1
|
Hanin A, Comi M, Sumida TS, Hafler DA. Cholesterol promotes IFNG mRNA expression in CD4 + effector/memory cells by SGK1 activation. Life Sci Alliance 2024; 7:e202402890. [PMID: 39366761 PMCID: PMC11452476 DOI: 10.26508/lsa.202402890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024] Open
Abstract
IFNγ-secreting T cells are central for the maintenance of immune surveillance within the central nervous system (CNS). It was previously reported in healthy donors that the T-cell environment in the CNS induces distinct signatures related to cytotoxic capacity, CNS trafficking, tissue adaptation, and lipid homeostasis. These findings suggested that the CNS milieu consisting predominantly of lipids mediated the metabolic conditions leading to IFNγ-secreting brain CD4 T cells. Here, we demonstrate that the supplementation of CD4+CD45RO+CXCR3+ cells with cholesterol modulates their function and increases IFNG expression. The heightened IFNG expression was mediated by the activation of the serum/glucocorticoid-regulated kinase (SGK1). Inhibition of SGK1 by a specific enzymatic inhibitor significantly reduces the expression of IFNG Our results confirm the crucial role of lipids in maintaining T-cell homeostasis and demonstrate a putative role of environmental factors to induce effector responses in CD4+ effector/memory cells. These findings offer potential avenues for further research targeting lipid pathways to modulate inflammatory conditions.
Collapse
Affiliation(s)
- Aurélie Hanin
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Sorbonne Université, Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
- AP-HP, Epilepsy Unit and Clinical Neurophysiology Department, DMU Neurosciences, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Michela Comi
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Tomokazu S Sumida
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - David A Hafler
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
2
|
Porsch F, Binder CJ. Autoimmune diseases and atherosclerotic cardiovascular disease. Nat Rev Cardiol 2024; 21:780-807. [PMID: 38937626 DOI: 10.1038/s41569-024-01045-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/28/2024] [Indexed: 06/29/2024]
Abstract
Autoimmune diseases are associated with a dramatically increased risk of atherosclerotic cardiovascular disease and its clinical manifestations. The increased risk is consistent with the notion that atherogenesis is modulated by both protective and disease-promoting immune mechanisms. Notably, traditional cardiovascular risk factors such as dyslipidaemia and hypertension alone do not explain the increased risk of cardiovascular disease associated with autoimmune diseases. Several mechanisms have been implicated in mediating the autoimmunity-associated cardiovascular risk, either directly or by modulating the effect of other risk factors in a complex interplay. Aberrant leukocyte function and pro-inflammatory cytokines are central to both disease entities, resulting in vascular dysfunction, impaired resolution of inflammation and promotion of chronic inflammation. Similarly, loss of tolerance to self-antigens and the generation of autoantibodies are key features of autoimmunity but are also implicated in the maladaptive inflammatory response during atherosclerotic cardiovascular disease. Therefore, immunomodulatory therapies are potential efficacious interventions to directly reduce the risk of cardiovascular disease, and biomarkers of autoimmune disease activity could be relevant tools to stratify patients with autoimmunity according to their cardiovascular risk. In this Review, we discuss the pathophysiological aspects of the increased cardiovascular risk associated with autoimmunity and highlight the many open questions that need to be answered to develop novel therapies that specifically address this unmet clinical need.
Collapse
Affiliation(s)
- Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Hu T, Liu CH, Lei M, Zeng Q, Li L, Tang H, Zhang N. Metabolic regulation of the immune system in health and diseases: mechanisms and interventions. Signal Transduct Target Ther 2024; 9:268. [PMID: 39379377 PMCID: PMC11461632 DOI: 10.1038/s41392-024-01954-6] [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: 03/27/2024] [Revised: 07/18/2024] [Accepted: 08/11/2024] [Indexed: 10/10/2024] Open
Abstract
Metabolism, including glycolysis, oxidative phosphorylation, fatty acid oxidation, and other metabolic pathways, impacts the phenotypes and functions of immune cells. The metabolic regulation of the immune system is important in the pathogenesis and progression of numerous diseases, such as cancers, autoimmune diseases and metabolic diseases. The concept of immunometabolism was introduced over a decade ago to elucidate the intricate interplay between metabolism and immunity. The definition of immunometabolism has expanded from chronic low-grade inflammation in metabolic diseases to metabolic reprogramming of immune cells in various diseases. With immunometabolism being proposed and developed, the metabolic regulation of the immune system can be gradually summarized and becomes more and more clearer. In the context of many diseases including cancer, autoimmune diseases, metabolic diseases, and many other disease, metabolic reprogramming occurs in immune cells inducing proinflammatory or anti-inflammatory effects. The phenotypic and functional changes of immune cells caused by metabolic regulation further affect and development of diseases. Based on experimental results, targeting cellular metabolism of immune cells becomes a promising therapy. In this review, we focus on immune cells to introduce their metabolic pathways and metabolic reprogramming, and summarize how these metabolic pathways affect immune effects in the context of diseases. We thoroughly explore targets and treatments based on immunometabolism in existing studies. The challenges of translating experimental results into clinical applications in the field of immunometabolism are also summarized. We believe that a better understanding of immune regulation in health and diseases will improve the management of most diseases.
