1
|
Li X, Zhu L. Comprehensive profiling of cell type-specific expression and distribution of complement genes in mouse and human kidneys: insights into normal physiology and response to kidney transplantations. Ren Fail 2025; 47:2471568. [PMID: 40015727 PMCID: PMC11869339 DOI: 10.1080/0886022x.2025.2471568] [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: 07/24/2024] [Revised: 01/24/2025] [Accepted: 02/15/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Recent studies innovatively revealed the localized expression of complement genes in kidneys and shed light on the vital roles of the intracellular complement system in the physiologic function and pathological conditions. However, a comprehensive analysis of the expression of complement genes in the context of the evolving cellular landscape of the kidney is not available. METHODS We analyzed single-cell RNA sequencing data from healthy human subjects, C57BL/6 mice, and kidney transplant-rejected mice. The data were sourced from the NCBI Gene Expression Omnibus and processed using quality control measures and unsupervised clustering. Differential gene analyses were based on expression levels. RESULTS In total, 50 complement genes were categorized into pattern recognition molecules, proteases, complement components, receptors, and regulators. In normal mice kidneys, complement genes were expressed at relatively low levels. Among different complement gene categories, receptor genes were most widely expressed in kidney cells. Comparatively, macrophages and mesangial cells are the most abundant immune and nonimmune cell types for complement gene expression. A comparison of human and mouse data showed similar expression patterns, but human kidney complement gene expression was more abundant. Comparative analysis between mouse transplant-rejected and normal kidneys demonstrated stronger complement gene expression in transplant-rejected kidneys. CONCLUSIONS This study illustrated significant similarities in complement gene expression between murine and human kidneys and highlighted the responsive nature of complement genes to kidney injury, underscoring the dynamic nature of local complement regulation. These findings enhance our understanding of the complex regulation of the complement system within the kidney, offering insights into its role in renal disease pathogenesis.
Collapse
Affiliation(s)
- Xianzhi Li
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Li Zhu
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease (Peking University), National Health Commission, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| |
Collapse
|
2
|
Ratajczak MZ, Konopko A, Jarczak J, Kazek M, Ratajczak J, Kucia M. Complosome as a new intracellular regulatory network in both normal and malignant hematopoiesis. Leukemia 2025:10.1038/s41375-025-02613-7. [PMID: 40269269 DOI: 10.1038/s41375-025-02613-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/25/2025]
Abstract
Hematopoietic cells and lymphocytes arise from a common stem cell for both lineages. This explains why similar signaling networks regulate the development and biological functions of these cells. One crucial regulatory mechanism involves interactions with soluble mediators of innate immunity, including activated elements of the complement cascade (ComC). For many years, ComC proteins were thought to be synthesized only in the liver and released into blood to be activated by one of the three proteolytic cascades. The regulatory effects of activated components of ComC on hematopoietic stem progenitor cells (HSPCs) and mature hematopoietic cells have been well demonstrated in the past. However, recent data indicate that complement proteins are also expressed in several cell types, including lymphocytes and innate immune cells. This intracellular complement network has been named the "complosome." Recent evidence from our group shows that the complosome is also expressed in HSPCs and plays an important yet underappreciated role in the expansion, trafficking, and metabolism of these cells. We propose that the complosome, like its role in lymphocytes, is necessary for the optimal function of mitochondria in hematopoietic cells, including HSPCs. This opens a new area for investigation and potential pharmacological intervention into the complosome network in normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Department of Regenerative Medicine Warsaw Medical University, Warsaw, Poland.
- Stem Cell Institute at Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
| | - Adrian Konopko
- Department of Regenerative Medicine Warsaw Medical University, Warsaw, Poland
| | - Justyna Jarczak
- Department of Regenerative Medicine Warsaw Medical University, Warsaw, Poland
| | - Michalina Kazek
- Department of Regenerative Medicine Warsaw Medical University, Warsaw, Poland
| | - Janina Ratajczak
- Stem Cell Institute at Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Magdalena Kucia
- Department of Regenerative Medicine Warsaw Medical University, Warsaw, Poland
| |
Collapse
|
3
|
van Haaren MJH, Steller LB, Vastert SJ, Calis JJA, van Loosdregt J. Get Spliced: Uniting Alternative Splicing and Arthritis. Int J Mol Sci 2024; 25:8123. [PMID: 39125692 PMCID: PMC11311815 DOI: 10.3390/ijms25158123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Immune responses demand the rapid and precise regulation of gene protein expression. Splicing is a crucial step in this process; ~95% of protein-coding gene transcripts are spliced during mRNA maturation. Alternative splicing allows for distinct functional regulation, as it can affect transcript degradation and can lead to alternative functional protein isoforms. There is increasing evidence that splicing can directly regulate immune responses. For several genes, immune cells display dramatic changes in isoform-level transcript expression patterns upon activation. Recent advances in long-read RNA sequencing assays have enabled an unbiased and complete description of transcript isoform expression patterns. With an increasing amount of cell types and conditions that have been analyzed with such assays, thousands of novel transcript isoforms have been identified. Alternative splicing has been associated with autoimmune diseases, including arthritis. Here, GWASs revealed that SNPs associated with arthritis are enriched in splice sites. In this review, we will discuss how alternative splicing is involved in immune responses and how the dysregulation of alternative splicing can contribute to arthritis pathogenesis. In addition, we will discuss the therapeutic potential of modulating alternative splicing, which includes examples of spliceform-based biomarkers for disease severity or disease subtype, splicing manipulation using antisense oligonucleotides, and the targeting of specific immune-related spliceforms using antibodies.
Collapse
Affiliation(s)
- Maurice J. H. van Haaren
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Levina Bertina Steller
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Sebastiaan J. Vastert
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Division of Pediatric Rheumatology and Immunology, Wilhelmina Children’s Hospital, 3584 CX Utrecht, The Netherlands
| | - Jorg J. A. Calis
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Jorg van Loosdregt
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
4
|
Nguyen VD, Hughes TR, Zhou Y. From complement to complosome in non-alcoholic fatty liver disease: When location matters. Liver Int 2024; 44:316-329. [PMID: 38010880 DOI: 10.1111/liv.15796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/21/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a growing public health threat and becoming the leading cause of liver transplantation. Nevertheless, no approved specific treatment is currently available for NAFLD. The pathogenesis of NAFLD is multifaceted and not yet fully understood. Accumulating evidence suggests a significant role of the complement system in the development and progression of NAFLD. Here, we provide an overview of the complement system, incorporating the novel concept of complosome, and summarise the up-to-date evidence elucidating the association between complement dysregulation and the pathogenesis of NAFLD. In this process, the extracellular complement system is activated through various pathways, thereby directly contributing to, or working together with other immune cells in the disease development and progression. We also introduce the complosome and assess the evidence that implicates its potential influence in NAFLD through its direct impact on hepatocytes or non-parenchymal liver cells. Additionally, we expound upon how complement system and the complosome may exert their effects in relation with hepatic zonation in NAFLD. Furthermore, we discuss the potential therapeutic implications of targeting the complement system, extracellularly and intracellularly, for NAFLD treatment. Finally, we present future perspectives towards a better understanding of the complement system's contribution to NAFLD.
Collapse
Affiliation(s)
- Van-Dien Nguyen
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Timothy R Hughes
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - You Zhou
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
5
|
Hayes CE, Astier AL, Lincoln MR. Vitamin D mechanisms of protection in multiple sclerosis. FELDMAN AND PIKE'S VITAMIN D 2024:1129-1166. [DOI: 10.1016/b978-0-323-91338-6.00051-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
Singh P, Kemper C. Complement, complosome, and complotype: A perspective. Eur J Immunol 2023; 53:e2250042. [PMID: 37120820 PMCID: PMC10613581 DOI: 10.1002/eji.202250042] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/01/2023]
Abstract
Recent rapid progress in key technological advances, including the broader accessibility of single-cell "omic" approaches, have allowed immunologists to gain important novel insights into the contributions of individual immune cells in protective immunity and immunopathologies. These insights also taught us that there is still much to uncover about the (cellular) networks underlying immune responses. For example, in the last decade, studies on a key component of innate immunity, the complement system, have defined intracellularly active complement (the complosome) as a key orchestrator of normal cell physiology. This added an unexpected facet to the biology of complement, which was long considered fully explored. Here, we will summarize succinctly the known activation modes and functions of the complosome and provide a perspective on the origins of intracellular complement. We will also make a case for extending assessments of the complotype, the individual inherited landscape of common variants in complement genes, to the complosome, and for reassessing patients with known serum complement deficiencies for complosome perturbations. Finally, we will discuss where we see current opportunities and hurdles for dissecting the compartmentalization of complement activities toward a better understanding of their contributions to cellular function in health and disease.
Collapse
Affiliation(s)
- Parul Singh
- Complement and Inflammation Research Section, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA
| | - Claudia Kemper
- Complement and Inflammation Research Section, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA
| |
Collapse
|
7
|
West EE, Kemper C. Complosome - the intracellular complement system. Nat Rev Nephrol 2023:10.1038/s41581-023-00704-1. [PMID: 37055581 PMCID: PMC10100629 DOI: 10.1038/s41581-023-00704-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2023] [Indexed: 04/15/2023]
Abstract
The complement system is a recognized pillar of host defence against infection and noxious self-derived antigens. Complement is traditionally known as a serum-effective system, whereby the liver expresses and secretes most complement components, which participate in the detection of bloodborne pathogens and drive an inflammatory reaction to safely remove the microbial or antigenic threat. However, perturbations in normal complement function can cause severe disease and, for reasons that are currently not fully understood, the kidney is particularly vulnerable to dysregulated complement activity. Novel insights into complement biology have identified cell-autonomous and intracellularly active complement - the complosome - as an unexpected central orchestrator of normal cell physiology. For example, the complosome controls mitochondrial activity, glycolysis, oxidative phosphorylation, cell survival and gene regulation in innate and adaptive immune cells, and in non-immune cells, such as fibroblasts and endothelial and epithelial cells. These unanticipated complosome contributions to basic cell physiological pathways make it a novel and central player in the control of cell homeostasis and effector responses. This discovery, together with the realization that an increasing number of human diseases involve complement perturbations, has renewed interest in the complement system and its therapeutic targeting. Here, we summarize the current knowledge about the complosome across healthy cells and tissues, highlight contributions from dysregulated complosome activities to human disease and discuss potential therapeutic implications.
Collapse
Affiliation(s)
- Erin E West
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Complement and Inflammation Research Section (CIRS), Bethesda, MD, USA
| | - Claudia Kemper
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Complement and Inflammation Research Section (CIRS), Bethesda, MD, USA.
| |
Collapse
|
8
|
Santarsiero D, Aiello S. The Complement System in Kidney Transplantation. Cells 2023; 12:cells12050791. [PMID: 36899927 PMCID: PMC10001167 DOI: 10.3390/cells12050791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Kidney transplantation is the therapy of choice for patients who suffer from end-stage renal diseases. Despite improvements in surgical techniques and immunosuppressive treatments, long-term graft survival remains a challenge. A large body of evidence documented that the complement cascade, a part of the innate immune system, plays a crucial role in the deleterious inflammatory reactions that occur during the transplantation process, such as brain or cardiac death of the donor and ischaemia/reperfusion injury. In addition, the complement system also modulates the responses of T cells and B cells to alloantigens, thus playing a crucial role in cellular as well as humoral responses to the allograft, which lead to damage to the transplanted kidney. Since several drugs that are capable of inhibiting complement activation at various stages of the complement cascade are emerging and being developed, we will discuss how these novel therapies could have potential applications in ameliorating outcomes in kidney transplantations by preventing the deleterious effects of ischaemia/reperfusion injury, modulating the adaptive immune response, and treating antibody-mediated rejection.
