD3 + /CD8 + T cells, CD3 + /CD4 + /CD8 + T cells, and CD3 + /CD4-/CD8- T cells may be associated with infection in children with SLE • B cells was used to predict the outcome of coinfection in children with SLE.
Collapse
Affiliation(s)
- Yan Deng
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Medicine of Guangxi, Department of Education, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ying-Ying Ou
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Medicine of Guangxi, Department of Education, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Cui-Ju Mo
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Medicine of Guangxi, Department of Education, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Li Huang
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Medicine of Guangxi, Department of Education, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xue Qin
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Medicine of Guangxi, Department of Education, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
18
|
Liu J, Peng H, Yu T, Huang Y, Tan N, Pang L, Wu Y, Wang L. Increased SLAMF7 +CD8 + T cells are associated with the pathogenesis of experimental autoimmune pancreatitis in mice. Pancreatology 2023; 23:767-776. [PMID: 37661465 DOI: 10.1016/j.pan.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND IgG4-related autoimmune pancreatitis (AIP) is considered to be a T cell-mediated autoimmune disease. However, CD8+ T cells have only received brief mention, and have yet to be completely studied. The study aimed to investigate the expression of signaling lymphocytic activation molecule family 7 (SLAMF7) on CD8+ T cells and the features of SLAMF7+CD8+ T cells in MRL/Mp mice with AIP. METHODS A murine model of AIP was established by intraperitoneal injection with polyinosinic:polycytidylic acid (poly I:C) for 8 weeks. Dexamethasone treatment was daily administrated for the last 2 weeks during a 6-week course of poly I:C. SLAMF7 expression on CD8+ T cells in the spleen and pancreas was detected by flow cytometry. Granzyme B (GZMB) and cytokines including IFN-γ, TNF-α, and IL-2, were monitored in an in vitro T cell activation assay. Dexamethasone suppression assays were performed to downregulate SLAMF7 expression on T cells upon T cell receptor stimulation. RESULTS AIP in MRL/Mp mice was induced by repeated intraperitoneal administration of poly I:C and CD8+ T cells were increased in the inflamed pancreas. SLAMF7+CD8+ T cells were elevated in the spleen and pancreas of AIP mice. SLAMF7+CD8+ T subsets produced more GZMB, IFN-γ, TNF-α and IL-2 than SLAMF7-CD8+ T subsets. Dexamethasone treatment ameliorated pancreatic inflammatory and fibrosis of AIP. Dexamethasone could downregulate SLAMF7+CD8+ T cells and reduce GZMB, IFN-γ and TNF-α levels both in vitro and in vivo. CONCLUSIONS Increased SLAMF7+CD8+ T cells exhibit enhanced cytotoxicity and cytokines secretion capacity, which may be involved in the pathogenesis of AIP.
Collapse
Affiliation(s)
- Jia Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hui Peng
- Department of Pathology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Tingfeng Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yanlin Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ning Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Li Pang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yongtong Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lingyun Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
19
|
Spinelli FR, Berti R, Farina G, Ceccarelli F, Conti F, Crescioli C. Exercise-induced modulation of Interferon-signature: a therapeutic route toward management of Systemic Lupus Erythematosus. Autoimmun Rev 2023; 22:103412. [PMID: 37597604 DOI: 10.1016/j.autrev.2023.103412] [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: 08/01/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
Systemic Lupus Erythematosus (SLE) is a multisystemic autoimmune disorder characterized by flares-ups/remissions with a complex clinical picture related to disease severity and organ/tissue injury, which, if left untreated, may result in permanent damage. Enhanced fatigue and pain perception, worsened quality of life (QoL) and outcome are constant, albeit symptoms may differ. An aberrant SLE immunoprofiling, note as "interferon (IFN)α-signature", is acknowledged to break immunotolerance. Recently, a deregulated "IFNγ-signature" is suggested to silently precede/trigger IFNα profile before clinical manifestations. IFNα- and IFNγ-over-signaling merge in cytokine/chemokine overexpression exacerbating autoimmunity. Remission achievement and QoL improvement are the main goals. The current therapy (i.e., corticosteroids, immunosuppressants) aims to downregulate immune over-response. Exercise could be a safe treatment due to its ever-emerging ability to shape and re-balance immune system without harmful side-effects; in addition, it improves cardiorespiratory capacity and musculoskeletal strength/power, usually impaired in SLE. Nevertheless, exercise is not yet included in SLE care plans. Furthermore, due to the fear to worsening pain/fatigue, SLE subjects experience kinesiophobia and sedentary lifestyle, worsening physical health. Training SLE patients to exercise is mandatory to fight inactive behavior and ameliorate health. This review aims to focus the attention on the role of exercise as a non-pharmacological therapy in SLE, considering its ability to mitigate IFN-signature and rebalance (auto)immune response. To this purpose, the significance of IFNα- and IFNγ-signaling in SLE etiopathogenesis will be addressed first and discussed thereafter as biotarget of exercise. Comments are addressed on the need to make aware all SLE care professional figures to promote exercise for health patients.
Collapse
Affiliation(s)
- Francesca Romana Spinelli
- Sapienza Università di Roma, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Roma, Italy
| | - Riccardo Berti
- University of Rome Foro Italico, Department of Movement, Human and Health Sciences, Rome, Italy
| | - Gabriele Farina
- University of Rome Foro Italico, Department of Movement, Human and Health Sciences, Rome, Italy
| | - Fulvia Ceccarelli
- Sapienza Università di Roma, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Roma, Italy
| | - Fabrizio Conti
- Sapienza Università di Roma, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Roma, Italy
| | - Clara Crescioli
- University of Rome Foro Italico, Department of Movement, Human and Health Sciences, Rome, Italy.
| |
Collapse
|
20
|
Han L, Zhang L. CCL21/CCR7 axis as a therapeutic target for autoimmune diseases. Int Immunopharmacol 2023; 121:110431. [PMID: 37331295 DOI: 10.1016/j.intimp.2023.110431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/19/2023] [Accepted: 05/30/2023] [Indexed: 06/20/2023]
Abstract
Chemokine receptor 7 (CCR7) is a G protein-coupled receptor containing 7 transmembrane domains that is expressed on various cells, such as naive T/B cells, central memory T cells, regulatory T cells, immature/mature dendritic cells (DCs), natural killer cells, and a minority of tumor cells. Chemokine ligand 21 (CCL21) is the known high-affinity ligand that binds to CCR7 and drives cell migration in tissues. CCL21 is mainly produced by stromal cells and lymphatic endothelial cells, and its expression is significantly increased under inflammatory conditions. Genome-wide association studies (GWAS) have shown a strong association between CCL21/CCR7 axis and disease severity in patients with rheumatoid arthritis, sjogren's syndrome, systemic lupus erythematosus, polymyositis, ankylosing spondylitis, and asthma. Disrupting CCL21/CCR7 interaction with antibodies or inhibitors prevents the migration of CCR7-expressing immune and non-immune cells at the site of inflammation and reduces disease severity. This review emphasizes the importance of the CCL21 /CCR7 axis in autoimmune diseases and evaluates its potential as a novel therapeutic target for these conditions.