Collapse
Affiliation(s)
- Tengyue Hu
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Chang-Hai Liu
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Min Lei
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qingmin Zeng
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Li Li
- Division of Renal and endocrinology, Qin Huang Hospital, Xi'an, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China.
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Nannan Zhang
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China.
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Katsuyama E, Humbel M, Suarez-Fueyo A, Satyam A, Yoshida N, Kyttaris VC, Tsokos MG, Tsokos GC. CD38 in SLE CD4 T cells promotes Ca 2+ flux and suppresses interleukin-2 production by enhancing the expression of GM2 on the surface membrane. Nat Commun 2024; 15:8304. [PMID: 39333474 PMCID: PMC11436706 DOI: 10.1038/s41467-024-52617-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 09/17/2024] [Indexed: 09/29/2024] Open
Abstract
CD38 has emerged as a potential therapeutic target for patients with systemic lupus erythematosus (SLE) but it is not known whether CD38 alters CD4+ T cell function. Using primary human T cells and CD38-sufficient and CD38-deficient Jurkat T cells, we demonstrate that CD38 shifts the T cell lipid profile of gangliosides from GM3 to GM2 by upregulating B4GALNT1 in a Sirtuin 1-dependent manner. Enhanced expression of GM2 causes ER stress by enhancing Ca2+ flux through the PLCγ1-IP3 pathway. Interestingly, correction of the calcium overload by an IP3 receptor inhibitor, but not by a store-operated calcium entry (SOCE) inhibitor, improves IL-2 production by CD4+ T cells in SLE. This study demonstrates that CD38 affects calcium homeostasis in CD4+ T cells by controlling cell membrane lipid composition that results in suppressed IL-2 production. CD38 inhibition with biologics or small drugs should be expected to benefit patients with SLE.
Collapse
Affiliation(s)
- Eri Katsuyama
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Morgane Humbel
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Abel Suarez-Fueyo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Abhigyan Satyam
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Nobuya Yoshida
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Vasileios C Kyttaris
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA.
| |
Collapse
|
5
|
Li W, Tang X, Zheng Y, Xu X, Zhao N, Tsao BP, Feng X, Sun L. Phosphatidic Acid Promoting the Generation of Interleukin-17A Producing Double-Negative T Cells by Enhancing mTORC1 Signaling in Lupus. Arthritis Rheumatol 2024; 76:1096-1108. [PMID: 38433594 DOI: 10.1002/art.42840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 12/30/2023] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE The goal was to investigate the role and intracellular regulatory mechanisms of double-negative T (DNT) cells in the pathogenesis of systemic lupus erythematosus (SLE). METHODS DNT cells were assessed in murine models, patients with SLE, and controls using flow cytometry (FCM). DNT cells from either resiquimod (R848) or vehicle-treated C57BL/6 (B6) mice were cultured with B cells from R848-treated mice to explore functions. Differential mechanistic target of rapamycin (mTOR) pathway signaling in DNT cells measured using FCM and quantitative polymerase chain reaction was validated by rapamycin inhibition. Candidate lipid metabolites detected using liquid chromatography with electrospray ionization mass spectrometry/mass spectrometry were functionally assessed in DNT cell cultures. RESULTS DNT cells were markedly increased in both spontaneous and induced mouse lupus models and in patients with SLE. Expanded DNT cells from R848-treated B6 mice produced elevated interleukin (IL)-17A and IgG with increased germinal center B (GCB) cells. Expansion of DNT cells associated with activation of mTORC1 pathway that both IL-17A levels and the number of DNT cells exhibited dose-dependent reduction with rapamycin treatment. Lipidomics studies revealed differential patterns of lipid metabolites in T cells of R848-treated mice. Among candidate metabolites, elevated phosphatidic acid (PA) that was partially controlled by phospholipase D2 increased the expression of the mTORC1 downstream target p-S6 and positively expanded IL-17A-producing DNT cells. Similarly, elevated proportions of circulating DNT cells in patients with SLE correlated with disease activity and proteinuria, and IL-17A secretion was elevated after in vitro PA stimulation. CONCLUSION The accumulation of PA in T cells could activate the mTORC1 pathway, promoting DNT cell expansion and IL-17A secretion, resulting in GCB cell abnormalities in lupus.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yuanyuan Zheng
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuefeng Xu
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Nan Zhao
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Betty P Tsao
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston
| | - Xuebing Feng
- Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lingyun Sun
- Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
6
|
Ou Y, Zhan Y, Shao X, Xu P, Ji L, Zhuang X, Chen H, Cheng Y. Lipoprotein lipids and apolipoproteins in primary immune thrombocytopenia: Results from a clinical characteristics and causal relationship verification, potential drug target identification by Mendelian randomization analyses. Br J Haematol 2024; 204:1483-1494. [PMID: 38031970 DOI: 10.1111/bjh.19229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 11/01/2023] [Accepted: 11/15/2023] [Indexed: 12/01/2023]
Abstract
Primary immune thrombocytopenia (ITP) is an acquired autoimmune disease. Cellular and systemic lipid metabolism plays a significant role in the regulation of immune cell activities. However, the role of lipoprotein lipids and apolipoproteins in ITP remains elusive. The automatic biochemistry analyser was used to measure the levels of serum total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), apolipoprotein A-I (apoA-I), apoB, apoE and lipoprotein a [LP(a)]. Genetic variants strongly associated with circulating lipoprotein lipids and apolipoproteins (LDL-C, apoB, TG, HDL-C and apoA-I) were extracted to perform Mendelian randomization (MR) analyses. Finally, drug-target MR and passive ITP mice model was used to investigate the potential druggable targets of ITP. Levels of HDL-C, apoA-I, decreased and LP(a) increased in ITP patients compared with healthy controls. Low HDL-C was causally associated with ITP susceptibility. Through drug-target MR and animal modelling, ABCA1 was identified as a potential target to design drugs for ITP. Our study found that lipid metabolism is related to ITP. The causative association between HDL-C and the risk of ITP was also established. The study provided new evidence of the aetiology of ITP. ABCA1 might be a potential drug target for ITP.