Collapse
|
9
|
Lupus nephritis with corticosteroid responsiveness: molecular changes of CD46-mediated type 1 regulatory T cells. Pediatr Res 2022; 92:1099-1107. [PMID: 34952938 DOI: 10.1038/s41390-021-01882-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND The engagement of the complement regulatory proteins CD46 and CD3 in human CD4+ T cells induces the type 1 regulatory T cells (Tr1) and interleukin-10 (IL-10) secretion. This study aimed to elucidate the molecular changes of Tr1 cells through CD46 cytoplasmic Cyt1 tail in lupus nephritis (LN) respond to intravenous methylprednisolone (ivMP) therapy. METHODS We enrolled 40 pediatric patients with LN and 30 healthy controls. Clinical characteristics and peripheral blood mononuclear cells were collected before and 3 days after the administration of ivMP. Kidney specimens were taken from five LN and five minimal-change nephrotic syndrome patients. RESULTS We found that defective CD46-mediated T-helper type 1 contraction (IL-10 switching) is present in active LN patients. The ivMP therapy enhanced LN remission, restored the production of IL-10, increased the CD46-Cyt1/Cyt2 ratio, AKT, and cAMP-responsive element-binding protein phosphorylation, and induced migration with the expression of chemokine receptor molecules CCR4, CCR6, and CCR7 of CD3/CD46-activated Tr1 cells. CONCLUSIONS Pharmacologic interventions that alter the patterns of CD46-Cyt1/Cyt2 expression and the secretion of IL-10 by CD3/CD46-activated Tr1 cells can be used in patients with active LN. IMPACT In patients with LN, ivMP was associated with increased IL-10 production and increased CD46-Cyt1/Cyt2 ratio and AKT phosphorylation by Tr1 cells, with enhanced potential to migration in response to CCL17. These results suggest that expression levels of CD46 isoforms Cyt1 and Cyt2 in CD4 + CD46 + Tr1 cells differ in patients with active LN but can be corrected by corticosteroid treatment. Enhancing the expression of functional CD4 + CD46 + Tr1 cells may be a useful therapeutic approach for LN.
Collapse
|
10
|
Qi R, Qin W. Role of Complement System in Kidney Transplantation: Stepping From Animal Models to Clinical Application. Front Immunol 2022; 13:811696. [PMID: 35281019 PMCID: PMC8913494 DOI: 10.3389/fimmu.2022.811696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/31/2022] [Indexed: 12/23/2022] Open
Abstract
Kidney transplantation is a life-saving strategy for patients with end-stage renal diseases. Despite the advances in surgical techniques and immunosuppressive agents, the long-term graft survival remains a challenge. Growing evidence has shown that the complement system, part of the innate immune response, is involved in kidney transplantation. Novel insights highlighted the role of the locally produced and intracellular complement components in the development of inflammation and the alloreactive response in the kidney allograft. In the current review, we provide the updated understanding of the complement system in kidney transplantation. We will discuss the involvement of the different complement components in kidney ischemia-reperfusion injury, delayed graft function, allograft rejection, and chronic allograft injury. We will also introduce the existing and upcoming attempts to improve allograft outcomes in animal models and in the clinical setting by targeting the complement system.
Collapse
Affiliation(s)
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
11
|
Abstract
CD46 is a receptor for human herpesvirus 6A (HHV-6A) and is in some cells also important for infection with HHV-6B. CD46 has several isoforms of which the most commonly expressed can be distinguished by expression of a BC domain or a C domain in a serine-threonine-proline rich (STP) extracellular region. Using a SupT1 CD46 CRISPR-Cas9 knockout model system reconstituted with specific CD46 isoforms, we demonstrated that HHV-6A infection was more efficient when BC-isoforms were expressed as opposed to C-isoforms, measured by higher levels of intracellular viral transcripts and recovery of more progeny virus. Although the B domain contains several O-glycosylations, mutations of Ser and Thr residues did not prevent infection with HHV-6A. The HHV-6A infection was blocked by inhibitors of clathrin-mediated endocytosis. In contrast, infection with HHV-6B was preferentially promoted by C-isoforms mediating fusion-from-without, and this infection was less affected by inhibitors of clathrin-mediated endocytosis. Taken together, HHV-6A preferred BC isoforms, mediating endocytosis, whereas HHV-6B preferred C isoforms, mediating fusion-from-without. This demonstrates that the STP region of CD46 is important for regulating the mode of infection in SupT1 cells and suggests an epigenetic regulation of the host susceptibility to HHV-6A and HHV-6B infection. Importance CD46 is the receptor used by human herpesvirus 6A (HHV-6A) during infection of T cells, but it is also involved in infection of certain T cells by HHV-6B. The gene for CD46 allows expression of several variants of CD46, known as isoforms, but whether the isoforms matter for infection of T cells is unknown. We used a genetic approach to delete CD46 from T cells and reconstituted them with separate isoforms to study these individually. We expressed the isoforms known as BC and C, which are distinguished by the potential inclusion of a B domain in the CD46 molecule. We demonstrate that HHV-6A prefers the BC isoform to infect T cells, and this occurs predominantly by clathrin-mediated endocytosis. In contrast, HHV-6B prefers the C isoform and infects predominantly by fusion-from-without. Thus, CD46 isoforms may affect susceptibility of T cells to infection with HHV-6A and HHV-6B.
Collapse
|
12
|
Freiwald T, Afzali B. Renal diseases and the role of complement: Linking complement to immune effector pathways and therapeutics. Adv Immunol 2021; 152:1-81. [PMID: 34844708 PMCID: PMC8905641 DOI: 10.1016/bs.ai.2021.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The complement system is an ancient and phylogenetically conserved key danger sensing system that is critical for host defense against pathogens. Activation of the complement system is a vital component of innate immunity required for the detection and removal of pathogens. It is also a central orchestrator of adaptive immune responses and a constituent of normal tissue homeostasis. Once complement activation occurs, this system deposits indiscriminately on any cell surface in the vicinity and has the potential to cause unwanted and excessive tissue injury. Deposition of complement components is recognized as a hallmark of a variety of kidney diseases, where it is indeed associated with damage to the self. The provenance and the pathophysiological role(s) played by complement in each kidney disease is not fully understood. However, in recent years there has been a renaissance in the study of complement, with greater appreciation of its intracellular roles as a cell-intrinsic system and its interplay with immune effector pathways. This has been paired with a profusion of novel therapeutic agents antagonizing complement components, including approved inhibitors against complement components (C)1, C3, C5 and C5aR1. A number of clinical trials have investigated the use of these more targeted approaches for the management of kidney diseases. In this review we present and summarize the evidence for the roles of complement in kidney diseases and discuss the available clinical evidence for complement inhibition.
Collapse
Affiliation(s)
- Tilo Freiwald
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, MD, United States; Department of Nephrology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Behdad Afzali
- Department of Nephrology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany.
| |
Collapse
|
13
|
Zeng J, Xu H, Huang C, Sun Y, Xiao H, Yu G, Zhou H, Zhang Y, Yao W, Xiao W, Hu J, Wu L, Xing J, Wang T, Chen Z, Ye Z, Chen K. CD46 splice variant enhances translation of specific mRNAs linked to an aggressive tumor cell phenotype in bladder cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:140-153. [PMID: 33767911 PMCID: PMC7972933 DOI: 10.1016/j.omtn.2021.02.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/19/2021] [Indexed: 01/02/2023]
Abstract
CD46 is well known to be involved in diverse biological processes. Although several splice variants of CD46 have been identified, little is known about the contribution of alternative splicing to its tumorigenic functions. In this study, we found that exclusion of CD46 exon 13 is significantly increased in bladder cancer (BCa) samples. In BCa cell lines, enforced expression of CD46-CYT2 (exon 13-skipping isoform) promoted, and CD46-CYT1 (exon 13-containing isoform) attenuated, cell growth, migration, and tumorigenicity in a xenograft model. We also applied interaction proteomics to identify exhaustively the complexes containing the CYT1 or CYT2 domain in EJ-1 cells. 320 proteins were identified that interact with the CYT1 and/or CYT2 domain, and most of them are new interactors. Using an internal ribosome entry site (IRES)-dependent reporter system, we established that CD46 could regulate mRNA translation through an interaction with the translation machinery. We also identified heterogeneous nuclear ribonucleoprotein (hnRNP)A1 as a novel CYT2 binding partner, and this interaction facilitates the interaction of hnRNPA1 with IRES RNA to promote IRES-dependent translation of HIF1a and c-Myc. Strikingly, the splicing factor SRSF1 is highly correlated with CD46 exon 13 exclusion in clinical BCa samples. Taken together, our findings contribute to understanding the role of CD46 in BCa development.
Collapse
Affiliation(s)
- Jin Zeng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Hubei Institute of Urology, Wuhan 430030, P.R. China
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang 330000, P.R. China
| | - Hua Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Hubei Institute of Urology, Wuhan 430030, P.R. China
| | - Chunhua Huang
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, P.R. China
| | - Yi Sun
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Hubei Institute of Urology, Wuhan 430030, P.R. China
| | - Haibing Xiao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Hubei Institute of Urology, Wuhan 430030, P.R. China
| | - Gan Yu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Hubei Institute of Urology, Wuhan 430030, P.R. China
| | - Hui Zhou
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Hubei Institute of Urology, Wuhan 430030, P.R. China
| | - Yangjun Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Hubei Institute of Urology, Wuhan 430030, P.R. China
| | - Weimin Yao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Hubei Institute of Urology, Wuhan 430030, P.R. China
| | - Wei Xiao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Hubei Institute of Urology, Wuhan 430030, P.R. China
| | - Junhui Hu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Lily Wu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Jinchun Xing
- Department of Urology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, P.R. China
| | - Tao Wang
- Department of Urology, The First Affiliated Hospital of Xiamen University, Xiamen 361003, P.R. China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Hubei Institute of Urology, Wuhan 430030, P.R. China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Hubei Institute of Urology, Wuhan 430030, P.R. China
| | - Ke Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- Hubei Institute of Urology, Wuhan 430030, P.R. China
| |
Collapse
|
14
|
Killick J, Hay J, Morandi E, Vermeren S, Kari S, Angles T, Williams A, Damoiseaux J, Astier AL. Vitamin D/CD46 Crosstalk in Human T Cells in Multiple Sclerosis. Front Immunol 2020; 11:598727. [PMID: 33329593 PMCID: PMC7732696 DOI: 10.3389/fimmu.2020.598727] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/27/2020] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS), in which T-cell migration into the CNS is key for pathogenesis. Patients with MS exhibit impaired regulatory T cell populations, and both Foxp3+ Tregs and type I regulatory T cells (Tr1) are dysfunctional. MS is a multifactorial disease and vitamin D deficiency is associated with disease. Herein, we examined the impact of 1,25(OH)2D3 on CD4+ T cells coactivated by either CD28 to induce polyclonal activation or by the complement regulator CD46 to promote Tr1 differentiation. Addition of 1,25(OH)2D3 led to a differential expression of adhesion molecules on CD28- and CD46-costimulated T cells isolated from both healthy donors or from patients with MS. 1,25(OH)2D3 favored Tr1 motility though a Vitamin D-CD46 crosstalk highlighted by increased VDR expression as well as increased CYP24A1 and miR-9 in CD46-costimulated T cells. Furthermore, analysis of CD46 expression on T cells from a cohort of patients with MS supplemented by vitamin D showed a negative correlation with the levels of circulating vitamin D. Moreover, t-Distributed Stochastic Neighbor Embedding (t-SNE) analysis allowed the visualization and identification of clusters increased by vitamin D supplementation, but not by placebo, that exhibited similar adhesion phenotype to what was observed in vitro. Overall, our data show a crosstalk between vitamin D and CD46 that allows a preferential effect of Vitamin D on Tr1 cells, providing novel key insights into the role of an important modifiable environmental factor in MS.