Collapse
Affiliation(s)
- Le Han
- Department of Pharmacy, The Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin 214400, China
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
21
|
Accapezzato D, Caccavale R, Paroli MP, Gioia C, Nguyen BL, Spadea L, Paroli M. Advances in the Pathogenesis and Treatment of Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:6578. [PMID: 37047548 PMCID: PMC10095030 DOI: 10.3390/ijms24076578] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a genetically predisposed, female-predominant disease, characterized by multiple organ damage, that in its most severe forms can be life-threatening. The pathogenesis of SLE is complex and involves cells of both innate and adaptive immunity. The distinguishing feature of SLE is the production of autoantibodies, with the formation of immune complexes that precipitate at the vascular level, causing organ damage. Although progress in understanding the pathogenesis of SLE has been slower than in other rheumatic diseases, new knowledge has recently led to the development of effective targeted therapies, that hold out hope for personalized therapy. However, the new drugs available to date are still an adjunct to conventional therapy, which is known to be toxic in the short and long term. The purpose of this review is to summarize recent advances in understanding the pathogenesis of the disease and discuss the results obtained from the use of new targeted drugs, with a look at future therapies that may be used in the absence of the current standard of care or may even cure this serious systemic autoimmune disease.
Collapse
Affiliation(s)
- Daniele Accapezzato
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Rosalba Caccavale
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Pia Paroli
- Eye Clinic, Department of Sense Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Chiara Gioia
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Bich Lien Nguyen
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Luca Spadea
- Post Graduate School of Public Health, University of Siena, 53100 Siena, Italy
| | - Marino Paroli
- Division of Clinical Immunology, Department of Clinical, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
22
|
Khunsriraksakul C, Li Q, Markus H, Patrick MT, Sauteraud R, McGuire D, Wang X, Wang C, Wang L, Chen S, Shenoy G, Li B, Zhong X, Olsen NJ, Carrel L, Tsoi LC, Jiang B, Liu DJ. Multi-ancestry and multi-trait genome-wide association meta-analyses inform clinical risk prediction for systemic lupus erythematosus. Nat Commun 2023; 14:668. [PMID: 36750564 PMCID: PMC9905560 DOI: 10.1038/s41467-023-36306-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 01/25/2023] [Indexed: 02/09/2023] Open
Abstract
Systemic lupus erythematosus is a heritable autoimmune disease that predominantly affects young women. To improve our understanding of genetic etiology, we conduct multi-ancestry and multi-trait meta-analysis of genome-wide association studies, encompassing 12 systemic lupus erythematosus cohorts from 3 different ancestries and 10 genetically correlated autoimmune diseases, and identify 16 novel loci. We also perform transcriptome-wide association studies, computational drug repurposing analysis, and cell type enrichment analysis. We discover putative drug classes, including a histone deacetylase inhibitor that could be repurposed to treat lupus. We also identify multiple cell types enriched with putative target genes, such as non-classical monocytes and B cells, which may be targeted for future therapeutics. Using this newly assembled result, we further construct polygenic risk score models and demonstrate that integrating polygenic risk score with clinical lab biomarkers improves the diagnostic accuracy of systemic lupus erythematosus using the Vanderbilt BioVU and Michigan Genomics Initiative biobanks.
Collapse
Affiliation(s)
- Chachrit Khunsriraksakul
- Program in Bioinformatics and Genomics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
- Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Qinmengge Li
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Havell Markus
- Program in Bioinformatics and Genomics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
- Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Matthew T Patrick
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Renan Sauteraud
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Daniel McGuire
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Xingyan Wang
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Chen Wang
- Program in Bioinformatics and Genomics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Lida Wang
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Siyuan Chen
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Ganesh Shenoy
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Bingshan Li
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, 37235, USA
| | - Xue Zhong
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Nancy J Olsen
- Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Laura Carrel
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Bibo Jiang
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Dajiang J Liu
- Program in Bioinformatics and Genomics, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
23
|
A single-cell map of peripheral alterations after FMT treatment in patients with systemic lupus erythematosus. J Autoimmun 2023; 135:102989. [PMID: 36610264 DOI: 10.1016/j.jaut.2022.102989] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 01/07/2023]
Abstract
Systemic lupus erythematosus (SLE) is characterized by loss of self-tolerance and persistent self-aggression, sustained chronic inflammation, production of autoantibodies and multi-system damage, and is largely incurable to date. The gut microbiota and its metabolites, now recognized as crucial environmental triggers of local/systemic immune reactions, have been implicated in the development and progression of SLE. Fecal microbiota transplantation (FMT) is restoration of disturbed microbiota by transplanting foreign gut microbiota from healthy individuals into the gastrointestinal tract of diseased individuals. Our previous clinical trial suggests that FMT is a potentially safe and effective treatment for SLE. In order to elucidate the potential effect of FMT on peripheral immune cells of patients with SLE, we collected PBMCs (n = 30) of 13 SLE patients who participated in the clinical trial before and after the FMT-treatment, and performed single-cell RNA sequencing. The results first revealed that peripheral T lymphocytes of SLE patients decreased and NK cells increased after the FMT treatment. Then, sub-clustering analysis discovered that total CD4+ T cells highly expressed genes of IL7R, CD28, and CD8+ T cells highly expressed genes of GZMH and NKG7 after FMT treatment. Moreover, FMT treatment reduced the expression of interferon-related genes (IRGs) in CD4+ T, CD8+ T, DP, NK, and B cells of SLE patients. More importantly, interferon-related pathways were more enriched in cells of the FMT non-responder group, and further the interferon genes expression of lymphocytes and myeloid cells was negatively correlated with the efficiency of FMT treatment. Collectively, our data identified various immunophenotypic and associated gene set changes following FMT treatment, illustrating the heterogeneity of response to FMT treatment in SLE.
Collapse
|
24
|
Zhang J, Zhang S, Qiao J, Qiu M, Li X. Risk factors analysis and risk assessment model construction of systemic lupus erythematosus patients with infection. Lupus 2023; 32:119-128. [PMID: 36433710 DOI: 10.1177/09612033221141255] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To analyze the characteristics of peripheral blood lymphocyte subsets in systemic lupus erythematosus (SLE) patients with infection and non-infection group. Explore the risk factors of infection in SLE patients and establish a risk matrix model to predict the occurrence of co-infection. METHODS total of 333 SLE patients without infection, 163 patients suffering from infection, and 132 healthy controls (HCs) were recruited. General clinical data and disease activity indicators were collected. The levels of total T, B, CD4+T, CD8+T, NK, Th1, Th2, Th17, and Treg cells in peripheral blood of HCs, SLE patients (including infected and non-infected group) were analyzed by flow cytometry. The risk assessment model was constructed, and the receiver operating characteristic curve was drawn. 39 SLE patients with infection and 20 patients without infection were randomly selected to evaluate the predictive power of the regression model. RESULTS The levels of T, B, CD4+T, CD8+T, and NK cells in the infected patients were significantly decreased when compared with that of both non-infected patients and HCs (p < .05). The non-infected patients had a higher level of Th17 than that of HCs (p < . 05), but the absolute numbers of Th17 in infected patients was the lowest among the three groups (p < .001). The number of Treg cells in SLE patients was significantly lower than that of HCs (p < .01), and the infected patients had the fewest Treg cells among all these groups (p < . 05). A risk assessment model for SLE with infection was established, p = 1/(1-e-y), Y = 1.763-0.004 × Absolute number of CD4 + T cells-0.005 × Absolute number of NK cells -0.005 × Platelet count(×1012/L) + 1.033 × Absolute number of lymphocytes (×109/L) + 0.023 × C-reactive protein (mg/dL), whose predictive sensitivity is 77.5%, and specificity is 78.3%. CONCLUSION The new risk assessment model exhibits good predictive ability to assess co-infection risk in SLE patients. T cells, NK cells, and CD4 + T cells along with other parameters help in differentiating Lupus with infection from Lupus alone.