Collapse
Affiliation(s)
- Yang Ou
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yanxia Zhan
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xia Shao
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Pengcheng Xu
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Lili Ji
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xibing Zhuang
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Chen
- Department of Thoracic Surgery, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China
| | - Yunfeng Cheng
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Hematology, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Holtkamp HU, Aguergaray C, Prangnell K, Pook C, Amirapu S, Grey A, Simpson C, Nieuwoudt M, Jarrett P. Raman spectroscopy and mass spectrometry identifies a unique group of epidermal lipids in active discoid lupus erythematosus. Sci Rep 2023; 13:16452. [PMID: 37777584 PMCID: PMC10542761 DOI: 10.1038/s41598-023-43331-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 09/22/2023] [Indexed: 10/02/2023] Open
Abstract
Discoid lupus erythematosus (DLE) is the most common form of cutaneous lupus1. It can cause permanent scarring. The pathophysiology of is not fully understood. Plasmacytoid dendritic cells are found in close association with apoptotic keratinocytes inferring close cellular signalling. Matrix Associated Laser Desorption Ionisation (MALDI) combined with Fourier Transform Ion Cyclotron Resonance Mass Spectrometry (FT-ICR-MS) is an exquisitely sensitive combination to examine disease processes at the cellular and molecular level. Active areas of discoid lupus erythematosus were compared with normal perilesional skin using MALDI combined with FT-ICR-MS. A unique set of biomarkers, including epidermal lipids is identified in active discoid lupus. These were assigned as sphingomyelins, phospholipids and ceramides. Additionally, increased levels of proteins from the keratin, and small proline rich family, and aromatic amino acids (tryptophan, phenylalanine, and tyrosine) in the epidermis are observed. These techniques, applied to punch biopsies of the skin, have shown a distinctive lipid profile of active discoid lupus. This profile may indicate specific lipid signalling pathways. Lipid rich microdomains (known as lipid rafts) are involved in cell signalling and lipid abnormalities have been described with systemic lupus erythematosus which correlate with disease activity.
Collapse
Affiliation(s)
- Hannah U Holtkamp
- The Photon Factory, The University of Auckland, Auckland, New Zealand
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
- The Dodd Walls Centre for Photonic and Quantum Technologies, Dunedin, New Zealand
| | - Claude Aguergaray
- The Photon Factory, The University of Auckland, Auckland, New Zealand
- The Dodd Walls Centre for Photonic and Quantum Technologies, Dunedin, New Zealand
- Department of Physics, The University of Auckland, Auckland, New Zealand
| | - Kalita Prangnell
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Christopher Pook
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Satya Amirapu
- Department of Anatomy and Medical Imaging, The University of Auckland, Auckland, New Zealand
| | - Angus Grey
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Cather Simpson
- The Photon Factory, The University of Auckland, Auckland, New Zealand
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
- The Dodd Walls Centre for Photonic and Quantum Technologies, Dunedin, New Zealand
- Department of Physics, The University of Auckland, Auckland, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, New Zealand
| | - Michel Nieuwoudt
- The Photon Factory, The University of Auckland, Auckland, New Zealand
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
- The Dodd Walls Centre for Photonic and Quantum Technologies, Dunedin, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, New Zealand
| | - Paul Jarrett
- Department of Dermatology, Middlemore Hospital, Auckland, New Zealand.
- Department of Medicine, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
8
|
Marques-da-Silva D, Lagoa R. Rafting on the Evidence for Lipid Raft-like Domains as Hubs Triggering Environmental Toxicants' Cellular Effects. Molecules 2023; 28:6598. [PMID: 37764374 PMCID: PMC10536579 DOI: 10.3390/molecules28186598] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The plasma membrane lipid rafts are cholesterol- and sphingolipid-enriched domains that allow regularly distributed, sub-micro-sized structures englobing proteins to compartmentalize cellular processes. These membrane domains can be highly heterogeneous and dynamic, functioning as signal transduction platforms that amplify the local concentrations and signaling of individual components. Moreover, they participate in cell signaling routes that are known to be important targets of environmental toxicants affecting cell redox status and calcium homeostasis, immune regulation, and hormonal functions. In this work, the evidence that plasma membrane raft-like domains operate as hubs for toxicants' cellular actions is discussed, and suggestions for future research are provided. Several studies address the insertion of pesticides and other organic pollutants into membranes, their accumulation in lipid rafts, or lipid rafts' disruption by polychlorinated biphenyls (PCBs), benzo[a]pyrene (B[a]P), and even metals/metalloids. In hepatocytes, macrophages, or neurons, B[a]P, airborne particulate matter, and other toxicants caused rafts' protein and lipid remodeling, oxidative changes, or amyloidogenesis. Different studies investigated the role of the invaginated lipid rafts present in endothelial cells in mediating the vascular inflammatory effects of PCBs. Furthermore, in vitro and in vivo data strongly implicate raft-localized NADPH oxidases, the aryl hydrocarbon receptor, caveolin-1, and protein kinases in the toxic mechanisms of occupational and environmental chemicals.