Collapse
Affiliation(s)
- Justin Killick
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Joanne Hay
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Elena Morandi
- Centre de Physiopathologie Toulouse-Purpan (CPTP), INSERM U1043, CNRS U5282, Université de Toulouse, Toulouse, France
| | - Sonja Vermeren
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Saniya Kari
- Centre de Physiopathologie Toulouse-Purpan (CPTP), INSERM U1043, CNRS U5282, Université de Toulouse, Toulouse, France
| | - Thibault Angles
- Centre de Physiopathologie Toulouse-Purpan (CPTP), INSERM U1043, CNRS U5282, Université de Toulouse, Toulouse, France
| | - Anna Williams
- Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Jan Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, Netherlands
| | - Anne L Astier
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, United Kingdom.,Centre de Physiopathologie Toulouse-Purpan (CPTP), INSERM U1043, CNRS U5282, Université de Toulouse, Toulouse, France
| |
Collapse
|
15
|
Paz S, Ritchie A, Mauer C, Caputi M. The RNA binding protein SRSF1 is a master switch of gene expression and regulation in the immune system. Cytokine Growth Factor Rev 2020; 57:19-26. [PMID: 33160830 DOI: 10.1016/j.cytogfr.2020.10.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/22/2022]
Abstract
Serine/Arginine splicing factor 1 (SRSF1) is an RNA binding protein abundantly expressed in most tissues. The pleiotropic functions of SRSF1 exert multiple roles in gene expression by regulating major steps in transcription, processing, export through the nuclear pores and translation of nascent RNA transcripts. The aim of this review is to highlight recent findings in the functions of this protein and to describe its role in immune system development, functions and regulation.
Collapse
Affiliation(s)
- Sean Paz
- Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431, United States
| | - Anastasia Ritchie
- Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431, United States
| | - Christopher Mauer
- Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431, United States
| | - Massimo Caputi
- Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431, United States.
| |
Collapse
|
16
|
Abstract
Cytometry technologies are able to profile immune cells at single-cell resolution. They are widely used for both clinical diagnosis and biological research. We developed a deep learning model for analyzing cytometry data. We demonstrated that the deep learning model accurately diagnoses the latent cytomegalovirus (CMV) in healthy individuals. In addition, we developed a method for interpreting the deep learning model, allowing us to identify biomarkers associated with latent CMV infection. The deep learning model is widely applicable to other cytometry data related to human diseases. Cytometry technologies are essential tools for immunology research, providing high-throughput measurements of the immune cells at the single-cell level. Existing approaches in interpreting and using cytometry measurements include manual or automated gating to identify cell subsets from the cytometry data, providing highly intuitive results but may lead to significant information loss, in that additional details in measured or correlated cell signals might be missed. In this study, we propose and test a deep convolutional neural network for analyzing cytometry data in an end-to-end fashion, allowing a direct association between raw cytometry data and the clinical outcome of interest. Using nine large cytometry by time-of-flight mass spectrometry or mass cytometry (CyTOF) studies from the open-access ImmPort database, we demonstrated that the deep convolutional neural network model can accurately diagnose the latent cytomegalovirus (CMV) in healthy individuals, even when using highly heterogeneous data from different studies. In addition, we developed a permutation-based method for interpreting the deep convolutional neural network model. We were able to identify a CD27- CD94+ CD8+ T cell population significantly associated with latent CMV infection, confirming the findings in previous studies. Finally, we provide a tutorial for creating, training, and interpreting the tailored deep learning model for cytometry data using Keras and TensorFlow (https://github.com/hzc363/DeepLearningCyTOF).
Collapse
|
17
|
Ma WJ, Shi YH, Chen J. Ayu (Plecoglossus altivelis) CD46 isoforms protect the cells from autologous complement attack. FISH & SHELLFISH IMMUNOLOGY 2020; 102:267-275. [PMID: 32360277 DOI: 10.1016/j.fsi.2020.04.057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/19/2020] [Accepted: 04/23/2020] [Indexed: 06/11/2023]
Abstract
CD46 is an important immune regulatory receptor with multiple functions. However, studies on the function of teleost CD46, especially the different CD46 isoforms are limited. In this study, we identified three membrane cofactor protein (MCP, CD46) gene isoforms from ayu (Plecoglossus altivelis) and tentatively named as PaCD46 isoforms. PaCD46 isoforms were generated by alternative splicing and all consisted of four conserved short consensus repeats (SCRs), and the variable serine-threonine-proline-rich domain, transmembrane hydrophobic domain, and cytoplasmic tail. Phylogenetic analysis showed that the isoforms clustered together with other fish CD46 and then with higher animal CD46. Western blotting analysis of peripheral blood mononuclear cells (PBMC) revealed three bands, all of which had much larger molecular weights than the theoretical values of the three PaCD46 isoforms. Moreover, three PaCD46 isoforms were individually expressed on HEK293 cells, and Western blotting showed the similar band profile to that of PBMC. The recombinant extracellular domain of the PaCD46 isoforms, obtained by expression in Pichia pastoris, significantly reduced hemolysis activity of ayu sera. Furthermore, each of the three PaCD46 isoforms respectively protected the HEK293 cells expressing the isoform. The isoforms were also identified for their protection of autologous PBMC from complement activation. These results provided the first evidence that PaCD46 isoforms may be complement regulatory proteins to prevent complement-induced damage to self-tissue.
Collapse
Affiliation(s)
- Wen-Jing Ma
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Yu-Hong Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
18
|
West EE, Kunz N, Kemper C. Complement and human T cell metabolism: Location, location, location. Immunol Rev 2020; 295:68-81. [PMID: 32166778 PMCID: PMC7261501 DOI: 10.1111/imr.12852] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/19/2020] [Accepted: 02/25/2020] [Indexed: 12/26/2022]
Abstract
The complement system represents one of the evolutionary oldest arms of our immune system and is commonly recognized as a liver-derived and serum-active system critical for providing protection against invading pathogens. Recent unexpected findings, however, have defined novel and rather "uncommon" locations and activities of complement. Specifically, the discovery of an intracellularly active complement system-the complosome-and its key role in the regulation of cell metabolic pathways that underly normal human T cell responses have taught us that there is still much to be discovered about this system. Here, we summarize the current knowledge about the emerging functions of the complosome in T cell metabolism. We further place complosome activities among the non-canonical roles of other intracellular innate danger sensing systems and argue that a "location-centric" view of complement evolution could logically justify its close connection with the regulation of basic cell physiology.
Collapse
Affiliation(s)
- Erin E. West
- Complement and Inflammation Research Section, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Natalia Kunz
- Complement and Inflammation Research Section, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Claudia Kemper
- Complement and Inflammation Research Section, National Heart, Lung and Blood Institute, Bethesda, MD, USA
- Faculty of Life Sciences and Medicine, King’s College London, London, UK
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| |
Collapse
|
19
|
Chiarini M, Capra R, Serana F, Bertoli D, Sottini A, Giustini V, Scarpazza C, Rovaris M, Torri Clerici V, Ferraro D, Galgani S, Solaro C, Conti MZ, Visconti A, Imberti L. Simultaneous quantification of natural and inducible regulatory T-cell subsets during interferon-β therapy of multiple sclerosis patients. J Transl Med 2020; 18:169. [PMID: 32299447 PMCID: PMC7161224 DOI: 10.1186/s12967-020-02329-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/03/2020] [Indexed: 02/06/2023] Open
Abstract
Background The mechanisms underlying the therapeutic activity of interferon-β in multiple sclerosis are still not completely understood. In the present study, we evaluated the short and long-term effects of interferon-β treatment on different subsets of regulatory T cells in relapsing–remitting multiple sclerosis patients biologically responsive to treatment because of mixovirus resistance protein A inducibility. Methods In this prospective longitudinal study, subsets of natural regulatory T cells (naïve, central memory and effector memory) and inducible regulatory T cells (Tr1), as well as in vitro-induced regulatory T cells (Tr1-like cells), were simultaneously quantified by flow cytometry in samples prepared from 148 therapy-naïve multiple sclerosis patients obtained before and after 6, 12, 18, and 24 months of interferon-β-1a treatment. mRNA for interleukin-10 and Tr1-related genes (CD18, CD49b, and CD46, together with Cyt-1 and Cyt-2 CD46-associated isoforms) were quantified in Tr1-like cells. Results Despite profound inter-individual variations in the modulation of all regulatory T-cell subsets, the percentage of natural regulatory T cells increased after 6, 12, and 24 months of interferon-β treatment. This increase was characterized by the expansion of central and effector memory regulatory T-cell subsets. The percentage of Tr1 significantly enhanced at 12 months of therapy and continued to be high at the subsequent evaluation points. Patients experiencing relapses displayed a higher percentage of naïve regulatory T cells and a lower percentage of central memory regulatory T cells and of Tr1 before starting interferon-β therapy. In addition, an increase over time of central memory and of Tr1 was observed only in patients with stable disease. However, in vitro-induced Tr1-like cells, prepared from patients treated for 24 months, produced less amount of interleukin-10 mRNA compared with pre-treatment Tr1-like cells. Conclusion Interferon-β induces the expansion of T regulatory subsets endowed with a high suppressive activity, especially in clinically stable patients. The overall concurrent modulation of natural and inducible regulatory T-cell subsets might explain the therapeutic effects of interferon-β in multiple sclerosis patients.
Collapse
Affiliation(s)
- Marco Chiarini
- Clinical Chemistry Laboratory, Diagnostic Department, ASST Spedali Civili, Brescia, Italy.,Centro di Ricerca Emato-oncologica AIL (CREA), ASST Spedali Civili, P.le Spedali Civili 1, 25123, Brescia, Italy
| | - Ruggero Capra
- Multiple Sclerosis Center, ASST Spedali Civili, Brescia, Italy
| | - Federico Serana
- Clinical Chemistry Laboratory, Diagnostic Department, ASST Spedali Civili, Brescia, Italy.,Centro di Ricerca Emato-oncologica AIL (CREA), ASST Spedali Civili, P.le Spedali Civili 1, 25123, Brescia, Italy
| | - Diego Bertoli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Centro di Ricerca Emato-oncologica AIL (CREA), ASST Spedali Civili, P.le Spedali Civili 1, 25123, Brescia, Italy
| | - Alessandra Sottini
- Centro di Ricerca Emato-oncologica AIL (CREA), ASST Spedali Civili, P.le Spedali Civili 1, 25123, Brescia, Italy
| | - Viviana Giustini
- Centro di Ricerca Emato-oncologica AIL (CREA), ASST Spedali Civili, P.le Spedali Civili 1, 25123, Brescia, Italy
| | - Cristina Scarpazza
- Multiple Sclerosis Center, ASST Spedali Civili, Brescia, Italy.,Department of General Psychology, University of Padua, Padova, Italy
| | | | | | - Diana Ferraro
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Claudio Solaro
- Department of Rehabilitation, CRRF Mons Luigi Novarese Moncrivello, Vercelli, Italy
| | | | | | - Luisa Imberti
- Centro di Ricerca Emato-oncologica AIL (CREA), ASST Spedali Civili, P.le Spedali Civili 1, 25123, Brescia, Italy.
| | | |
Collapse
|
20
|
Eyoh E, McCallum P, Killick J, Amanfo S, Mutapi F, Astier AL. The anthelmintic drug praziquantel promotes human Tr1 differentiation. Immunol Cell Biol 2019; 97:512-518. [PMID: 30623486 DOI: 10.1111/imcb.12229] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/28/2018] [Accepted: 01/07/2019] [Indexed: 12/20/2022]
Abstract
Praziquantel (PZQ) is an anthelminthic human and veterinary drug used to treat trematode and cestode worms. Changes in immune responses have been demonstrated in humans following curative PZQ treatment of schistosome infections. These changes have been attributed to the removal of immunosupressive worms and immune responses to parasite antigens exposed from dying worms. To date, there has been no study investigating the potential direct effect of PZQ on the host immune cells. Herein, we analyzed the effect of PZQ on human CD4+ T cells classically costimulated by CD3/CD28 or costimulated by the complement regulator CD46 to induce Type 1 regulatory T cells (Tr1). Our results show that PZQ enhanced T-cell proliferation, increased secretion of IL-17 and IL-10 but had no effect on secretion of GM-CSF or IFNγ. Moreover, PZQ increased the coexpression of CD49b and LAG-3, a hallmark of Tr1 cells, suggesting increased Tr1 differentiation. Indeed, supernatants from PZQ-treated cells were able to decrease bystander T-cell activation, and this was partly reduced when blocking IL-10. Hence, our study demonstrates that PZQ directly modulates human T-cell activation and promotes Tr1 differentiation, suggesting that PZQ may have immunomodulatory functions in parasite-unrelated human inflammatory diseases.