Collapse
Affiliation(s)
- Jiaqian Zhang
- Department of Rheumatology, 74761The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Shengxiao Zhang
- Department of Rheumatology, 74761The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jun Qiao
- Department of Rheumatology, 74761The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Mengting Qiu
- Department of Rheumatology, 74761The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaofeng Li
- Department of Rheumatology, 74761The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
25
|
Yang Y, Liu K, Liu M, Zhang H, Guo M. EZH2: Its regulation and roles in immune disturbance of SLE. Front Pharmacol 2022; 13:1002741. [DOI: 10.3389/fphar.2022.1002741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
The pathogenesis of systemic lupus erythematosus (SLE) is related to immune homeostasis imbalance. Epigenetic mechanisms have played a significant role in breaking immune tolerance. Enhancer of zeste homolog 2 (EZH2), the specific methylation transferase of lysine at position 27 of histone 3, is currently found to participate in the pathogenesis of SLE through affecting multiple components of the immune system. This review mainly expounds the mechanisms underlying EZH2-mediated disruption of immune homeostasis in SLE patients, hoping to provide new ideas in the pathogenesis of SLE and new targets for future treatment.
Collapse
|
26
|
Perl A, Agmon-Levin N, Crispín JC, Jorgensen TN. Editorial: New biomarkers for the diagnosis and treatment of systemic lupus erythematosus. Front Immunol 2022; 13:1009038. [PMID: 36311710 PMCID: PMC9599399 DOI: 10.3389/fimmu.2022.1009038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/20/2022] [Indexed: 01/17/2023] Open
Affiliation(s)
- Andras Perl
- Department of Medicine, College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY, United States,Department of Biochemistry and Molecular Biology, College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY, United States,Department of Microbiology and Immunology, College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY, United States,*Correspondence: Andras Perl,
| | - Nancy Agmon-Levin
- The Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat Gan, Israel
| | - José C. Crispín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico
| | - Trine N. Jorgensen
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
27
|
Radziszewska A, Moulder Z, Jury EC, Ciurtin C. CD8 + T Cell Phenotype and Function in Childhood and Adult-Onset Connective Tissue Disease. Int J Mol Sci 2022; 23:11431. [PMID: 36232733 PMCID: PMC9569696 DOI: 10.3390/ijms231911431] [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: 08/10/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022] Open
Abstract
CD8+ T cells are cytotoxic lymphocytes that destroy pathogen infected and malignant cells through release of cytolytic molecules and proinflammatory cytokines. Although the role of CD8+ T cells in connective tissue diseases (CTDs) has not been explored as thoroughly as that of other immune cells, research focusing on this key component of the immune system has recently gained momentum. Aberrations in cytotoxic cell function may have implications in triggering autoimmunity and may promote tissue damage leading to exacerbation of disease. In this comprehensive review of current literature, we examine the role of CD8+ T cells in systemic lupus erythematosus, Sjögren's syndrome, systemic sclerosis, polymyositis, and dermatomyositis with specific focus on comparing what is known about CD8+ T cell peripheral blood phenotypes, CD8+ T cell function, and CD8+ T cell organ-specific profiles in adult and juvenile forms of these disorders. Although, the precise role of CD8+ T cells in the initiation of autoimmunity and disease progression remains to be elucidated, increasing evidence indicates that CD8+ T cells are emerging as an attractive target for therapy in CTDs.
Collapse
Affiliation(s)
- Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London WC1E 6JF, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| | - Zachary Moulder
- University College London Medical School, University College London, London WC1E 6DE, UK
| | - Elizabeth C. Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London WC1E 6JF, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| |
Collapse
|
28
|
Abstract
Systemic lupus erythematosus (SLE) is a typical autoimmune disease with a complex pathogenesis and genetic predisposition. With continued understanding of this disease, it was found that SLE is related to the interferon gene signature. Most studies have emphasized the important role of IFN-α in SLE, but our previous study suggested a nonnegligible role of IFN-γ in SLE. Some scholars previously found that IFN-γ is abnormally elevated as early as before the classification of SLE and before the emergence of autoantibodies and IFN-α. Due to the large overlap between IFN-α and IFN-γ, SLE is mostly characterized by expression of the IFN-α gene after onset. Therefore, the role of IFN-γ in SLE may be underestimated. This article mainly reviews the role of IFN-γ in SLE and focuses on the nonnegligible role of IFN-γ in SLE to gain a more comprehensive understanding of the disease.
Collapse
|
29
|
Li H, Boulougoura A, Endo Y, Tsokos GC. Abnormalities of T cells in systemic lupus erythematosus: new insights in pathogenesis and therapeutic strategies. J Autoimmun 2022; 132:102870. [PMID: 35872102 DOI: 10.1016/j.jaut.2022.102870] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 11/25/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by loss of immune tolerance and sustained production of autoantibodies. Multiple and profound T cell abnormalities in SLE are intertwined with disease expression. Both numerical and functional disturbances have been reported in main CD4+ T helper cell subsets including Th1, Th2, Th17, regulatory, and follicular helper cells. SLE CD4+ T cells are known to provide help to B cells, produce excessive IL-17 but insufficient IL-2, and infiltrate tissues. In the absence of sufficient amounts of IL-2, regulatory T cells, do not function properly to constrain inflammation. A complicated series of early signaling defects and aberrant activation of kinases and phosphatases result in complex cell phenotypes by altering the metabolic profile and the epigenetic landscape. All main metabolic pathways including glycolysis, glutaminolysis and oxidative phosphorylation are altered in T cells from lupus prone mice and patients with SLE. SLE CD8+ cytotoxic T cells display reduced cytolytic activity which accounts for higher rates of infection and the sustenance of autoimmunity. Further, CD8+ T cells in the context of rheumatic diseases lose the expression of CD8, acquire IL-17+CD4-CD8- double negative T (DNT) cell phenotype and infiltrate tissues. Herein we present an update on these T cell abnormalities along with underlying mechanisms and discuss how these advances can be exploited therapeutically. Novel strategies to correct these aberrations in T cells show promise for SLE treatment.
Collapse
Affiliation(s)
- Hao Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Afroditi Boulougoura
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yushiro Endo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
Chen PM, Katsuyama E, Satyam A, Li H, Rubio J, Jung S, Andrzejewski S, Becherer JD, Tsokos MG, Abdi R, Tsokos GC. CD38 reduces mitochondrial fitness and cytotoxic T cell response against viral infection in lupus patients by suppressing mitophagy. SCIENCE ADVANCES 2022; 8:eabo4271. [PMID: 35704572 PMCID: PMC9200274 DOI: 10.1126/sciadv.abo4271] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023]
Abstract
Infection is one of the major causes of mortality in patients with systemic lupus erythematosus (SLE). We previously found that CD38, an ectoenzyme that regulates the production of NAD+, is up-regulated in CD8+ T cells of SLE patients and correlates with the risk of infection. Here, we report that CD38 reduces CD8+ T cell function by negatively affecting mitochondrial fitness through the inhibition of multiple steps of mitophagy, a process that is critical for mitochondria quality control. Using a murine lupus model, we found that administration of a CD38 inhibitor in a CD8+ T cell-targeted manner reinvigorated their effector function, reversed the defects in autophagy and mitochondria, and improved viral clearance. We conclude that CD38 represents a target to mitigate infection rates in people with SLE.