Collapse
Affiliation(s)
- Dorinda Marques-da-Silva
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| | - Ricardo Lagoa
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| |
Collapse
|
9
|
Abstract
T cell activation is initiated by the recognition of specific antigenic peptides and subsequently accomplished by complex signaling cascades. These aspects have been extensively studied for decades as pivotal factors in the establishment of adaptive immunity. However, how receptors or signaling molecules are organized in the resting state prior to encountering antigens has received less attention. Recent advancements in super-resolution microscopy techniques have revealed topographically controlled pre-formed organization of key molecules involved in antigen recognition and signal transduction on microvillar projections of T cells before activation and substantial effort has been dedicated to characterizing the topological structure of resting T cells over the past decade. This review will summarize our current understanding of how key surface receptors are pre-organized on the T-cell plasma membrane and discuss the potential role of these receptors, which are preassembled prior to ligand binding in the early activation events of T cells.
Collapse
Affiliation(s)
- Yunmin Jung
- Department of Nano-Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science, Seoul, Republic of Korea
| |
Collapse
|
10
|
Hjazi A, Ahsan M, Alghamdi MI, Kareem AK, Al-Saidi DN, Qasim MT, Romero-Parra RM, Zabibah RS, Ramírez-Coronel AA, Mustafa YF, Hosseini-Fard SR, Karampoor S, Mirzaei R. Unraveling the impact of 27-hydroxycholesterol in autoimmune diseases: Exploring promising therapeutic approaches. Pathol Res Pract 2023; 248:154737. [PMID: 37542860 DOI: 10.1016/j.prp.2023.154737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
The role of 27-hydroxycholesterol (27-OHC) in autoimmune diseases has become a subject of intense research in recent years. This oxysterol, derived from cholesterol, has been identified as a significant player in modulating immune responses and inflammation. Its involvement in autoimmune pathogenesis has drawn attention to its potential as a therapeutic target for managing autoimmune disorders effectively. 27-OHC, an oxysterol derived from cholesterol, has emerged as a key player in modulating immune responses and inflammatory processes. It exerts its effects through various mechanisms, including activation of nuclear receptors, interaction with immune cells, and modulation of neuroinflammation. Additionally, 27-OHC has been implicated in the dysregulation of lipid metabolism, neurotoxicity, and blood-brain barrier (BBB) disruption. Understanding the intricate interplay between 27-OHC and autoimmune diseases, particularly neurodegenerative disorders, holds promise for developing targeted therapeutic strategies. Additionally, emerging evidence suggests that 27-OHC may interact with specific receptors and transcription factors, thus influencing gene expression and cellular processes in autoimmune disorders. Understanding the intricate mechanisms by which 27-OHC influences immune dysregulation and tissue damage in autoimmune diseases is crucial for developing targeted therapeutic interventions. Further investigations into the molecular pathways and signaling networks involving 27-OHC are warranted to unravel its full potential as a therapeutic target in autoimmune diseases, thereby offering new avenues for disease intervention and management.
Collapse
Affiliation(s)
- Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Maria Ahsan
- King Edward Medical University Lahore, Pakistan
| | - Mohammed I Alghamdi
- Department of Computer Science, Al-Baha University, Al-Baha City, Kingdom of Saudi Arabia
| | - A K Kareem
- Biomedical Engineering Department, Al-Mustaqbal University College, Babylon, Iraq
| | - Dahlia N Al-Saidi
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | - Maytham T Qasim
- Department of Anesthesia, College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Iraq
| | | | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Andrés Alexis Ramírez-Coronel
- Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Ecuador; University of Palermo, Buenos Aires, Argentina; Research group in educational statistics, National University of Education, Azogues, Ecuador; Epidemiology and Biostatistics Research Group, CES University, Colombia
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| | - Seyed Reza Hosseini-Fard
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
11
|
Jia L, Zhang L, Liu M, Ji H, Wen Z, Wang C. Mitochondrial Control for Healthy and Autoimmune T Cells. Cells 2023; 12:1800. [PMID: 37443834 PMCID: PMC10340733 DOI: 10.3390/cells12131800] [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: 06/07/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
T cells are critical players in adaptive immunity, driving the tissue injury and organ damage of patients with autoimmune diseases. Consequently, investigations on T cell activation, differentiation, and function are valuable in uncovering the disease pathogenesis, thus exploring promising therapeutics for autoimmune diseases. In recent decades, accumulating studies have pinpointed immunometabolism as the fundamental determinant in controlling T cell fate. Specifically, mitochondria, as a hub of intracellular metabolism, connect glucose, lipid, and amino acid metabolic pathways. Herein, we summarize metabolic adaptations of mitochondrial oxidative phosphorylation and the relevant glucose, lipid, and amino acid metabolism during T cell activation, differentiation, and function. Further, we focused on current updates of the molecular bases for metabolic reprogramming in autoimmune T cells and advances in exploring metabolic-targeted therapeutics against autoimmune diseases. This might facilitate the in-depth understanding of autoimmune pathogeneses and the clinical management of autoimmune diseases.