Collapse
Affiliation(s)
- Enwono Eyoh
- Institute of Immunology & Infection Research and Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, King's Buildings, Edinburgh, UK
| | - Patrick McCallum
- Institute of Immunology & Infection Research and Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, King's Buildings, Edinburgh, UK
| | - Justin Killick
- The MRC Centre for Inflammation Research, Edinburgh Centre for MS research, University of Edinburgh, Edinburgh, UK
| | - Seth Amanfo
- Institute of Immunology & Infection Research and Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, King's Buildings, Edinburgh, UK
| | - Francisca Mutapi
- Institute of Immunology & Infection Research and Centre for Immunity, Infection and Evolution, School of Biological Sciences, Ashworth Laboratories, University of Edinburgh, King's Buildings, Edinburgh, UK.,NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), Ashworth Laboratories, University of Edinburgh, King's Buildings, Edinburgh, UK
| | - Anne L Astier
- The MRC Centre for Inflammation Research, Edinburgh Centre for MS research, University of Edinburgh, Edinburgh, UK.,Centre de Physiopathologie Toulouse-Purpan (CPTP) INSERM U1043, CNRS U5282, Université de Toulouse, Toulouse, France
| |
Collapse
|
21
|
Abstract
The classical complement system is engrained in the mind of scientists and clinicians as a blood-operative key arm of innate immunity, critically required for the protection against invading pathogens. Recent work, however, has defined a novel and unexpected role for an intracellular complement system-the complosome-in the regulation of key metabolic events that underlie peripheral human T cell survival as well as the induction and cessation of their effector functions. This review summarizes the current knowledge about the emerging vital role of the complosome in T cell metabolism and discusses how viewing the evolution of the complement system from an "unconventional" vantage point could logically account for the development of its metabolic activities.
Collapse
|
22
|
Kitz A, Singer E, Hafler D. Regulatory T Cells: From Discovery to Autoimmunity. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a029041. [PMID: 29311129 DOI: 10.1101/cshperspect.a029041] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) is a genetically mediated autoimmune disease of the central nervous system. Allelic variants lead to lower thresholds of T-cell activation resulting in activation of autoreactive T cells. Environmental factors, including, among others, diet, vitamin D, and smoking, in combination with genetic predispositions, play a substantial role in disease development and activation of autoreactive T cells. FoxP3+ regulatory T cells (Tregs) have emerged as central in the control of autoreactive T cells. A consistent finding in patients with MS is defects in Treg cell function with reduced suppression of effector T cells and production of proinflammatory cytokines. Emerging data suggests that functional Tregs become effector-like T cells with loss of function associated with T-bet expression and interferon γ (IFN-γ) secretion.
Collapse
Affiliation(s)
- Alexandra Kitz
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut 06520
| | - Emily Singer
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut 06520
| | - David Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
23
|
Freeley S, Cardone J, Günther SC, West EE, Reinheckel T, Watts C, Kemper C, Kolev MV. Asparaginyl Endopeptidase (Legumain) Supports Human Th1 Induction via Cathepsin L-Mediated Intracellular C3 Activation. Front Immunol 2018; 9:2449. [PMID: 30405635 PMCID: PMC6207624 DOI: 10.3389/fimmu.2018.02449] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 10/04/2018] [Indexed: 12/31/2022] Open
Abstract
Autocrine activation of the complement receptors C3aR and CD46 by complement activation components C3a and C3b produced through C3 cleavage by the protease cathepsin L (CTSL) during T cell stimulation is a requirement for IFN-γ production and Th1 induction in human CD4+ T cells. Thus, lack of autocrine CD46 activation, such as in CD46-deficient patients, is associated with defective Th1 responses and recurrent infections. We have identified LGMN [the gene coding for legumain, also known as asparaginyl endopeptidase (AEP)] as one of the key genes induced by CD46 co-stimulation during human CD4+ T cell activation. AEP processes and activates a range of proteins, among those α1-thymosin and CTSL, which both drive intrinsically Th1 activity-but has so far not been described to be functionally active in human T cells. Here we found that pharmacological inhibition of AEP during activation of human CD4+ T cells reduced CTSL activation and the CTSL-mediated generation of intracellular C3a. This translated into a specific reduction of IFN-γ production without affecting cell proliferation or survival. In line with these findings, CD4+ T cells isolated from Lgmn -/- mice also displayed a specific defect in IFN-γ secretion and Th1 induction. Furthermore, we did not observe a role for AEP-driven autocrine α1-thymosin activation in T cell-derived IFN-γ production. These data suggest that AEP is an "upstream" activator of the CTSL-C3-IFN-γ axis in human CD4+ T cells and hence an important supporter of human Th1 induction.
Collapse
Affiliation(s)
- Simon Freeley
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - John Cardone
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Sira C Günther
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom.,Institut für Medizinische Virologie, University of Zurich, Zurich, Switzerland
| | - Erin E West
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Thomas Reinheckel
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs University Freiburg, and German Cancer Consortium (DKTK), Freiburg, Germany
| | - Colin Watts
- Division of Cell Signaling & Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Claudia Kemper
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom.,Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, United States.,Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Martin V Kolev
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| |
Collapse
|
24
|
Hansen AS, Biltoft M, Bundgaard B, Bohn AB, Møller BK, Höllsberg P. CD46 activation induces distinct CXCL-10 response in monocytes and monocyte-derived dendritic cells. Cytokine 2018; 113:466-469. [PMID: 29980471 DOI: 10.1016/j.cyto.2018.06.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 06/25/2018] [Accepted: 06/26/2018] [Indexed: 12/15/2022]
Abstract
CD46 is an important immune regulatory receptor with dual functions, however, the CD46 isoform distribution and the effect of CD46 activation on the cytokine production in monocytes and monocyte-derived dendritic cells (moDCs) is unclear. Here, we show that CD46 activation of moDCs downregulates LPS-induced CXCL-10 expression, while the expression of CXCL-10 in monocytes is unaffected. Furthermore, the differentiation of moDCs induces a switch towards dominance of CYT-2 isoforms of CD46. These data indicate that CD46 activation exerts different functions in monocytes and moDCs and this correlates with a switch in CD46 isoform expression upon differentiation of moDCs.
Collapse
Affiliation(s)
- Aida S Hansen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| | - Mette Biltoft
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Anja B Bohn
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Bjarne K Møller
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Per Höllsberg
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
25
|
Hansen AS, Slater J, Biltoft M, Bundgaard BB, Møller BK, Höllsberg P. CD46 is a potent co-stimulatory receptor for expansion of human IFN-γ-producing CD8 + T cells. Immunol Lett 2018; 200:26-32. [PMID: 29902483 PMCID: PMC7112827 DOI: 10.1016/j.imlet.2018.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/18/2018] [Accepted: 06/08/2018] [Indexed: 11/18/2022]
Abstract
Similar to CD4+ T cells, precursor CD8+ T cells are thought to depend on a co-stimulatory signal through CD28 for proliferation and differentiation into effector cells. CD46 is another co-stimulatory receptor that promotes differentiation of CD4+ T-helper cells type 1 (Th1 cells) into a regulatory phenotype with a switch from IFN-γ towards IL-10-secretion over time. Whether CD46 exerts a similar function on CD8+ T cells remains to be fully elucidated. Here, we demonstrate that CD46 co-stimulation induced secretion of IFN-γ as well as expansion of IFN-γ-secreting CD8+ T cells. In contrast to CD46 co-stimulation of CD4+ T cells, CD8+ T cells did not differentiate into a regulatory IL-10-secreting phenotype. This demonstrates that CD46 is a co-stimulatory receptor on CD8+ T cells, and that it exerts separate functions during CD4+ and CD8+ T-cell differentiation.
Collapse
Affiliation(s)
- Aida S Hansen
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Josefine Slater
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Mette Biltoft
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Bjarne K Møller
- Department of Clinical Immunology, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Per Höllsberg
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| |
Collapse
|
26
|
Abstract
The complement system is an evolutionarily ancient key component of innate immunity required for the detection and removal of invading pathogens. It was discovered more than 100 years ago and was originally defined as a liver-derived, blood-circulating sentinel system that classically mediates the opsonization and lytic killing of dangerous microbes and the initiation of the general inflammatory reaction. More recently, complement has also emerged as a critical player in adaptive immunity via its ability to instruct both B and T cell responses. In particular, work on the impact of complement on T cell responses led to the surprising discoveries that the complement system also functions within cells and is involved in regulating basic cellular processes, predominantly those of metabolic nature. Here, we review current knowledge about complement's role in T cell biology, with a focus on the novel intracellular and noncanonical activities of this ancient system.
Collapse
Affiliation(s)
- Erin E West
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, Maryland 20892, United States; ,
| | - Martin Kolev
- Division of Transplant Immunology and Mucosal Biology, King's College London, London SE1 9RT, United Kingdom;
| | - Claudia Kemper
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, Maryland 20892, United States; ,
- Division of Transplant Immunology and Mucosal Biology, King's College London, London SE1 9RT, United Kingdom;
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
27
|
West EE, Afzali B, Kemper C. Unexpected Roles for Intracellular Complement in the Regulation of Th1 Responses. Adv Immunol 2018; 138:35-70. [PMID: 29731006 DOI: 10.1016/bs.ai.2018.02.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The complement system is generally recognized as an evolutionarily ancient and critical part of innate immunity required for the removal of pathogens that have breached the protective host barriers. It was originally defined as a liver-derived serum surveillance system that induces the opsonization and killing of invading microbes and amplifies the general inflammatory reactions. However, studies spanning the last four decades have established complement also as a vital bridge between innate and adaptive immunity. Furthermore, recent work on complement, and in particular its impact on human T helper 1 (Th1) responses, has led to the unexpected findings that the complement system also functions within cells and that it participates in the regulation of basic processes of the cell, including metabolism. These recent new insights into the unanticipated noncanonical activities of this ancient system suggest that the functions of complement extend well beyond mere host protection and into cellular physiology.