Collapse
Affiliation(s)
- Ping-Min Chen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Eri Katsuyama
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Abhigyan Satyam
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hao Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jose Rubio
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sungwook Jung
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Maria G. Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - George C. Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
31
|
dos Santos CC, Walburg KV, van Veen S, Wilson LG, Trufen CEM, Nascimento IP, Ottenhoff THM, Leite LCC, Haks MC. Recombinant BCG-LTAK63 Vaccine Candidate for Tuberculosis Induces an Inflammatory Profile in Human Macrophages. Vaccines (Basel) 2022; 10:vaccines10060831. [PMID: 35746439 PMCID: PMC9227035 DOI: 10.3390/vaccines10060831] [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: 04/07/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 11/29/2022] Open
Abstract
Tuberculosis (TB) is one of the top 10 leading causes of death worldwide. The recombinant BCG strain expressing the genetically detoxified A subunit of the thermolabile toxin from Escherichia coli (LTAK63) adjuvant (rBCG-LTAK63) has previously been shown to confer superior protection and immunogenicity compared to BCG in a murine TB infection model. To further investigate the immunological mechanisms induced by rBCG-LTAK63, we evaluated the immune responses induced by rBCG-LTAK63, BCG, and Mycobacterium tuberculosis (Mtb) H37Rv strains in experimental infections of primary human M1 and M2 macrophages at the transcriptomic and cytokine secretion levels. The rBCG-LTAK63-infected M1 macrophages more profoundly upregulated interferon-inducible genes such as IFIT3, OAS3, and antimicrobial gene CXCL9 compared to BCG, and induced higher levels of inflammatory cytokines such as IL-12(p70), TNF-β, and IL-15. The rBCG-LTAK63-infected M2 macrophages more extensively upregulated transcripts of inflammation-related genes, TAP1, GBP1, SLAMF7, TNIP1, and IL6, and induced higher levels of cytokines related to inflammation and tissue repair, MCP-3 and EGF, as compared to BCG. Thus, our data revealed an important signature of immune responses induced in human macrophages by rBCG-LTAK63 associated with increased inflammation, activation, and tissue repair, which may be correlated with a protective immune response against TB.
Collapse
Affiliation(s)
- Carina C. dos Santos
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil;
- Programa de Pós-Graduação Interunidades em Biotecnologia, Universidade de São Paulo, São Paulo 05508-900, Brazil
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (K.V.W.); (S.v.V.); (L.G.W.); (T.H.M.O.); (M.C.H.)
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador 40170-115, Brazil
- Correspondence: (C.C.d.S.); (L.C.C.L.)
| | - Kimberley V. Walburg
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (K.V.W.); (S.v.V.); (L.G.W.); (T.H.M.O.); (M.C.H.)
| | - Suzanne van Veen
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (K.V.W.); (S.v.V.); (L.G.W.); (T.H.M.O.); (M.C.H.)
| | - Louis G. Wilson
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (K.V.W.); (S.v.V.); (L.G.W.); (T.H.M.O.); (M.C.H.)
| | | | - Ivan P. Nascimento
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (K.V.W.); (S.v.V.); (L.G.W.); (T.H.M.O.); (M.C.H.)
| | - Luciana C. C. Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil;
- Programa de Pós-Graduação Interunidades em Biotecnologia, Universidade de São Paulo, São Paulo 05508-900, Brazil
- Correspondence: (C.C.d.S.); (L.C.C.L.)
| | - Mariëlle C. Haks
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (K.V.W.); (S.v.V.); (L.G.W.); (T.H.M.O.); (M.C.H.)
| |
Collapse
|
32
|
Akama-Garren EH, Carroll MC. T Cell Help in the Autoreactive Germinal Center. Scand J Immunol 2022; 95:e13192. [PMID: 35587582 DOI: 10.1111/sji.13192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/29/2022]
Abstract
The germinal center serves as a site of B cell selection and affinity maturation, critical processes for productive adaptive immunity. In autoimmune disease tolerance is broken in the germinal center reaction, leading to production of autoreactive B cells that may propagate disease. Follicular T cells are crucial regulators of this process, providing signals necessary for B cell survival in the germinal center. Here we review the emerging roles of follicular T cells in the autoreactive germinal center. Recent advances in immunological techniques have allowed study of the gene expression profiles and repertoire of follicular T cells at unprecedented resolution. These studies provide insight into the potential role follicular T cells play in preventing or facilitating germinal center loss of tolerance. Improved understanding of the mechanisms of T cell help in autoreactive germinal centers provides novel therapeutic targets for diseases of germinal center dysfunction.
Collapse
Affiliation(s)
- Elliot H Akama-Garren
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard-MIT Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Michael C Carroll
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
33
|
Akama-Garren EH, Carroll MC. Lupus Susceptibility Loci Predispose Mice to Clonal Lymphocytic Responses and Myeloid Expansion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2403-2424. [PMID: 35477687 PMCID: PMC9254690 DOI: 10.4049/jimmunol.2200098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/14/2022] [Indexed: 05/17/2023]
Abstract
Lupus susceptibility results from the combined effects of numerous genetic loci, but the contribution of these loci to disease pathogenesis has been difficult to study due to the large cellular heterogeneity of the autoimmune immune response. We performed single-cell RNA, BCR, and TCR sequencing of splenocytes from mice with multiple polymorphic lupus susceptibility loci. We not only observed lymphocyte and myeloid expansion, but we also characterized changes in subset frequencies and gene expression, such as decreased CD8 and marginal zone B cells and increased Fcrl5- and Cd5l-expressing macrophages. Clonotypic analyses revealed expansion of B and CD4 clones, and TCR repertoires from lupus-prone mice were distinguishable by algorithmic specificity prediction and unsupervised machine learning classification. Myeloid differential gene expression, metabolism, and altered ligand-receptor interaction were associated with decreased Ag presentation. This dataset provides novel mechanistic insight into the pathophysiology of a spontaneous model of lupus, highlighting potential therapeutic targets for autoantibody-mediated disease.