Collapse
Affiliation(s)
- Li Jia
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Lei Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Mengdi Liu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Huiyan Ji
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Zhenke Wen
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Chunhong Wang
- Cyrus Tang Hematology Center, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| |
Collapse
|
12
|
Psarras A, Clarke A. A cellular overview of immunometabolism in systemic lupus erythematosus. OXFORD OPEN IMMUNOLOGY 2023; 4:iqad005. [PMID: 37554724 PMCID: PMC10264559 DOI: 10.1093/oxfimm/iqad005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/16/2023] [Accepted: 05/02/2023] [Indexed: 08/10/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease, characterized by a breakdown of immune tolerance and the development of autoantibodies against nucleic self-antigens. Immunometabolism is a rapidly expanding scientific field investigating the metabolic programming of cells of the immune system. During the normal immune response, extensive reprogramming of cellular metabolism occurs, both to generate adenosine triphosphate and facilitate protein synthesis, and also to manage cellular stress. Major pathways upregulated include glycolysis, oxidative phosphorylation, the tricarboxylic acid cycle and the pentose phosphate pathway, among others. Metabolic reprogramming also occurs to aid resolution of inflammation. Immune cells of both patients with SLE and lupus-prone mice are characterized by metabolic abnormalities resulting in an altered functional and inflammatory state. Recent studies have described how metabolic reprogramming occurs in many cell populations in SLE, particularly CD4+ T cells, e.g. favouring a glycolytic profile by overactivation of the mechanistic target of rapamycin pathway. These advances have led to an increased understanding of the metabolic changes affecting the inflammatory profile of T and B cells, monocytes, dendritic cells and neutrophils, and how they contribute to autoimmunity and SLE pathogenesis. In the current review, we aim to summarize recent advances in the field of immunometabolism involved in SLE and how these could potentially lead to new therapeutic strategies in the future.
Collapse
Affiliation(s)
- Antonios Psarras
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Alexander Clarke
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| |
Collapse
|
13
|
Saadh MJ, Kazemi K, Khorramdelazad H, Mousavi MJ, Noroozi N, Masoumi M, Karami J. Role of T cells in the pathogenesis of systemic lupus erythematous: Focus on immunometabolism dysfunctions. Int Immunopharmacol 2023; 119:110246. [PMID: 37148769 DOI: 10.1016/j.intimp.2023.110246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/08/2023]
Abstract
Evidence demonstrates that T cells are implicated in developing SLE, and each of them dominantly uses distinct metabolic pathways. Indeed, intracellular enzymes and availability of specific nutrients orchestrate fate of T cells and lead to differentiation of regulatory T cells (Treg), memory T cells, helper T cells, and effector T cells. The function of T cells in inflammatory and autoimmune responses is determined by metabolic processes and activity of their enzymes. Several studies were conducted to determine metabolic abnormalities in SLE patients and clarify how these modifications could control the functions of the involved T cells. Metabolic pathways such as glycolysis, mitochondrial pathways, oxidative stress, mTOR pathway, fatty acid and amino acid metabolisms are dysregulated in SLE T cells. Moreover, immunosuppressive drugs used in treating autoimmune diseases, including SLE, could affect immunometabolism. Developing drugs to regulate autoreactive T cell metabolism could be a promising therapeutic approach for SLE treatment. Accordingly, increased knowledge about metabolic processes paves the way to understanding SLE pathogenesis better and introduces novel therapeutic options for SLE treatment. Although monotherapy with metabolic pathways modulators might not be sufficient to prevent autoimmune disease, they may be an ideal adjuvant to reduce administration doses of immunosuppressive drugs, thus reducing drug-associated adverse effects. This review summarized emerging data about T cells that are involved in SLE pathogenesis, focusing on immunometabolism dysregulation and how these modifications could affect the disease development.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Department of Basic Sciences, Faculty of Pharmacy, Middle East University, Amman, Jordan; Applied Science Private University, Amman, Jordan
| | | | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Mousavi
- Department of Hematology, School of Para-Medicine, Bushehr University of Medical Sciences, Bushehr, Iran; Student Research and Technology Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Negar Noroozi
- Student Research and Technology Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Maryam Masoumi
- Clinical Research Development Center, Shahid Beheshti Hospital, Qom University of Medical Sciences, Qom, Iran.
| | - Jafar Karami
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran.