Collapse
Affiliation(s)
- Erin E West
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, United States
| | - Behdad Afzali
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, United States; Lymphocyte Cell Biology Section (Molecular Immunology and Inflammation Branch), National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Claudia Kemper
- Laboratory of Molecular Immunology and Immunology Center, National Heart, Lung and Blood Institute, Bethesda, MD, United States; Division of Transplant Immunology and Mucosal Biology, King's College London, London, United Kingdom; Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
28
|
Killick J, Morisse G, Sieger D, Astier AL. Complement as a regulator of adaptive immunity. Semin Immunopathol 2018; 40:37-48. [PMID: 28842749 PMCID: PMC5794818 DOI: 10.1007/s00281-017-0644-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/03/2017] [Indexed: 11/30/2022]
Abstract
The complement system is an ancient and evolutionarily conserved effector system comprising in mammals over 50 circulating and membrane bound proteins. Complement has long been described as belonging to the innate immune system; however, a number of recent studies have demonstrated its key role in the modulation of the adaptive immune response. This review does not set out to be an exhaustive list of the numerous interactions of the many complement components with adaptive immunity; rather, we will focus more precisely on the role of some complement molecules in the regulation of antigen presenting cells, as well as on their direct effect on the activation of the core adaptive immune cells, B and T lymphocytes. Recent reports on the local production and activation of complement proteins also suggest a major role in the control of effector responses. The crucial role of complement in adaptive immunity is further highlighted by several examples of dysregulation of these pathways in human diseases.
Collapse
Affiliation(s)
- Justin Killick
- MRC Centre for Inflammation Research, Edinburgh Centre for MS Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Gregoire Morisse
- MRC Centre for Inflammation Research, Edinburgh Centre for MS Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
- Centre for NeuroRegeneration, Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Dirk Sieger
- Centre for NeuroRegeneration, Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Anne L Astier
- MRC Centre for Inflammation Research, Edinburgh Centre for MS Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK.
- Inserm U1043, CNRS U5282, Université de Toulouse, Centre de Physiopathologie Toulouse-Purpan (CPTP), F-31300, Toulouse, France.
| |
Collapse
|
29
|
Ni Choileain S, Hay J, Thomas J, Williams A, Vermeren MM, Benezech C, Gomez-Salazar M, Hugues OR, Vermeren S, Howie SEM, Dransfield I, Astier AL. TCR-stimulated changes in cell surface CD46 expression generate type 1 regulatory T cells. Sci Signal 2017; 10:10/502/eaah6163. [PMID: 29066539 DOI: 10.1126/scisignal.aah6163] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A lack of regulatory T cell function is a critical factor in the pathogenesis of autoimmune diseases, such as multiple sclerosis (MS). Ligation of the complement regulatory protein CD46 facilitates the differentiation of T helper 1 (TH1) effector cells into interleukin-10 (IL-10)-secreting type 1 regulatory T cells (Tr1 cells), and this pathway is defective in MS patients. Cleavage of the ectodomain of CD46, which contains three N-glycosylation sites and multiple O-glycosylation sites, enables CD46 to activate T cells. We found that stimulation of the T cell receptor (TCR)-CD3 complex was associated with a reduction in the apparent molecular mass of CD46 in a manner that depended on O-glycosylation. CD3-stimulated changes in CD46 O-glycosylation status reduced CD46 processing and subsequent T cell signaling. During T cell activation, CD46 was recruited to the immune synapse in a manner that required its serine-, threonine-, and proline-rich (STP) region, which is rich in O-glycosylation sites. Recruitment of CD46 to the immune synapse switched T cells from producing the inflammatory cytokine interferon-γ (IFN-γ) to producing IL-10. Furthermore, CD4+ T cells isolated from MS patients did not exhibit a CD3-stimulated reduction in the mass of CD46 and thus showed increased amounts of cell surface CD46. Together, these data suggest a possible mechanism underlying the regulatory function of CD46 on T cells. Our findings may explain why this pathway is defective in patients with MS and provide insights into MS pathogenesis that could help to design future immunotherapies.
Collapse
Affiliation(s)
- Siobhan Ni Choileain
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Joanne Hay
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Joelle Thomas
- Université Claude Bernard Lyon I, CNRS UMR 5310-INSERM U1217, F-69100 Lyon, France
| | - Anna Williams
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Matthieu M Vermeren
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Cecile Benezech
- UK Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Mario Gomez-Salazar
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Owen R Hugues
- Millipore (U.K.) Limited, Croxley Green Business Park, Watford, Hertfordshire WD18 8ZB, UK
| | - Sonja Vermeren
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Sarah E M Howie
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Ian Dransfield
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Anne L Astier
- Medical Research Council (MRC) Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK. .,Centre de Physiopathologie Toulouse-Purpan, INSERM U1043, CNRS U5282, Université de Toulouse, Toulouse F-31300, France
| |
Collapse
|
30
|
Freeley S, Kemper C, Le Friec G. The "ins and outs" of complement-driven immune responses. Immunol Rev 2017; 274:16-32. [PMID: 27782335 DOI: 10.1111/imr.12472] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The complement system represents an evolutionary old and critical component of innate immunity where it forms the first line of defense against invading pathogens. Originally described as a heat-labile fraction of the serum responsible for the opsonization and subsequent lytic killing of bacteria, work over the last century firmly established complement as a key mediator of the general inflammatory response but also as an acknowledged vital bridge between innate and adaptive immunity. However, recent studies particularly spanning the last decade have provided new insights into the novel modes and locations of complement activation and highlighted unexpected additional biological functions for this ancient system, for example, in regulating basic processes of the cell. In this review, we will cover the current knowledge about complement's established and novel roles in innate and adaptive immunity with a focus on the functional differences between serum circulating and intracellularly active complement and will describe and discuss the newly discovered cross-talks of complement with other cell effector systems particularly during T-cell induction and contraction.
Collapse
Affiliation(s)
- Simon Freeley
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Claudia Kemper
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK. .,Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Gaëlle Le Friec
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| |
Collapse
|
31
|
Intracellular complement - the complosome - in immune cell regulation. Mol Immunol 2017; 89:2-9. [PMID: 28601357 PMCID: PMC7112704 DOI: 10.1016/j.molimm.2017.05.012] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 05/14/2017] [Accepted: 05/19/2017] [Indexed: 12/18/2022]
Abstract
The complement system was defined over a century ago based on its ability to "complement" the antibody-mediated and cell-mediated immune responses against pathogens. Today our understanding of this ancient part of innate immunity has changed substantially and we know now that complement plays an undisputed pivotal role in the regulation of both innate and adaptive immunity. The complement system consists of over 50 blood-circulating, cell-surface expressed and intracellular proteins. It is key in the recognition and elimination of invading pathogens, also in the removal of self-derived danger such as apoptotic cells, and it supports innate immune responses and the initiation of the general inflammatory reactions. The long prevailing classic view of complement was that of a serum-operative danger sensor and first line of defence system, however, recent experimental and clinical evidences have demonstrated that "local" tissue and surprisingly intracellular complement (the complosome) activation impacts on normal cell physiology. This review will focus on novel aspects of intracellular complement activation and its unexpected roles in basic cell processes such as metabolism. We also discuss what the existence of the complosome potentially means for how the host handles intracellular pathogens such as viruses.
Collapse
|
32
|
Ly PT, Tang SJ, Roca X. Alternative polyadenylation expands the mRNA isoform repertoire of human CD46. Gene 2017; 625:21-30. [PMID: 28476687 DOI: 10.1016/j.gene.2017.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/28/2017] [Accepted: 05/02/2017] [Indexed: 11/28/2022]
Abstract
Alternative polyadenylation is a prevalent mechanism regulating mammalian gene expression. While tandem 3'-Untranslated-Region (3'UTR) polyadenylation changes expression levels, Intronic PolyAdenylation generates shorter transcripts encoding truncated proteins. Intronic PolyAdenylation regulates 20% of genes and is especially common in receptor tyrosine-kinase transcripts, generating soluble repressors. Here we report that human CD46, encoding a TransMembrane repressor of complement and T-cell co-stimulator, expresses multiple isoforms by alternative polyadenylation. We provide evidence for polyadenylation at several introns by RT-PCR of 5' intronic fragments, and by increase in such isoforms via functional U1 knockdown. We mapped various Intronic PolyAdenylation Sites by 3' Rapid Amplification of cDNA Ends (3'RACE), which could generate soluble or membrane-bound but tail-less CD46. Intronic PolyAdenylation could add to the source of soluble CD46 isoforms in fluids and tissues, which increase in cancers and autoimmune syndromes. Furthermore, 3'RACE identified three PolyAdenylation Sites within the last intron and exon, whose transcripts with shortened 3'UTRs could support higher CD46 expression. Finally, 3'RACE revealed that the CD46 Pseudogene only expresses short transcripts by early polyadenylation in intron 2. Overall, we report a wide variety of CD46 mRNA isoforms which could generate new protein isoforms, adding to the diverse physiological and pathological roles of CD46.
Collapse
Affiliation(s)
- Phuong Thao Ly
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore; The Neuroscience and Behavioral Disorders Programme, Duke-NUS Graduate Medical School, Singapore
| | - Sze Jing Tang
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Xavier Roca
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore.
| |
Collapse
|
33
|
Kolev M, Kemper C. Keeping It All Going-Complement Meets Metabolism. Front Immunol 2017; 8:1. [PMID: 28149297 PMCID: PMC5241319 DOI: 10.3389/fimmu.2017.00001] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/03/2017] [Indexed: 01/22/2023] Open
Abstract
The complement system is an evolutionary old and crucial component of innate immunity, which is key to the detection and removal of invading pathogens. It was initially discovered as a liver-derived sentinel system circulating in serum, the lymph, and interstitial fluids that mediate the opsonization and lytic killing of bacteria, fungi, and viruses and the initiation of the general inflammatory responses. Although work performed specifically in the last five decades identified complement also as a critical instructor of adaptive immunity—indicating that complement’s function is likely broader than initially anticipated—the dominant opinion among researchers and clinicians was that the key complement functions were in principle defined. However, there is now a growing realization that complement activity goes well beyond “classic” immune functions and that this system is also required for normal (neuronal) development and activity and general cell and tissue integrity and homeostasis. Furthermore, the recent discovery that complement activation is not confined to the extracellular space but occurs within cells led to the surprising understanding that complement is involved in the regulation of basic processes of the cell, particularly those of metabolic nature—mostly via novel crosstalks between complement and intracellular sensor, and effector, pathways that had been overlooked because of their spatial separation. These paradigm shifts in the field led to a renaissance in complement research and provide new platforms to now better understand the molecular pathways underlying the wide-reaching effects of complement functions in immunity and beyond. In this review, we will cover the current knowledge about complement’s emerging relationship with the cellular metabolism machinery with a focus on the functional differences between serum-circulating versus intracellularly active complement during normal cell survival and induction of effector functions. We will also discuss how taking a closer look into the evolution of key complement components not only made the functional connection between complement and metabolism rather “predictable” but how it may also give clues for the discovery of additional roles for complement in basic cellular processes.