Collapse
Affiliation(s)
- Elliot H Akama-Garren
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA; and
- Harvard-MIT Health Sciences and Technology, Harvard Medical School, Boston, MA
| | - Michael C Carroll
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA; and
| |
Collapse
|
34
|
Humbel M, Bellanger F, Horisberger A, Suffiotti M, Fluder N, Makhmutova M, Mathias A, Du Pasquier R, Fenwick C, Ribi C, Comte D. SLAMF Receptor Expression Identifies an Immune Signature That Characterizes Systemic Lupus Erythematosus. Front Immunol 2022; 13:843059. [PMID: 35603218 PMCID: PMC9120573 DOI: 10.3389/fimmu.2022.843059] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/11/2022] [Indexed: 11/25/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease of unknown etiology, linked to alterations in both the innate and the adaptive immune system. Due to the heterogeneity of the clinical presentation, the diagnosis of SLE remains complicated and is often made years after the first symptoms manifest, delaying treatment, and worsening the prognosis. Several studies have shown that signaling lymphocytic activation molecule family (SLAMF) receptors are aberrantly expressed and dysfunctional in SLE immune cells, contributing to the altered cellular function observed in these patients. The aim of this study was to determine whether altered co-/expression of SLAMF receptors on peripheral blood mononuclear cells (PBMC) identifies SLE characteristic cell populations. To this end, single cell mass cytometry and bioinformatic analysis were exploited to compare SLE patients to healthy and autoimmune diseases controls. First, the expression of each SLAMF receptor on all PBMC populations was investigated. We observed that SLAMF1+ B cells (referred to as SLEB1) were increased in SLE compared to controls. Furthermore, the frequency of SLAMF4+ monocytes and SLAMF4+ NK were inversely correlated with disease activity, whereas the frequency SLAMF1+ CD4+ TDEM cells were directly correlated with disease activity. Consensus clustering analysis identified two cell clusters that presented significantly increased frequency in SLE compared to controls: switch memory B cells expressing SLAMF1, SLAMF3, SLAMF5, SLAMF6 (referred to as SLESMB) and circulating T follicular helper cells expressing the same SLAMF receptors (referred to as SLEcTFH). Finally, the robustness of the identified cell populations as biomarkers for SLE was evaluated through ROC curve analysis. The combined measurement of SLEcTFH and SLEB1 or SLESMB cells identified SLE patients in 90% of cases. In conclusion, this study identified an immune signature for SLE based on the expression of SLAMF receptors on PBMC, further highlighting the involvement of SLAMF receptors in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Morgane Humbel
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Florence Bellanger
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Alice Horisberger
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Madeleine Suffiotti
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Natalia Fluder
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Mariko Makhmutova
- Machine Learning and Optimization Laboratory, Swiss Federal Institute of Technology in Lausanne, Lausanne, Switzerland
| | - Amandine Mathias
- Service of Neurology, Department of Clinical Neurosciences, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Renaud Du Pasquier
- Service of Neurology, Department of Clinical Neurosciences, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Craig Fenwick
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Camillo Ribi
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Denis Comte
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
35
|
Iwata S, Tanaka Y. Association of Viral Infection With the Development and Pathogenesis of Systemic Lupus Erythematosus. Front Med (Lausanne) 2022; 9:849120. [PMID: 35280878 PMCID: PMC8914279 DOI: 10.3389/fmed.2022.849120] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/03/2022] [Indexed: 12/27/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that causes multiple organ damage in women of childbearing age and has a relapsing-remitting course. SLE is caused by the interaction between genetic and environmental factors, however, its underlying triggers remain unknown. Among the environmental factors, the involvement of infections as a trigger for SLE, especially those of viral etiology, has been widely reported. Human endogenous retroviruses (HERVs) may put patients at a genetic predisposition to SLE, while the Epstein-Barr virus (EBV) may play a role as an environmental factor that triggers the development of SLE. It has been suggested that EBV-infected B-cells may become resistant to apoptosis, resulting in the activation, proliferation, and antibody production of autoreactive B-cells, which cause tissue damage in SLE. However, the interaction between the virus and immune cells, as well as the impact of the virus on the differentiation and dysfunction of immune cells, remain unclear. In this review, we focus on the relationship between the development and pathogenesis of SLE and viral infections, as well as the mechanism of SLE exacerbation via activation of immune cells, such as B-cells, based on the latest findings.
Collapse
|
36
|
CD4 T cell help prevents CD8 T cell exhaustion and promotes control of Mycobacterium tuberculosis infection. Cell Rep 2021; 36:109696. [PMID: 34525366 PMCID: PMC8466141 DOI: 10.1016/j.celrep.2021.109696] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/09/2021] [Accepted: 08/19/2021] [Indexed: 11/23/2022] Open
Abstract
CD4 T cells are essential for immunity to tuberculosis because they produce cytokines, including interferon-γ. Whether CD4 T cells act as "helper" cells to promote optimal CD8 T cell responses during Mycobacterium tuberculosis is unknown. Using two independent models, we show that CD4 T cell help enhances CD8 effector functions and prevents CD8 T cell exhaustion. We demonstrate synergy between CD4 and CD8 T cells in promoting the survival of infected mice. Purified helped, but not helpless, CD8 T cells efficiently restrict intracellular bacterial growth in vitro. Thus, CD4 T cell help plays an essential role in generating protective CD8 T cell responses against M. tuberculosis infection in vitro and in vivo. We infer vaccines that elicit both CD4 and CD8 T cells are more likely to be successful than vaccines that elicit only CD4 or CD8 T cells.
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is a serious autoimmune disease with a wide range of organ involvement. In addition to aberrant B-cell responses leading to autoantibody production, T-cell abnormalities are important in the induction of autoimmunity and the ensuing downstream organ damage. In this article, we present an update on how subsets of CD8+ T cells contribute to SLE pathogenesis. RECENT FINDINGS Reduced cytolytic function of CD8+ T cells not only promotes systemic autoimmunity but also accounts for the increased risk of infections. Additional information suggests that effector functions of tissue CD8+ T cells contribute to organ damage. The phenotypic changes in tissue CD8+ T cells likely arise from exposure to tissue microenvironment and crosstalk with tissue resident cells. Research on pathogenic IL-17-producing double negative T cells also suggests their origin from autoreactive CD8+ T cells, which also contribute to the induction and maintenance of systemic autoimmunity. SUMMARY Reduced CD8+ T-cell effector function illustrates their role in peripheral tolerance in the control of autoimmunity and to the increased risk of infections. Inflammatory cytokine producing double negative T cells and functional defects of regulatory CD8+ T cell both contribute to SLE pathogenesis. Further in depth research on these phenotypic changes are warranted for the development of new therapeutics for people with SLE.
Collapse
|
38
|
Paredes JL, Fernandez-Ruiz R, Niewold TB. T Cells in Systemic Lupus Erythematosus. Rheum Dis Clin North Am 2021; 47:379-393. [PMID: 34215369 DOI: 10.1016/j.rdc.2021.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
T-cell dysregulation has been implicated in the loss of tolerance and overactivation of B cells in systemic lupus erythematosus (SLE). Recent studies have identified T-cell subsets and genetic, epigenetic, and environmental factors that contribute to pathogenic T-cell differentiation, as well as disease pathogenesis and clinical phenotypes in SLE. Many therapeutics targeting T-cell pathways are under development, and although many have not progressed in clinical trials, the recent approval of the calcineurin inhibitor voclosporin is encouraging. Further study of T-cell subsets and biomarkers of T-cell action may pave the way for specific targeting of pathogenic T-cell populations in SLE.
Collapse
Affiliation(s)
- Jacqueline L Paredes
- Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Avenue, New York, NY 10016, USA
| | - Ruth Fernandez-Ruiz
- Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Avenue, New York, NY 10016, USA; Division of Rheumatology, NYU Grossman School of Medicine, 550 1st Avenue, New York, NY 10016, USA
| | - Timothy B Niewold
- Colton Center for Autoimmunity, NYU Grossman School of Medicine, 550 1st Avenue, New York, NY 10016, USA.
| |
Collapse
|
39
|
Dörner T, Szelinski F, Lino AC, Lipsky PE. Therapeutic implications of the anergic/postactivated status of B cells in systemic lupus erythematosus. RMD Open 2021; 6:rmdopen-2020-001258. [PMID: 32675278 PMCID: PMC7425190 DOI: 10.1136/rmdopen-2020-001258] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/28/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is characterised by numerous abnormalities in B lineage cells, including increased CD27++ plasmablasts/plasma cells, atypical CD27-IgD- B cells with increased CD95, spleen tyrosine kinase (Syk)++, CXCR5- and CXCR5+ subsets and anergic CD11c+Tbet+ age-associated B cells. Most findings, together with preclinical lupus models, support the concept of B cell hyperactivity in SLE. However, it remains largely unknown whether these specific B cell subsets have pathogenic consequences and whether they provide relevant therapeutic targets. Recent findings indicate a global distortion of B cell functional capability, in which the entire repertoire of naïve and memory B cells in SLE exhibits an anergic or postactivated (APA) functional phenotype. The APA status of SLE B cells has some similarities to the functional derangement of lupus T cells. APA B cells are characterised by reduced global cytokine production, diminished B cell receptor (BCR) signalling with decreased Syk and Bruton's tyrosine kinase phosphorylation related to repeated in vivo BCR stimulation as well as hyporesponsiveness to toll-like receptor 9 engagement, but intact CD40 signalling. This APA status was related to constitutive co-localisation of CD22 linked to phosphatase SHP-1 and increased overall protein phosphatase activities. Notably, CD40 co-stimulation could revert this APA status and restore BCR signalling, downregulate protein tyrosine phosphatase transcription and promote B cell proliferation and differentiation. The APA status and their potential rescue by bystander help conveyed through CD40 stimulation not only provides insights into possible mechanisms of escape of autoreactive clones from negative selection but also into novel ways to target B cells therapeutically.