| |
Collapse
|
14
|
Ali AA, Bagheri Y, Tian Q, You M. Advanced DNA Zipper Probes for Detecting Cell Membrane Lipid Domains. NANO LETTERS 2022; 22:7579-7587. [PMID: 36084301 PMCID: PMC10368464 DOI: 10.1021/acs.nanolett.2c02605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The cell membrane is a complex mixture of lipids, proteins, and other components. By forming dynamic lipid domains, different membrane molecules can selectively interact with each other to control cell signaling. Herein, we report several new types of lipid-DNA conjugates, termed as "DNA zippers", which can be used to measure cell membrane dynamic interactions and the formation of lipid domains. Dependent on the choice of lipid moieties, cholesterol- and sphingomyelin-conjugated DNA zippers specifically locate in and detect membrane lipid-ordered domains, while in contrast, a tocopherol-DNA zipper can be applied for the selective imaging of lipid-disordered phases. These versatile and programmable probes can be further engineered into membrane competition assays to simultaneously detect multiple types of membrane dynamic interactions. These DNA zipper probes can be broadly used to study the correlation between lipid domains and various cellular processes, such as the epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Ahsan Ausaf Ali
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Yousef Bagheri
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Qian Tian
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Mingxu You
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, USA
| |
Collapse
|
15
|
Li H, Boulougoura A, Endo Y, Tsokos GC. Abnormalities of T cells in systemic lupus erythematosus: new insights in pathogenesis and therapeutic strategies. J Autoimmun 2022; 132:102870. [PMID: 35872102 DOI: 10.1016/j.jaut.2022.102870] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 11/25/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by loss of immune tolerance and sustained production of autoantibodies. Multiple and profound T cell abnormalities in SLE are intertwined with disease expression. Both numerical and functional disturbances have been reported in main CD4+ T helper cell subsets including Th1, Th2, Th17, regulatory, and follicular helper cells. SLE CD4+ T cells are known to provide help to B cells, produce excessive IL-17 but insufficient IL-2, and infiltrate tissues. In the absence of sufficient amounts of IL-2, regulatory T cells, do not function properly to constrain inflammation. A complicated series of early signaling defects and aberrant activation of kinases and phosphatases result in complex cell phenotypes by altering the metabolic profile and the epigenetic landscape. All main metabolic pathways including glycolysis, glutaminolysis and oxidative phosphorylation are altered in T cells from lupus prone mice and patients with SLE. SLE CD8+ cytotoxic T cells display reduced cytolytic activity which accounts for higher rates of infection and the sustenance of autoimmunity. Further, CD8+ T cells in the context of rheumatic diseases lose the expression of CD8, acquire IL-17+CD4-CD8- double negative T (DNT) cell phenotype and infiltrate tissues. Herein we present an update on these T cell abnormalities along with underlying mechanisms and discuss how these advances can be exploited therapeutically. Novel strategies to correct these aberrations in T cells show promise for SLE treatment.
Collapse
Affiliation(s)
- Hao Li
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Afroditi Boulougoura
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yushiro Endo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Pan J, Zhou L, Zhang C, Xu Q, Sun Y. Targeting protein phosphatases for the treatment of inflammation-related diseases: From signaling to therapy. Signal Transduct Target Ther 2022; 7:177. [PMID: 35665742 PMCID: PMC9166240 DOI: 10.1038/s41392-022-01038-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/28/2022] [Accepted: 05/25/2022] [Indexed: 11/09/2022] Open
Abstract
Inflammation is the common pathological basis of autoimmune diseases, metabolic diseases, malignant tumors, and other major chronic diseases. Inflammation plays an important role in tissue homeostasis. On one hand, inflammation can sense changes in the tissue environment, induce imbalance of tissue homeostasis, and cause tissue damage. On the other hand, inflammation can also initiate tissue damage repair and maintain normal tissue function by resolving injury and restoring homeostasis. These opposing functions emphasize the significance of accurate regulation of inflammatory homeostasis to ameliorate inflammation-related diseases. Potential mechanisms involve protein phosphorylation modifications by kinases and phosphatases, which have a crucial role in inflammatory homeostasis. The mechanisms by which many kinases resolve inflammation have been well reviewed, whereas a systematic summary of the functions of protein phosphatases in regulating inflammatory homeostasis is lacking. The molecular knowledge of protein phosphatases, and especially the unique biochemical traits of each family member, will be of critical importance for developing drugs that target phosphatases. Here, we provide a comprehensive summary of the structure, the "double-edged sword" function, and the extensive signaling pathways of all protein phosphatases in inflammation-related diseases, as well as their potential inhibitors or activators that can be used in therapeutic interventions in preclinical or clinical trials. We provide an integrated perspective on the current understanding of all the protein phosphatases associated with inflammation-related diseases, with the aim of facilitating the development of drugs that target protein phosphatases for the treatment of inflammation-related diseases.
Collapse
Affiliation(s)
- Jie Pan
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Lisha Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Chenyang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China.
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
17
|
Muñoz-Urbano M, Quintero-González DC, Vasquez G. T cell metabolism and possible therapeutic targets in systemic lupus erythematosus: a narrative review. Immunopharmacol Immunotoxicol 2022; 44:457-470. [PMID: 35352607 DOI: 10.1080/08923973.2022.2055568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In the immunopathogenesis of systemic lupus erythematosus (SLE), there is a dysregulation of specific immune cells, including T cells. The metabolic reprogramming in T cells causes different effects. Metabolic programs are critical checkpoints in immune responses and are involved in the etiology of autoimmune disease. For instance, resting lymphocytes generate energy through oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO), whereas activated lymphocytes rapidly shift to the glycolytic pathway. Specifically, mitochondrial dysfunction, oxidative stress, abnormal metabolism (including glucose, lipid, and amino acid metabolism), and mTOR signaling are hallmarks of T lymphocyte metabolic dysfunction in SLE. Herein it is summarized how metabolic defects contribute to T cell responses in SLE, and some epigenetic alterations involved in the disease. Finally, it is shown how metabolic defects could be modified therapeutically.