Collapse
Affiliation(s)
- Martin Kolev
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital , London , UK
| | - Claudia Kemper
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK; Laboratory of Molecular Immunology, The Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
34
|
Hansen AS, Bundgaard BB, Møller BK, Höllsberg P. Non-random pairing of CD46 isoforms with skewing towards BC2 and C2 in activated and memory/effector T cells. Sci Rep 2016; 6:35406. [PMID: 27739531 PMCID: PMC5064401 DOI: 10.1038/srep35406] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 09/27/2016] [Indexed: 11/24/2022] Open
Abstract
CD46 is a glycoprotein with important functions in innate and adaptive immune responses. Functionally different isoforms are generated by alternative splicing at exons 7–9 (BC and C isoforms) and exon 13 (CYT-1 and CYT-2 isoforms) giving rise to BC1, BC2, C1 and C2. We developed a novel real-time PCR assay that allows quantitative comparisons between these isoforms. Their relative frequency in CD4+ T cells from 100 donors revealed a distribution with high interpersonally variability. Importantly, the distribution between the isoforms was not random and although splicing favoured inclusion of exon 8 (BC isoforms), exclusion of exon 8 (C isoforms) was significantly linked to exclusion of exon 13 (CYT-2 isoforms). Despite inter-individual differences, CD4+ and CD8+ T cells, B cells, NK cells and monocytes expressed similar isoform profiles intra-individually. However, memory/effector CD4+ T cells had a significantly higher frequency of CYT-2 when compared with naïve CD4+ T cells. Likewise, in vitro activation of naïve and total CD4+ T cells increased the expression of CYT-2. This indicates that although splicing factors determine a certain expression profile in an individual, the profile can be modulated by external stimuli. This suggests a mechanism by which alterations in CD46 isoforms may temporarily regulate the immune response.
Collapse
Affiliation(s)
- Aida S Hansen
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | | | - Bjarne K Møller
- Department of Clinical Immunology, Aarhus University Hospital, DK-8200 Aarhus N, Denmark
| | - Per Höllsberg
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
35
|
Tang SJ, Luo S, Ho JXJ, Ly PT, Goh E, Roca X. Characterization of the Regulation of CD46 RNA Alternative Splicing. J Biol Chem 2016; 291:14311-14323. [PMID: 27226545 DOI: 10.1074/jbc.m115.710350] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Indexed: 11/06/2022] Open
Abstract
Here we present a detailed analysis of the alternative splicing regulation of human CD46, which generates different isoforms with distinct functions. CD46 is a ubiquitous membrane protein that protects host cells from complement and plays other roles in immunity, autophagy, and cell adhesion. CD46 deficiency causes an autoimmune disorder, and this protein is also involved in pathogen infection and cancer. Before this study, the mechanisms of CD46 alternative splicing remained unexplored even though dysregulation of this process has been associated with autoimmune diseases. We proved that the 5' splice sites of CD46 cassette exons 7 and 8 encoding extracellular domains are defined by noncanonical mechanisms of base pairing to U1 small nuclear RNA. Next we characterized the regulation of CD46 cassette exon 13, whose inclusion or skipping generates different cytoplasmic tails with distinct functions. Using splicing minigenes, we identified multiple exonic and intronic splicing enhancers and silencers that regulate exon 13 inclusion via trans-acting splicing factors like PTBP1 and TIAL1. Interestingly, a common splicing activator such as SRSF1 appears to repress CD46 exon 13 inclusion. We also report that expression of CD46 mRNA isoforms is further regulated by non-sense-mediated mRNA decay and transcription speed. Finally, we successfully manipulated CD46 exon 13 inclusion using antisense oligonucleotides, opening up opportunities for functional studies of the isoforms as well as for therapeutics for autoimmune diseases. This study provides insight into CD46 alternative splicing regulation with implications for its function in the immune system and for genetic disease.
Collapse
Affiliation(s)
- Sze Jing Tang
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Shufang Luo
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Jia Xin Jessie Ho
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Phuong Thao Ly
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Eling Goh
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Xavier Roca
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
36
|
Preclinical safety, pharmacokinetics, pharmacodynamics, and biodistribution studies with Ad35K++ protein: a novel rituximab cotherapeutic. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 5:16013. [PMID: 27069950 PMCID: PMC4813608 DOI: 10.1038/mtm.2016.13] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 12/10/2015] [Accepted: 12/11/2015] [Indexed: 12/31/2022]
Abstract
Rituximab is a mouse/human chimeric monoclonal antibody targeted toward CD20. It is efficient as first-line therapy of CD20-positive B-cell malignancies. However, a large fraction of treated patients relapse with rituximab-resistant disease. So far, only modest progress has been made in treatment options for rituximab refractory patients. One of the mechanisms for rituximab resistance involves the upregulation of CD46, which is a key cell surface protein that blocks the activation of complement. We have recently developed a technology that depletes CD46 from the cell surface and thereby sensitizes tumor cells to complement-dependent cytotoxicity. This technology is based on a small recombinant protein, Ad35K++ that binds with high affinity to CD46. In preliminary studies using a 6 × histidinyl tagged protein, we had demonstrated that intravenous Ad35K++ injection in combination with rituximab was safe and increased rituximab-mediated killing of CD20-positive target cells in mice and nonhuman primates (NHPs). The presence of the tag, while allowing for easy purification by Ni-NTA chromatography, has the potential to increase the immunogenicity of the recombinant protein. For clinical application, we therefore developed an Ad35K++ protein without His-tag. In the present study, we performed preclinical studies in two animal species (mice and NHPs) with this protein demonstrating its safety and efficacy. These studies estimated the Ad35K++ dose range and treatment regimen to be used in patients. Furthermore, we showed that intravenous Ad35K++ injection triggers the shedding of the CD46 extracellular domain in xenograft mouse tumor models and in macaques. Shed serum CD46 can be measured in the serum and can potentially be used as a pharmacodynamic marker for monitoring Ad35K++ activity in patient undergoing treatment with this agent. These studies create the basis for an investigational new drug application for the use of Ad35K++ in combination with rituximab in the treatment of patients with B-cell malignancies.
Collapse
|
37
|
Tilib Shamoun S, Le Friec G, Spinner N, Kemper C, Baker AJ. Immune dysregulation in Alagille syndrome: A new feature of the evolving phenotype. Clin Res Hepatol Gastroenterol 2015; 39:566-9. [PMID: 26026399 DOI: 10.1016/j.clinre.2015.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 01/20/2015] [Accepted: 02/01/2015] [Indexed: 02/04/2023]
Abstract
Alagille syndrome (ALGS) is a rare autosomal dominant, multi-system disease caused by mutations in one of two NOTCH signaling pathway genes. Mutations in JAG1 are found in more than 94% of patients, with associated Jagged1 defects. We previously showed that CD46, which is a complement and immune regulator, regulates NOTCH expression during T cell activation after binding to C3b/C4b. We have identified 25% of our ALGS cohort with frequent infections and studied a subgroup of 4 in detail who were not showing current features of infections in order to show if Jagged1 abnormalities could affect immune function. We used cytometric bead arrays and FACS to measure cytokines and cell membrane expression. Resting and activated T cells were studied in both low and high IL-2 concentration to assess the TH1 ability to shift from INFγ to IL-10 production. In vitro initial PBMC cell population and subpopulation assessment were normal but further assessment of the lymphocytes revealed that while NOTCH1 expression and regulation was normal on resting TH1, Jagged1 expression was exaggerated. Resting TH1 cells from some patients exhibited high CD132 levels. Upon activating T cells, TH1 cells managed to produce TNF but failed to produce sufficient IFNγ levels (in two patients TH1 produced no IFNγ). TH2 exhibited exaggerated response with high IL-4 and IL-5 levels. TH1 were unable to down-regulate CD127, resulting in prolonged immune activation, and failed to shift from IFNγ to IL-10 production maintaining high IL-2 levels suggesting an impaired T cell response. Disturbed CD46-Jagged1 interaction may explain recurrent infections among ALGS patients, and could predispose to Th2-driven conditions such as asthma, eczema, food allergies and airway atopy and otitis media. The ALGS description could now be extended to include immune dysregulation.
Collapse
Affiliation(s)
- S Tilib Shamoun
- King's College Hospital, Denmark Hill, London SE5 9RS, United Kingdom
| | - G Le Friec
- MRC Center for Transplantation, Guys' Hospital, King's College London, 5th Floor Tower Wing, London SE1 9RT, United Kingdom
| | - N Spinner
- Abramson Research Center, The Children's Hospital of Philadelphia, 3615, Civic Center Boulevard, Philadelphia 19104-4318, PA, United States
| | - C Kemper
- MRC Center for Transplantation, Guys' Hospital, King's College London, 5th Floor Tower Wing, London SE1 9RT, United Kingdom
| | - A J Baker
- King's College Hospital, Denmark Hill, London SE5 9RS, United Kingdom.
| |
Collapse
|
38
|
Kolev M, Dimeloe S, Le Friec G, Navarini A, Arbore G, Povoleri GA, Fischer M, Belle R, Loeliger J, Develioglu L, Bantug GR, Watson J, Couzi L, Afzali B, Lavender P, Hess C, Kemper C. Complement Regulates Nutrient Influx and Metabolic Reprogramming during Th1 Cell Responses. Immunity 2015; 42:1033-47. [PMID: 26084023 PMCID: PMC4518498 DOI: 10.1016/j.immuni.2015.05.024] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 03/24/2015] [Accepted: 04/10/2015] [Indexed: 01/02/2023]
Abstract
Expansion and acquisition of Th1 cell effector function requires metabolic reprogramming; however, the signals instructing these adaptations remain poorly defined. Here we found that in activated human T cells, autocrine stimulation of the complement receptor CD46, and specifically its intracellular domain CYT-1, was required for induction of the amino acid (AA) transporter LAT1 and enhanced expression of the glucose transporter GLUT1. Furthermore, CD46 activation simultaneously drove expression of LAMTOR5, which mediated assembly of the AA-sensing Ragulator-Rag-mTORC1 complex and increased glycolysis and oxidative phosphorylation (OXPHOS), required for cytokine production. T cells from CD46-deficient patients, characterized by defective Th1 cell induction, failed to upregulate the molecular components of this metabolic program as well as glycolysis and OXPHOS, but IFN-γ production could be reinstated by retrovirus-mediated CD46-CYT-1 expression. These data establish a critical link between the complement system and immunometabolic adaptations driving human CD4+ T cell effector function. CD46 regulates GLUT1 and LAT1 and enhances glucose and AA uptake in T cells LAMTOR5 mediates Ragulator-Rag-mTORC1 assembly in activated T cells Complement drives glycolysis and oxidative phosphorylation critical to Th1 cell induction
Collapse
Affiliation(s)
- Martin Kolev
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Sarah Dimeloe
- Department of Biomedicine, Immunobiology, University of Basel, 20 Hebelstrasse, 4031 Basel, Switzerland
| | - Gaelle Le Friec
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Alexander Navarini
- Department of Dermatology, University Hospital Zurich, 31 Gloriastrasse, 8091 Zürich, Switzerland
| | - Giuseppina Arbore
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Giovanni A Povoleri
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK; Biomedical Research Centre, King's Health Partners, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Marco Fischer
- Department of Biomedicine, Immunobiology, University of Basel, 20 Hebelstrasse, 4031 Basel, Switzerland
| | - Réka Belle
- Department of Biomedicine, Immunobiology, University of Basel, 20 Hebelstrasse, 4031 Basel, Switzerland
| | - Jordan Loeliger
- Department of Biomedicine, Immunobiology, University of Basel, 20 Hebelstrasse, 4031 Basel, Switzerland
| | - Leyla Develioglu
- Department of Biomedicine, Immunobiology, University of Basel, 20 Hebelstrasse, 4031 Basel, Switzerland
| | - Glenn R Bantug
- Department of Biomedicine, Immunobiology, University of Basel, 20 Hebelstrasse, 4031 Basel, Switzerland
| | - Julie Watson
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Lionel Couzi
- Nephrology Transplantation, CHU Bordeaux, Hospital Pellegrin, CNRS UMR 1564, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Behdad Afzali
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK; Biomedical Research Centre, King's Health Partners, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK; Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Paul Lavender
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Christoph Hess
- Department of Biomedicine, Immunobiology, University of Basel, 20 Hebelstrasse, 4031 Basel, Switzerland.