Collapse
Affiliation(s)
| | | | - Andreia C Lino
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Peter E Lipsky
- RILITE Research Institute, Charlottesville, Virginia, USA
| |
Collapse
|
40
|
Gartshteyn Y, Askanase AD, Mor A. SLAM Associated Protein Signaling in T Cells: Tilting the Balance Toward Autoimmunity. Front Immunol 2021; 12:654839. [PMID: 33936082 PMCID: PMC8086963 DOI: 10.3389/fimmu.2021.654839] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/25/2021] [Indexed: 11/13/2022] Open
Abstract
T cell activation is the result of the integration of signals across the T cell receptor and adjacent co-receptors. The signaling lymphocyte activation molecules (SLAM) family are transmembrane co-receptors that modulate antigen driven T cell responses. Signal transduction downstream of the SLAM receptor is mediated by the adaptor protein SLAM Associated Protein (SAP), a small intracellular protein with a single SH2 binding domain that can recruit tyrosine kinases as well as shield phosphorylated sites from dephosphorylation. Balanced SLAM-SAP signaling within T cells is required for healthy immunity, with deficiency or overexpression prompting autoimmune diseases. Better understanding of the molecular pathways involved in the intracellular signaling downstream of SLAM could provide treatment targets for these autoimmune diseases.
Collapse
Affiliation(s)
- Yevgeniya Gartshteyn
- Division of Rheumatology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Anca D Askanase
- Division of Rheumatology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Adam Mor
- Division of Rheumatology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States.,Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
41
|
Humbel M, Bellanger F, Fluder N, Horisberger A, Suffiotti M, Fenwick C, Ribi C, Comte D. Restoration of NK Cell Cytotoxic Function With Elotuzumab and Daratumumab Promotes Elimination of Circulating Plasma Cells in Patients With SLE. Front Immunol 2021; 12:645478. [PMID: 33828555 PMCID: PMC8019934 DOI: 10.3389/fimmu.2021.645478] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/26/2021] [Indexed: 11/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease characterized by multiple cellular and molecular dysfunctions of the innate and adaptive immunity. Cytotoxic function of NK cells is compromised in patients with SLE. Herein, we characterized the phenotypic alterations of SLE NK cells in a comprehensive manner to further delineate the mechanisms underlying the cytotoxic dysfunction of SLE NK cells and identify novel potential therapeutic targets. Therefore, we examined PBMC from SLE patients and matched healthy controls by single-cell mass cytometry to assess the phenotype of NK cells. In addition, we evaluated the cell function of NK cells (degranulation and cytokine production) and the killing of B cell subpopulations in a B cell-NK cell in vitro co-culture model. We found that SLE NK cells expressed higher levels of CD38 and were not able to adequately upregulate SLAMF1 and SLAMF7 following activation. In addition, ligation of SLAMF7 with elotuzumab or of CD38 with daratumumab on SLE NK cells enhanced degranulation of both healthy and SLE NK cells and primed them to kill circulating plasma cells in an in vitro co-culture system. Overall, our data indicated that dysregulated expression of CD38, SLAMF1 and SLAMF7 on SLE NK cells is associated with an altered interplay between SLE NK cells and plasma cells, thus suggesting their contribution to the accumulation of (auto)antibody producing cells. Accordingly, targeting SLAMF7 and CD38 may represent novel therapeutic approaches in SLE by enhancing NK cell function and promoting elimination of circulating plasma cell.
Collapse
Affiliation(s)
- Morgane Humbel
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Florence Bellanger
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Natalia Fluder
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Alice Horisberger
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Madeleine Suffiotti
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Craig Fenwick
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Camillo Ribi
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Denis Comte
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
42
|
Chen PM, Tsokos GC. T Cell Abnormalities in the Pathogenesis of Systemic Lupus Erythematosus: an Update. Curr Rheumatol Rep 2021; 23:12. [PMID: 33512577 DOI: 10.1007/s11926-020-00978-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2020] [Indexed: 12/01/2022]
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus is a complex disease with broad spectrum of clinical manifestations. In addition to abnormal B cell responsive leading to autoantibody production, various T cells also play different roles in promoting systemic autoimmunity and end organ damage. We aim to provide a review on recent developments in how abnormalities in different T cells subsets contribute to systemic lupus erythematosus pathogenesis and how they inform the consideration of new promising therapeutics. RECENT FINDINGS Distinct subsets of T cells known as T follicular helper cells enable the production of pathogenic autoantibodies. Detailed understanding of the B cell helping T cell subsets should improve the performance of clinical trials targeting the cognate T:B cell interaction. CD8+ T cells play a role in peripheral tolerance and reversal of its exhausted phenotype could potentially alleviate both systemic autoimmunity and the risk of infection. Research on the abnormal lupus T cell signaling also leads to putative therapeutic targets able to restore interleukin-2 production and suppress the production of the pathogenic IL-17 cytokine. Recently, several studies have focused on dissecting T cell populations located in the damaged organs, aiming to target the pathogenic processes specific to each organ. Numerous T cell subsets play distinct roles in SLE pathogenesis and recent research in understanding abnormal signaling pathways, cellular metabolism, and environmental cues pave the way for the development of novel therapeutics.
Collapse
Affiliation(s)
- Ping-Min Chen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
43
|
Katsuyama E, Suarez-Fueyo A, Bradley SJ, Mizui M, Marin AV, Mulki L, Krishfield S, Malavasi F, Yoon J, Sui SJH, Kyttaris VC, Tsokos GC. The CD38/NAD/SIRTUIN1/EZH2 Axis Mitigates Cytotoxic CD8 T Cell Function and Identifies Patients with SLE Prone to Infections. Cell Rep 2021; 30:112-123.e4. [PMID: 31914379 PMCID: PMC7577012 DOI: 10.1016/j.celrep.2019.12.014] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 10/28/2019] [Accepted: 12/05/2019] [Indexed: 12/01/2022] Open
Abstract
Patients with systemic lupus erythematosus (SLE) suffer frequent infections that account for significant morbidity and mortality. T cell cytotoxic responses are decreased in patients with SLE, yet the responsible molecular events are largely unknown. We find an expanded CD8CD38high T cell subset in a sub-group of patients with increased rates of infections. CD8CD38high T cells from healthy subjects and patients with SLE display decreased cytotoxic capacity, degranulation, and expression of granzymes A and B and perforin. The key cytotoxicity-related transcription factors T-bet, RUNX3, and EOMES are decreased in CD8CD38high T cells. CD38 leads to increased acetylated EZH2 through inhibition of the deacetylase Sirtuin1. Acetylated EZH2 represses RUNX3 expression, whereas inhibition of EZH2 restores CD8 T cell cytotoxic responses. We propose that high levels of CD38 lead to decreased CD8 T cell-mediated cytotoxicity and increased propensity to infections in patients with SLE, a process that can be reversed pharmacologically. Katsuyama et al. find that an expanded CD8CD38high T cell population in SLE patients is linked to infections. CD8CD38high T cells display decreased cytotoxic capacity by suppressing the expression of related molecules through an NAD+/Sirtuin1/EZH2 pathway. EZH2 inhibitors increase cytotoxicity offering a means to mitigate infection rates in SLE.