Collapse
Affiliation(s)
| | | | - Gloria Vasquez
- Rheumatology Section, Universidad de Antioquia, Medellín, Colombia.,Grupo de Inmunología Celular e Inmunogenética, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
18
|
Sun W, Li P, Cai J, Ma J, Zhang X, Song Y, Liu Y. Lipid Metabolism: Immune Regulation and Therapeutic Prospectives in Systemic Lupus Erythematosus. Front Immunol 2022; 13:860586. [PMID: 35371016 PMCID: PMC8971568 DOI: 10.3389/fimmu.2022.860586] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/28/2022] [Indexed: 12/31/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous disease characterized by the production of abnormal autoantibodies and immune complexes that can affect the organ and organ systems, particularly the kidneys and the cardiovascular system. Emerging evidence suggests that dysregulated lipid metabolism, especially in key effector cells, such as T cells, B cells, and innate immune cells, exerts complex effects on the pathogenesis and progression of SLE. Beyond their important roles as membrane components and energy storage, different lipids can also modulate different cellular processes, such as proliferation, differentiation, and survival. In this review, we summarize altered lipid metabolism and the associated mechanisms involved in the pathogenesis and progression of SLE. Furthermore, we discuss the recent progress in the role of lipid metabolism as a potential therapeutic target in SLE.
Collapse
Affiliation(s)
- Wei Sun
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Southeast University, Nanjing, China
| | - Pengchong Li
- Department of Rheumatology and Clinical Immunology, The Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Beijing, China
- Department of Gastroenterology, Beijing Friendship Hospital, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease center, Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases, Capital Medical University, Beijing, China
| | - Jianping Cai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jie Ma
- Center of Biotherapy, Beijing Hospital, National Center of Gerontolog, Beijing, China
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Southeast University, Nanjing, China
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing Medical University, Nanjing, China
- *Correspondence: Yudong Liu, ; Yong Song,
| | - Yudong Liu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Center of Biotherapy, Beijing Hospital, National Center of Gerontolog, Beijing, China
- *Correspondence: Yudong Liu, ; Yong Song,
| |
Collapse
|
19
|
T Cells, Interleukin-2 and Systemic Lupus Erythematosus—From Pathophysiology to Therapy. Cells 2022; 11:cells11060980. [PMID: 35326431 PMCID: PMC8946767 DOI: 10.3390/cells11060980] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 02/06/2023] Open
Abstract
The phenotypic and functional complexities of T cells engender complicated and often confusing concepts as to how T cells ignite, accelerate and brake the inflammatory processes involved in systemic lupus erythematosus (SLE), let alone the plasticity of T cells that takes place under different immunological contexts. Nevertheless, being one of the prime survival factors of T cells, interleukin (IL)-2 plays a potentially critical role in many immunological scenarios during the pathophysiological process of SLE. Here, the pathophysiology of lupus T cells and current, as well as ongoing, therapeutic approaches of SLE that involve low-dose IL-2 administration will be highlighted. The mechanisms of IL-2 deficiency in SLE pathophysiology, the effects of low-dose IL-2 on T cells and restoration of lupus manifestations in murine SLE models, as well as the efficacy and safety of clinical trials that evaluated low-dose IL-2-containing regimens in patients with SLE will be discussed.
Collapse
|
20
|
Inokuchi JI, Nagafuku M. Gangliosides in T cell development and function of mice. Glycoconj J 2022; 39:229-238. [DOI: 10.1007/s10719-021-10037-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/19/2021] [Accepted: 12/29/2021] [Indexed: 11/30/2022]
|
21
|
Bagheri Y, Ali AA, Keshri P, Chambers J, Gershenson A, You M. Imaging Membrane Order and Dynamic Interactions in Living Cells with a DNA Zipper Probe. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202112033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yousef Bagheri
- Department of Chemistry University of Massachusetts Amherst MA 01003 USA
| | - Ahsan Ausaf Ali
- Department of Chemistry University of Massachusetts Amherst MA 01003 USA
| | - Puspam Keshri
- Department of Chemistry University of Massachusetts Amherst MA 01003 USA
| | - James Chambers
- Institute for Applied Life Sciences University of Massachusetts Amherst MA 01003 USA
| | - Anne Gershenson
- Department of Biochemistry and Molecular Biology University of Massachusetts Amherst MA 01003 USA
| | - Mingxu You
- Department of Chemistry University of Massachusetts Amherst MA 01003 USA
| |
Collapse
|
22
|
Bagheri Y, Ali AA, Keshri P, Chambers J, Gershenson A, You M. Imaging Membrane Order and Dynamic Interactions in Living Cells with a DNA Zipper Probe. Angew Chem Int Ed Engl 2022; 61:e202112033. [PMID: 34767659 PMCID: PMC8792286 DOI: 10.1002/anie.202112033] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/19/2021] [Indexed: 02/03/2023]
Abstract
The cell membrane is a dynamic and heterogeneous structure composed of distinct sub-compartments. Within these compartments, preferential interactions occur among various lipids and proteins. Currently, it is still challenging to image these short-lived membrane complexes, especially in living cells. In this work, we present a DNA-based probe, termed "DNA Zipper", which allows the membrane order and pattern of transient interactions to be imaged in living cells using standard fluorescence microscopes. By fine-tuning the length and binding affinity of DNA duplex, these probes can precisely extend the duration of membrane lipid interactions via dynamic DNA hybridization. The correlation between membrane order and the activation of T-cell receptor signaling has also been studied. These programmable DNA probes function after a brief cell incubation, which can be easily adapted to study lipid interactions and membrane order during different membrane signaling events.