| | - Claudia Kemper
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
| |
Collapse
|
39
|
Monocyte:T-cell interaction regulates human T-cell activation through a CD28/CD46 crosstalk. Immunol Cell Biol 2015; 93:796-803. [PMID: 25787182 PMCID: PMC4519525 DOI: 10.1038/icb.2015.42] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/20/2015] [Accepted: 03/15/2015] [Indexed: 01/20/2023]
Abstract
T cell activation requires engagement of the T cell receptor and of at least one costimulatory molecule. The key role of CD28 in inducing T cell activation has been reported several decades ago and the molecular mechanisms involved well described. The complement regulator CD46 also acts as a costimulatory molecule for T cells but, in contrast to CD28, has the ability to drive T cell differentiation from producing some IFNγ to secreting some potent anti-inflammatory IL-10, acquiring a so-called Type I regulatory phenotype (Tr1). Proteolytic cleavage of CD46 occurs upon costimulation and is important for T cell activation and IL-10 production. The observation that CD46 cleavage was reduced when PBMC were costimulated compared to purified naive T cells led us to hypothesize that interactions between different cell types within the PBMC were able to modulate the CD46 pathway. We show that CD46 downregulation is also reduced when CD4+ T cells are co-cultured with autologous monocytes. Indeed, monocyte:T cell co-cultures impaired CD46–mediated T cell differentiation and coactivation, by reducing downregulation of surface CD46, lowering induction of the early activation marker CD69, as well as reducing the levels of IL-10 secretion. Blocking of CD86 could partly restore CD69 expression and cytokine secretion, demonstrating that the CD28-CD86 pathway regulates CD46 activation. Direct concomitant ligation of CD28 and CD46 on CD4+ T cells also modulated CD46 expression and regulated cytokine production. These data identify a crosstalk between two main costimulatory pathways and provide novel insights into the regulation of human T cell activation.
Collapse
|
40
|
Hayes CE, Hubler SL, Moore JR, Barta LE, Praska CE, Nashold FE. Vitamin D Actions on CD4(+) T Cells in Autoimmune Disease. Front Immunol 2015; 6:100. [PMID: 25852682 PMCID: PMC4364365 DOI: 10.3389/fimmu.2015.00100] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/23/2015] [Indexed: 12/11/2022] Open
Abstract
This review summarizes and integrates research on vitamin D and CD4+ T-lymphocyte biology to develop new mechanistic insights into the molecular etiology of autoimmune disease. A deep understanding of molecular mechanisms relevant to gene–environment interactions is needed to deliver etiology-based autoimmune disease prevention and treatment strategies. Evidence linking sunlight, vitamin D, and the risk of multiple sclerosis and type 1 diabetes is summarized to develop the thesis that vitamin D is the environmental factor that most strongly influences autoimmune disease development. Evidence for CD4+ T-cell involvement in autoimmune disease pathogenesis and for paracrine calcitriol signaling to CD4+ T lymphocytes is summarized to support the thesis that calcitriol is sunlight’s main protective signal transducer in autoimmune disease risk. Animal modeling and human mechanistic data are summarized to support the view that vitamin D probably influences thymic negative selection, effector Th1 and Th17 pathogenesis and responsiveness to extrinsic cell death signals, FoxP3+CD4+ T-regulatory cell and CD4+ T-regulatory cell type 1 (Tr1) cell functions, and a Th1–Tr1 switch. The proposed Th1–Tr1 switch appears to bridge two stable, self-reinforcing immune states, pro- and anti-inflammatory, each with a characteristic gene regulatory network. The bi-stable switch would enable T cells to integrate signals from pathogens, hormones, cell–cell interactions, and soluble mediators and respond in a biologically appropriate manner. Finally, unanswered questions and potentially informative future research directions are highlighted to speed delivery of etiology-based strategies to reduce autoimmune disease.
Collapse
Affiliation(s)
- Colleen Elizabeth Hayes
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison , Madison, WI , USA
| | - Shane L Hubler
- Department of Statistics, College of Letters and Sciences, University of Wisconsin-Madison , Madison, WI , USA
| | - Jerott R Moore
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison , Madison, WI , USA
| | - Lauren E Barta
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison , Madison, WI , USA
| | - Corinne E Praska
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison , Madison, WI , USA
| | - Faye E Nashold
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison , Madison, WI , USA
| |
Collapse
|
41
|
Abstract
In addition to its established contribution to innate immunity, recent studies have suggested novel roles for the complement system in the development of various lung diseases. Several studies have demonstrated that complement may serve as a key link between innate and adaptive immunity in a variety of pulmonary conditions. However, the specific contributions of complement to lung diseases based on innate and adaptive immunity are just beginning to emerge. Elucidating the role of complement-mediated immune regulation in these diseases will help to identify new targets for therapeutic interventions.
Collapse
|
42
|
Cheng YL, Choi Y, Sobey CG, Arumugam TV, Jo DG. Emerging roles of the γ-secretase-notch axis in inflammation. Pharmacol Ther 2014; 147:80-90. [PMID: 25448038 DOI: 10.1016/j.pharmthera.2014.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/03/2014] [Indexed: 12/14/2022]
Abstract
γ-Secretase is a distinct proteolytic complex required for the activation of many transmembrane proteins. The cleavage of substrates by γ-secretase plays diverse biological roles in producing essential products for the organism. More than 90 transmembrane proteins have been reported to be substrates of γ-secretase. Two of the most widely known and studied of these substrates are the amyloid precursor protein (APP) and the Notch receptor, which are precursors for the generation of amyloid-β (Aβ) and the Notch intracellular domain (NICD), respectively. The wide spectrum of γ-secretase substrates has made analyses of the pathology of γ-secretase-related diseases and underlying mechanisms challenging. Inflammation is an important aspect of disease pathology that requires an in-depth analysis. γ-Secretase may contribute to disease development or progression by directly increasing and regulating production of pro-inflammatory cytokines. This review summarizes recent evidence for a role of γ-secretase in inflammatory diseases, and discusses the potential use of γ-secretase inhibitors as an effective future treatment option.
Collapse
Affiliation(s)
- Yi-Lin Cheng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - Yuri Choi
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | | | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
43
|
Hay J, Carter D, Lieber A, Astier AL. Recombinant Ad35 adenoviral proteins as potent modulators of human T cell activation. Immunology 2014; 144:453-460. [PMID: 25251258 PMCID: PMC4557682 DOI: 10.1111/imm.12391] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 09/03/2014] [Accepted: 09/16/2014] [Indexed: 11/30/2022] Open
Abstract
The protein CD46 protects cells from complement attack by regulating cleavage of C3b and C3d. CD46 also regulates the adaptive immune response by controlling T cell activation and differentiation. Co-engagement of the T cell receptor and CD46 notably drives T cell differentiation by switching production of IFNγ to secretion of anti-inflammatory IL-10. This regulatory pathway is altered in several chronic inflammatory diseases highlighting its key role for immune homeostasis. The manipulation of the CD46 pathway may therefore provide a powerful means to regulate immune responses. Herein, we investigated the effect of recombinant proteins derived from the fiber knob of the adenovirus serotype 35 (Ad35) that uses CD46 as its entry receptor, on human T cell activation. We compared the effects of Ad35K++, engineered to exhibit enhanced affinity to CD46, and of Ad35K-, mutated in the binding site for CD46. Ad35K++ profoundly affects T cell activation by decreasing the levels of CD46 at the surface of primary T cells, and impairing T cell co-activation, shown by decreased CD25 expression, reduced proliferation and lower secretion of IL-10 and IFNγ. In contrast, Ad35K- acts a potent coactivator of T cells, enhancing T cell proliferation and cytokine production. These data show that recombinant Ad35 proteins are potent modulators of human T cell activation, and support their further development as potential drugs targeting T cell responses. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Joanne Hay
- MRC Centre for Inflammation Research, University of Edinburgh, Queen’s Medical Research InstituteEdinburgh, UK
| | - Darrick Carter
- PAI Life Sciences Inc.Seattle, WA, USA
- Compliment Corp.Seattle, WA, USA
| | - André Lieber
- Department of Medical Genetics, University of WashingtonSeattle, WA, USA
| | - Anne L Astier
- MRC Centre for Inflammation Research, University of Edinburgh, Queen’s Medical Research InstituteEdinburgh, UK
| |
Collapse
|
44
|
Abstract
Regulatory T cells are the central element for the maintenance of peripheral tolerance. Several subtypes of regulatory T (Treg) cells have been described, and most of them belong to the CD4(+) T-helper (Th) cell lineage. These specific subtypes can be discriminated according to phenotype and function. Forkhead box protein 3 (FoxP3)-expressing natural Treg cells (Tregs) and IL-10-producing, T-regulatory type 1 cells (Tr1) are the best-studied types of CD4(+) regulatory T cells in humans and experimental animal models. It was shown that they play a crucial role during autoimmune neuroinflammation. Both cells types seem to be particularly important for multiple sclerosis (MS). Here, we discuss the role of CD4(+) regulatory T cells in autoimmune neuroinflammation with an emphasis on Tregs and Tr1 cells in MS.
Collapse
Affiliation(s)
- Markus Kleinewietfeld
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
- Faculty of Medicine, Dresden University of Technology (TUD), Dresden, Germany
| | - David A. Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
45
|
Jurisch-Yaksi N, Sannerud R, Annaert W. A fast growing spectrum of biological functions of γ-secretase in development and disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:2815-27. [PMID: 24099003 DOI: 10.1016/j.bbamem.2013.04.016] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 04/03/2013] [Accepted: 04/11/2013] [Indexed: 12/17/2022]
Abstract
γ-secretase, which assembles as a tetrameric complex, is an aspartyl protease that proteolytically cleaves substrate proteins within their membrane-spanning domain; a process also known as regulated intramembrane proteolysis (RIP). RIP regulates signaling pathways by abrogating or releasing signaling molecules. Since the discovery, already >15 years ago, of its catalytic component, presenilin, and even much earlier with the identification of amyloid precursor protein as its first substrate, γ-secretase has been commonly associated with Alzheimer's disease. However, starting with Notch and thereafter a continuously increasing number of novel substrates, γ-secretase is becoming linked to an equally broader range of biological processes. This review presents an updated overview of the current knowledge on the diverse molecular mechanisms and signaling pathways controlled by γ-secretase, with a focus on organ development, homeostasis and dysfunction. This article is part of a Special Issue entitled: Intramembrane Proteases.