Collapse
Affiliation(s)
- Eri Katsuyama
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Abel Suarez-Fueyo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sean J Bradley
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Masayuki Mizui
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ana V Marin
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lama Mulki
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Suzanne Krishfield
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Fabio Malavasi
- Laboratory of Immunogenetics, Department of Genetics, Biology and Biochemistry, University of Torino, and Fondazione Ricerca Molinette, Torino, Italy
| | - Joon Yoon
- Harvard Chan Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Shannan J Ho Sui
- Harvard Chan Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Vasileios C Kyttaris
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
44
|
SLAMF7 and IL-6R define distinct cytotoxic versus helper memory CD8 + T cells. Nat Commun 2020; 11:6357. [PMID: 33311473 PMCID: PMC7733515 DOI: 10.1038/s41467-020-19002-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 09/18/2020] [Indexed: 12/17/2022] Open
Abstract
The prevailing ‘division of labor’ concept in cellular immunity is that CD8+ T cells primarily utilize cytotoxic functions to kill target cells, while CD4+ T cells exert helper/inducer functions. Multiple subsets of CD4+ memory T cells have been characterized by distinct chemokine receptor expression. Here, we demonstrate that analogous CD8+ memory T-cell subsets exist, characterized by identical chemokine receptor expression signatures and controlled by similar generic programs. Among them, Tc2, Tc17 and Tc22 cells, in contrast to Tc1 and Tc17 + 1 cells, express IL-6R but not SLAMF7, completely lack cytotoxicity and instead display helper functions including CD40L expression. CD8+ helper T cells exhibit a unique TCR repertoire, express genes related to skin resident memory T cells (TRM) and are altered in the inflammatory skin disease psoriasis. Our findings reveal that the conventional view of CD4+ and CD8+ T cell capabilities and functions in human health and disease needs to be revised. We classically consider the T cell compartment divided into cytotoxic CD8+ T cells and multiple, different helper CD4+ T cell subsets. Here the authors demonstrate that distinct memory CD8+ T cell subsets phenotypically inhabit CD4+ T cell like populations including some with helper-like characteristics.
Collapse
|
45
|
O'Connell P, Hyslop S, Blake MK, Godbehere S, Amalfitano A, Aldhamen YA. SLAMF7 Signaling Reprograms T Cells toward Exhaustion in the Tumor Microenvironment. THE JOURNAL OF IMMUNOLOGY 2020; 206:193-205. [PMID: 33288545 DOI: 10.4049/jimmunol.2000300] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 10/26/2020] [Indexed: 12/21/2022]
Abstract
T cell exhaustion represents one of the most pervasive strategies tumors employ to circumvent the immune system. Although repetitive, cognate TCR signaling is recognized as the primary driving force behind this phenomenon, and it remains unknown what other forces drive T cell exhaustion in the tumor microenvironment (TME). In this study, we show that activation of the self-ligand SLAMF7 immune receptor on T cells induced STAT1 and STAT3 phosphorylation, expression of multiple inhibitory receptors, and transcription factors associated with T cell exhaustion. Analysis of The Cancer Genome Atlas revealed that SLAMF7 transcript levels were strongly correlated with various inhibitory receptors and that high SLAMF7 expression was indicative of poor survival in clear cell renal cell carcinoma (ccRCC). Targeted reanalysis of a CyTOF dataset, which profiled the TME in 73 ccRCC patients, revealed cell-type-specific SLAMF7 expression patterns, strong correlations between exhausted T cells and SLAMF7+ tumor-associated macrophages (TAMs), and a unique subset of SLAMF7highCD38high TAMs. These SLAMF7highCD38high TAMs showed the strongest correlations with exhausted T cells and were an independent prognostic factor in ccRCC. Confirmatory ex vivo coculture studies validated that SLAMF7-SLAMF7 interactions between murine TAMs and CD8+ T cells induce expression of multiple inhibitory receptors. Finally, mice lacking SLAMF7 show restricted growth of B16-F10 tumors, and CD8+ T cells from these mice express less PD-1 and TOX and exhibited an impaired ability to progress through the exhaustion developmental trajectory to terminal exhaustion. These findings suggest that SLAMF7 might play an important role in modulating T cell function in the TME.
Collapse
Affiliation(s)
- Patrick O'Connell
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824; and
| | - Sean Hyslop
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824; and
| | - Maja K Blake
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824; and
| | - Sarah Godbehere
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824; and
| | - Andrea Amalfitano
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824; and.,Department of Pediatrics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824
| | - Yasser A Aldhamen
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824; and
| |
Collapse
|
46
|
Higashioka K, Ota Y, Maehara T, Moriyama M, Ayano M, Mitoma H, Akahoshi M, Arinobu Y, Horiuchi T, Nakamura S, Akashi K, Niiro H. Association of circulating SLAMF7 +Tfh1 cells with IgG4 levels in patients with IgG4-related disease. BMC Immunol 2020; 21:31. [PMID: 32487061 PMCID: PMC7268355 DOI: 10.1186/s12865-020-00361-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Follicular helper CD4+ T (Tfh) cells have a critical role in IgG4 production by B cells in IgG4-related disease (IgG4-RD). Recent studies including ours showed that SLAMF7+CD4+ T cells are an important pathological driver of IgG4-RD. In this study, we have sought to elucidate a relationship between helper CD4+ T (Th), particularly Tfh, cells and SLAMF7+ CD4+ T cells in IgG4-RD. RESULTS The patients with IgG4-RD enrolled in this study were aged 66 ± 12 years and their titers of serum IgG4 were 372 ± 336 mg/dl. Th1 cells, activated circulating Tfh1 (cTfh1), and activated cTfh2 cells increased in IgG4-RD. SLAMF7 was mainly expressed on Th1 and cTfh1, but not cTfh2, cells in the patients. SLAMF7+ cTfh1 cells were PD-1/CD28 double-positive, whereas SLAMF7+ Th1 cells were CD28 negative. Positive correlations were noted between serum IgG4 levels and the number of activated cTfh2 cells and SLAMF7+ cTfh1 cells, but not SLAMF7+ Th1 cells. Intriguingly, among cTfh1 cells, activated SLAMF7+ cTfh1 cells were high producers of IL-10 along with IL-21. Blimp-1, but not Bcl-6, mRNA was expressed at high levels in activated SLAMF7+ cTfh1 cells. In addition to CD4+ T cells, the frequency of SLAMF7+ fraction was higher in memory B cells than naïve B cells in patients with IgG4RD. Finally, upon stimulation via B-cell receptor and CD40, Tfh1-associated cytokines, IL-21 and IFN-γ, most significantly induced SLAMF7 expression in memory B cells. CONCLUSIONS Together, these results suggest that circulating SLAMF7+ Tfh1 cells, along with Tfh2 cells, play a pathologic role in IgG4 production in IgG4-RD.
Collapse
Affiliation(s)
- Kazuhiko Higashioka
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuri Ota
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takashi Maehara
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masafumi Moriyama
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masahiro Ayano
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroki Mitoma
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Mitsuteru Akahoshi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yojiro Arinobu
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takahiko Horiuchi
- Department of Internal Medicine, Kyushu University Beppu Hospital, 4546 Tsurumihara, Beppu, Oita, 874-0838, Japan
| | - Seiji Nakamura
- Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroaki Niiro
- Department of Medical Education, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
47
|
JACKSON KAYLAC, SUN KATHERINE, BARBOUR CHRISTOPHER, HERNANDEZ DENA, KOSA PETER, TANIGAWA MAKOTO, WEIDEMAN ANNMARIE, BIELEKOVA BIBIANA. Genetic model of MS severity predicts future accumulation of disability. Ann Hum Genet 2020; 84:1-10. [PMID: 31396954 PMCID: PMC6898742 DOI: 10.1111/ahg.12342] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 02/06/2023]
Abstract
No genetic modifiers of multiple sclerosis (MS) severity have been independently validated, leading to a lack of insight into genetic determinants of the rate of disability progression. We investigated genetic modifiers of MS severity in prospectively acquired training (N = 205) and validation (N = 94) cohorts, using the following advances: (1) We focused on 113 genetic variants previously identified as related to MS severity; (2) We used a novel, sensitive outcome: MS Disease Severity Scale (MS-DSS); (3) Instead of validating individual alleles, we used a machine learning technique (random forest) that captures linear and complex nonlinear effects between alleles to derive a single Genetic Model of MS Severity (GeM-MSS). The GeM-MSS consists of 19 variants located in vicinity of 12 genes implicated in regulating cytotoxicity of immune cells, complement activation, neuronal functions, and fibrosis. GeM-MSS correlates with MS-DSS (r = 0.214; p = 0.043) in a validation cohort that was not used in the modeling steps. The recognized biology identifies novel therapeutic targets for inhibiting MS disability progression.
Collapse
Affiliation(s)
- KAYLA C. JACKSON
- Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - KATHERINE SUN
- Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - CHRISTOPHER BARBOUR
- Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
- Department of Mathematical Sciences, Montana State University, Bozeman, MT
| | - DENA HERNANDEZ
- Laboratory of Neurogenetics, National Institute of Aging, National Institutes of Health, Bethesda, MD
| | - PETER KOSA
- Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - MAKOTO TANIGAWA
- Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - ANN MARIE WEIDEMAN
- Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - BIBIANA BIELEKOVA
- Neuroimmunological Diseases Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
48
|
Karampetsou MP, Comte D, Suárez-Fueyo A, Katsuyama E, Yoshida N, Kono M, Kyttaris VC, Tsokos GC. Signaling Lymphocytic Activation Molecule Family Member 1 Engagement Inhibits T Cell-B Cell Interaction and Diminishes Interleukin-6 Production and Plasmablast Differentiation in Systemic Lupus Erythematosus. Arthritis Rheumatol 2019; 71:99-108. [PMID: 30058241 DOI: 10.1002/art.40682] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/26/2018] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Signaling lymphocytic activation molecule family member 1 (SLAMF1) homophilic interactions promote immunoglobulin production and T cell-B cell cross-talk. SLAMF1 is overexpressed on T and B cells in patients with systemic lupus erythematosus (SLE). This study was undertaken to determine the role of SLAMF1 monoclonal antibody (mAb) in modulating T cell-B cell interaction and B cell activation. METHODS Anti-IgM-prestimulated naive or total B cells from either healthy donors or patients with SLE were cocultured with autologous T cells under CD3/CD28 stimulation, in the presence or absence of the SLAMF1 mAb. Naive B cells were stimulated with anti-IgM and CD40L in the presence of the SLAMF1 antibody. Cytokine production by CD4+ T cells and B cells was examined by flow cytometry and/or quantitative polymerase chain reaction. Plasmablast formation and T cell and B cell conjugates were assessed by flow cytometry. IgG and antinuclear antibody production was determined by enzyme-linked immunosorbent assay. RESULTS SLAMF1 ligation in a human peripheral blood T cell-B cell culture system reduced the following in both healthy controls and patients with SLE: conjugate formation, interleukin-6 (IL-6) production by B cells, IL-21 and IL-17A production by T cells, and Ig and autoantibody production. Whereas the SLAMF1 mAb directly affected the function of isolated peripheral B cells by decreasing IL-6 and Ig production in vitro, it did not affect cytokine production by isolated T cells stimulated in vitro. CONCLUSION The SLAMF1 antibody inhibits T cell-B cell interaction and suppresses B cell cytokine production and differentiation, thereby acting as a potential therapeutic tool in the treatment of patients with SLE.
Collapse
Affiliation(s)
- Maria P Karampetsou
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Denis Comte
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, and Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Abel Suárez-Fueyo
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Eri Katsuyama
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Nobuya Yoshida
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Michihito Kono
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Vasileios C Kyttaris
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - George C Tsokos
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
49
|
Weißenberg SY, Szelinski F, Schrezenmeier E, Stefanski AL, Wiedemann A, Rincon-Arevalo H, Welle A, Jungmann A, Nordström K, Walter J, Imgenberg-Kreuz J, Nordmark G, Rönnblom L, Bachali P, Catalina MD, Grammer AC, Lipsky PE, Lino AC, Dörner T. Identification and Characterization of Post-activated B Cells in Systemic Autoimmune Diseases. Front Immunol 2019; 10:2136. [PMID: 31616406 PMCID: PMC6768969 DOI: 10.3389/fimmu.2019.02136] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/27/2019] [Indexed: 12/16/2022] Open
Abstract
Autoimmune diseases (AID) such as systemic lupus erythematosus (SLE), primary Sjögren's syndrome (pSS), and rheumatoid arthritis (RA) are chronic inflammatory diseases in which abnormalities of B cell function play a central role. Although it is widely accepted that autoimmune B cells are hyperactive in vivo, a full understanding of their functional status in AID has not been delineated. Here, we present a detailed analysis of the functional capabilities of AID B cells and dissect the mechanisms underlying altered B cell function. Upon BCR activation, decreased spleen tyrosine kinase (Syk) and Bruton's tyrosine kinase (Btk) phosphorylation was noted in AID memory B cells combined with constitutive co-localization of CD22 and protein tyrosine phosphatase (PTP) non-receptor type 6 (SHP-1) along with hyporesponsiveness to TLR9 signaling, a Syk-dependent response. Similar BCR hyporesponsiveness was also noted specifically in SLE CD27− B cells together with increased PTP activities and increased transcripts for PTPN2, PTPN11, PTPN22, PTPRC, and PTPRO in SLE B cells. Additional studies revealed that repetitive BCR stimulation of normal B cells can induce BCR hyporesponsiveness and that tissue-resident memory B cells from AID patients also exhibited decreased responsiveness immediately ex vivo, suggesting that the hyporesponsive status can be acquired by repeated exposure to autoantigen(s) in vivo. Functional studies to overcome B cell hyporesponsiveness revealed that CD40 co-stimulation increased BCR signaling, induced proliferation, and downregulated PTP expression (PTPN2, PTPN22, and receptor-type PTPs). The data support the conclusion that hyporesponsiveness of AID and especially SLE B cells results from chronic in vivo stimulation through the BCR without T cell help mediated by CD40–CD154 interaction and is manifested by decreased phosphorylation of BCR-related proximal signaling molecules and increased PTPs. The hyporesponsiveness of AID B cells is similar to a form of functional anergy.
Collapse
Affiliation(s)
- Sarah Y Weißenberg
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Franziska Szelinski
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Eva Schrezenmeier
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
| | - Ana-Luisa Stefanski
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
| | - Annika Wiedemann
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
| | - Hector Rincon-Arevalo
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany.,Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Instituto de Investigaciones Médicas, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Anna Welle
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Annemarie Jungmann
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Karl Nordström
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Jörn Walter
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Juliana Imgenberg-Kreuz
- Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Gunnel Nordmark
- Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Rönnblom
- Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | | | - Amrie C Grammer
- RILITE Research Institute, Charlottesville, VA, United States
| | - Peter E Lipsky
- RILITE Research Institute, Charlottesville, VA, United States
| | - Andreia C Lino
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| |
Collapse
|
50
|
Affiliation(s)
- Tetsuji Sawada
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Daiki Fujimori
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Yusuke Yamamoto
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| |
Collapse
|