Collapse
Affiliation(s)
- Nathalie Jurisch-Yaksi
- Laboratory for Membrane Trafficking, VIB-Center for the Biology of Disease & Department for Human Genetics (KU Leuven), Leuven, Belgium
| | | | | |
Collapse
|
46
|
Liszewski MK, Kolev M, Le Friec G, Leung M, Bertram PG, Fara AF, Subias M, Pickering MC, Drouet C, Meri S, Arstila TP, Pekkarinen PT, Ma M, Cope A, Reinheckel T, Rodriguez de Cordoba S, Afzali B, Atkinson JP, Kemper C. Intracellular complement activation sustains T cell homeostasis and mediates effector differentiation. Immunity 2013; 39:1143-57. [PMID: 24315997 PMCID: PMC3865363 DOI: 10.1016/j.immuni.2013.10.018] [Citation(s) in RCA: 433] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 10/19/2013] [Indexed: 01/27/2023]
Abstract
Complement is viewed as a critical serum-operative component of innate immunity, with processing of its key component, C3, into activation fragments C3a and C3b confined to the extracellular space. We report here that C3 activation also occurred intracellularly. We found that the T cell-expressed protease cathepsin L (CTSL) processed C3 into biologically active C3a and C3b. Resting T cells contained stores of endosomal and lysosomal C3 and CTSL and substantial amounts of CTSL-generated C3a. While “tonic” intracellular C3a generation was required for homeostatic T cell survival, shuttling of this intracellular C3-activation-system to the cell surface upon T cell stimulation induced autocrine proinflammatory cytokine production. Furthermore, T cells from patients with autoimmune arthritis demonstrated hyperactive intracellular complement activation and interferon-γ production and CTSL inhibition corrected this deregulated phenotype. Importantly, intracellular C3a was observed in all examined cell populations, suggesting that intracellular complement activation might be of broad physiological significance. Complement C3 is activated intracellularly in human T cells by cathepsin L Intracellular C3 activation mediates cell survival and Th1 induction Increased intracellular C3 activation underlies T effector dysregulation in arthritis Patients with serum C3-deficiency retain intracellular C3a generation
Collapse
Affiliation(s)
- M Kathryn Liszewski
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Martin Kolev
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Gaelle Le Friec
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Marilyn Leung
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Paula G Bertram
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Antonella F Fara
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Marta Subias
- Departamento de Immunología, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid 28006, Spain
| | - Matthew C Pickering
- Centre for Complement and Inflammation Research, Imperial College, London SW7 2AZ, UK
| | - Christian Drouet
- Université Joseph Fourier, GREPI/AGIM CNRS FRE3405, Grenoble, F-38041, France
| | - Seppo Meri
- Haartman Institute and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, FI-00014, Finland
| | - T Petteri Arstila
- Haartman Institute and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, FI-00014, Finland
| | - Pirkka T Pekkarinen
- Haartman Institute and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, FI-00014, Finland
| | - Margaret Ma
- Biomedical Research Centre, King's Health Partners, Guy's Hospital, London SE1 9RT, UK; Academic Department of Rheumatology, King's College London, London SE1 9RT, UK
| | - Andrew Cope
- Biomedical Research Centre, King's Health Partners, Guy's Hospital, London SE1 9RT, UK; Academic Department of Rheumatology, King's College London, London SE1 9RT, UK
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, and BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, D-79104, Germany
| | - Santiago Rodriguez de Cordoba
- Departamento de Immunología, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid 28006, Spain
| | - Behdad Afzali
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's College London, Guy's Hospital, London SE1 9RT, UK; Biomedical Research Centre, King's Health Partners, Guy's Hospital, London SE1 9RT, UK
| | - John P Atkinson
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Claudia Kemper
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's College London, Guy's Hospital, London SE1 9RT, UK.
| |
Collapse
|
47
|
Yamamoto H, Fara AF, Dasgupta P, Kemper C. CD46: the 'multitasker' of complement proteins. Int J Biochem Cell Biol 2013; 45:2808-20. [PMID: 24120647 DOI: 10.1016/j.biocel.2013.09.016] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 09/23/2013] [Accepted: 09/30/2013] [Indexed: 12/12/2022]
Abstract
Complement is undeniably quintessential for innate immunity by detecting and eliminating infectious microorganisms. Recent work, however, highlights an equally profound impact of complement on the induction and regulation of a wide range of immune cells. In particular, the complement regulator CD46 emerges as a key sensor of immune activation and a vital modulator of adaptive immunity. In this review, we summarize the current knowledge of CD46-mediated signalling events and their functional consequences on immune-competent cells with a specific focus on those in CD4(+) T cells. We will also discuss the promises and challenges that potential therapeutic modulation of CD46 may hold and pose.
Collapse
Affiliation(s)
- Hidekazu Yamamoto
- Division of Transplant Immunology and Mucosal Biology, MRC Centre for Transplantation, King's College London, Guy's Hospital, London SE1 9RT, UK; The Urology Centre, Guy's and St. Thomas' NHS Foundations Trust, London SE1 9RT, UK
| | | | | | | |
Collapse
|
48
|
Le Friec G, Sheppard D, Whiteman P, Karsten CM, Shamoun SAT, Laing A, Bugeon L, Dallman MJ, Melchionna T, Chillakuri C, Smith RA, Drouet C, Couzi L, Fremeaux-Bacchi V, Köhl J, Waddington SN, McDonnell JM, Baker A, Handford PA, Lea SM, Kemper C. The CD46-Jagged1 interaction is critical for human TH1 immunity. Nat Immunol 2012; 13:1213-21. [PMID: 23086448 PMCID: PMC3505834 DOI: 10.1038/ni.2454] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 09/19/2012] [Indexed: 12/13/2022]
Abstract
CD46 is a complement regulator with important roles related to the immune response. CD46 functions as a pathogen receptor and is a potent costimulator for the induction of interferon-γ (IFN-γ)-secreting effector T helper type 1 (T(H)1) cells and their subsequent switch into interleukin 10 (IL-10)-producing regulatory T cells. Here we identified the Notch family member Jagged1 as a physiological ligand for CD46. Furthermore, we found that CD46 regulated the expression of Notch receptors and ligands during T cell activation and that disturbance of the CD46-Notch crosstalk impeded induction of IFN-γ and switching to IL-10. Notably, CD4(+) T cells from CD46-deficient patients and patients with hypomorphic mutations in the gene encoding Jagged1 (Alagille syndrome) failed to mount appropriate T(H)1 responses in vitro and in vivo, which suggested that CD46-Jagged1 crosstalk is responsible for the recurrent infections in subpopulations of these patients.
Collapse
Affiliation(s)
- Gaëlle Le Friec
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King’s College London, Guy’s Hospital, London, UK
| | - Devon Sheppard
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Pat Whiteman
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Christian M. Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Salley Al-Tilib Shamoun
- Child Health Clinical Academic Grouping, King’s Health Partners, Denmark Hill Campus, London, UK
| | - Adam Laing
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King’s College London, Guy’s Hospital, London, UK
| | - Laurence Bugeon
- Division of Cell and Molecular Biology, Department of Life Sciences, Imperial College London, London, UK
| | - Margaret J. Dallman
- Division of Cell and Molecular Biology, Department of Life Sciences, Imperial College London, London, UK
| | - Teresa Melchionna
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King’s College London, Guy’s Hospital, London, UK
| | | | - Richard A. Smith
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King’s College London, Guy’s Hospital, London, UK
| | - Christian Drouet
- Université Joseph Fourier, GREPI/AGIM CNRS FRE3405, CHU de Grenoble, Grenoble, France
| | - Lionel Couzi
- Nephrology-Transplantation, CHU Bordeaux, Bordeaux, France
| | - Veronique Fremeaux-Bacchi
- Cordeliers Research Center, Inserm Unite Mixte de Recherche en Sante (UMRS) 872, Paris, France
- Hopital Europeen Georges Pompidou, Service d’Immunologie Biologique, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Cellular and Molecular Immunology, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Simon N. Waddington
- Institute for Women’s Health, Gene Transfer Technology Group, University College London, London
| | - James M. McDonnell
- Randall Division of Cell & Molecular Biophysics, King’s College London, UK
| | - Alastair Baker
- Child Health Clinical Academic Grouping, King’s Health Partners, Denmark Hill Campus, London, UK
| | | | - Susan M. Lea
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Claudia Kemper
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, King’s College London, Guy’s Hospital, London, UK
| |
Collapse
|
49
|
Kickler K, Ni Choileain S, Williams A, Richards A, Astier AL. Calcitriol modulates the CD46 pathway in T cells. PLoS One 2012; 7:e48486. [PMID: 23144765 PMCID: PMC3483209 DOI: 10.1371/journal.pone.0048486] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 10/02/2012] [Indexed: 11/19/2022] Open
Abstract
The complement regulator CD46 is a costimulatory molecule for human T cells that induces a regulatory Tr1 phenotype, characterized by large amounts of IL-10 secretion. Secretion of IL-10 upon CD46 costimulation is largely impaired in T cells from patients with multiple sclerosis (MS). Vitamin D can exert a direct effect on T cells, and may be beneficial in several pathologies, including MS. In this pilot study, we examined whether active vitamin D (1,25(OH)(2)D(3) or calcitriol) could modulate the CD46 pathway and restore IL-10 production by CD46-costimulated CD4+ T cells from patients with MS. In healthy T cells, calcitriol profoundly affects the phenotype of CD46-costimulated CD4+ T cells, by increasing the expression of CD28, CD25, CTLA-4 and Foxp3 while it concomitantly decreased CD46 expression. Similar trends were observed in MS CD4+ T cells except for CD25 for which a striking opposite effect was observed: while CD25 was normally induced on MS T cells by CD46 costimulation, addition of calcitriol consistently inhibited its induction. Despite the aberrant effect on CD25 expression, calcitriol increased the IL-10:IFNγ ratio, characteristic of the CD46-induced Tr1 phenotype, in both T cells from healthy donors and patients with MS. Hence, we show that calcitriol affects the CD46 pathway, and that it promotes anti-inflammatory responses mediated by CD46. Moreover, it might be beneficial for T cell responses in MS.
Collapse
Affiliation(s)
- Karoline Kickler
- MRC Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Siobhan Ni Choileain
- MRC Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, United Kingdom
- Multiple Sclerosis Research Centre, MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh Bioquarter, Edinburgh, United Kingdom
| | - Anna Williams
- Multiple Sclerosis Research Centre, MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh Bioquarter, Edinburgh, United Kingdom
| | - Anna Richards
- MRC Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Anne L. Astier
- MRC Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, United Kingdom
- Multiple Sclerosis Research Centre, MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh Bioquarter, Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
50
|
Tsai YG, Niu DM, Yang KD, Hung CH, Yeh YJ, Lee CY, Lin CY. Functional defects of CD46-induced regulatory T cells to suppress airway inflammation in mite allergic asthma. J Transl Med 2012; 92:1260-9. [PMID: 22751347 DOI: 10.1038/labinvest.2012.86] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Defective recruitment of regulatory T cells (Treg) function to the airway is important in the pathogenesis of allergic asthma. Complement regulatory protein (CD46) is a newly defined costimulatory molecule for Treg activation, which together with IL-10/granzyme B production may aid in suppressing asthmatic inflammation. This study examines chemotaxis and adhesion molecule expression on CD3/CD46-activated CD4(+) T cells (Tregs) from patients with and without asthma to suppress mite allergen-induced respiratory epithelial cells inflammation and to elucidate the mechanism of CD46-mediated Treg activation. Diminished IL-10/granzyme B and CCR4 expression from CD3/CD46-activated Tregs appeared in asthmatic subjects. CD3/CD46-activated Tregs from asthma patients co-cultured with BEAS-2B cells suppressed Dermatophagoides pteronyssinus 2 induced nuclear factor-κB/p65 by cell contact inhibition. Decreased interaction of CD3/CD46-mediated Tregs and BEAS-2B cells from asthmatics was associated with downregulated phosphorylation of protein kinase B (AKT) expression. Results provide the first evidence that decreased interaction between CD46-mediated Tregs and lung epithelial cells with less IL-10/granzyme B production may cause airway inflammation in allergic asthma.
Collapse
Affiliation(s)
- Yi-Giien Tsai
- Department of Pediatrics, Changhua Christian Hospital, Changhua, Taiwan
| | | | | | | | | | | | | |
Collapse
|