Collapse
Affiliation(s)
- Yousef Bagheri
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003 (USA)
| | - Ahsan Ausaf Ali
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003 (USA)
| | - Puspam Keshri
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003 (USA)
| | - James Chambers
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003 (USA)
| | - Anne Gershenson
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, MA 01003 (USA)
| | - Mingxu You
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003 (USA)
| |
Collapse
|
23
|
Shiraz AK, Panther EJ, Reilly CM. Altered Germinal-Center Metabolism in B Cells in Autoimmunity. Metabolites 2022; 12:metabo12010040. [PMID: 35050162 PMCID: PMC8780703 DOI: 10.3390/metabo12010040] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/27/2022] Open
Abstract
B lymphocytes play an important role in the pathophysiology of many autoimmune disorders by producing autoantibodies, secreting cytokines, and presenting antigens. B cells undergo extreme physiological changes as they develop and differentiate. Aberrant function in tolerogenic checkpoints and the metabolic state of B cells might be the contributing factors to the dysfunctionality of autoimmune B cells. Understanding B-cell metabolism in autoimmunity is important as it can give rise to new treatments. Recent investigations have revealed that alterations in metabolism occur in the activation of B cells. Several reports have suggested that germinal center (GC) B cells of individuals with systemic lupus erythematosus (SLE) have altered metabolic function. GCs are unique microenvironments in which the delicate and complex process of B-cell affinity maturation occurs through somatic hypermutation (SHM) and class switching recombination (CSR) and where Bcl6 tightly regulates B-cell differentiation into memory B-cells or plasma cells. GC B cells rely heavily on glucose, fatty acids, and oxidative phosphorylation (OXPHOS) for their energy requirements. However, the complicated association between GC B cells and their metabolism is still not clearly understood. Here, we review several studies of B-cell metabolism, highlighting the significant transformations that occur in GC progression, and suggest possible approaches that may be investigated to more precisely target aberrant B-cell metabolism in SLE.
Collapse
Affiliation(s)
- Ashton K. Shiraz
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, 205 Duck Pond Drive, Blacksburg, VA 24061, USA;
- Correspondence: (A.K.S.); (C.M.R.); Tel.: +1-540-231-9365 (C.M.R.)
| | - Eric J. Panther
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, 205 Duck Pond Drive, Blacksburg, VA 24061, USA;
| | - Christopher M. Reilly
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, 205 Duck Pond Drive, Blacksburg, VA 24061, USA;
- Via College of Osteopathic Medicine, Blacksburg, VA 24060, USA
- Correspondence: (A.K.S.); (C.M.R.); Tel.: +1-540-231-9365 (C.M.R.)
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW This study reviews the mechanisms of HDL cholesterol immunomodulation in the context of the mechanisms of chronic inflammation and immunosuppression causing persistent inflammation, immunosuppression and catabolism syndrome (PICS) and describes potential therapies and gaps in current research. RECENT FINDINGS Low HDL cholesterol is predictive of acute sepsis severity and outcome. Recent research has indicated apolipoprotein is a prognostic indicator of long-term outcomes. The pathobiologic mechanisms of PICS have been elucidated in the past several years. Recent research of the interaction of HDL pathways in related chronic inflammatory diseases may provide insights into further mechanisms and therapeutic targets. SUMMARY HDL significantly influences innate and adaptive immune pathways relating to chronic disease and inflammation. Further research is needed to better characterize these interactions in the setting of PICS.
Collapse
Affiliation(s)
- Grant Barker
- Department of Emergency Medicine, University of Florida College of Medicine, Jacksonville
| | - Julia R Winer
- University of Florida College of Medicine, Gainesville, Florida
| | - Faheem W Guirgis
- Department of Emergency Medicine, University of Florida College of Medicine, Jacksonville
| | - Srinivasa Reddy
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
| |
Collapse
|
25
|
Iwata S, Tanaka Y. Therapeutic perspectives on the metabolism of lymphocytes in patients with rheumatoid arthritis and systemic lupus erythematosus. Expert Rev Clin Immunol 2021; 17:1121-1130. [PMID: 34351835 DOI: 10.1080/1744666x.2021.1964957] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The activation of autoreactive T- and B-cells and production of autoantibodies by B cells are involved in the pathogenesis of autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Recently, the concept of 'immunometabolism' has attracted significant attention. Immune cells produce large amounts of energy in the form of ATP and biosynthesize biological components such as nucleic acids and lipids via metabolic reprogramming to activate, differentiate, and exert their functions. AREAS COVERED While the mechanisms underlying the metabolism of CD4+ T cells in SLE have been extensively studied, the metabolic changes underlying B cell activation, differentiation, and function remain unclear. Drugs targeting mTOR and AMPK, such as sirolimus, rapamycin, and metformin, have shown some efficacy and tolerability in clinical trials on patients with SLE, but have not led to breakthroughs. In this review, we summarize the current knowledge on the immunometabolic mechanisms involved in SLE and RA and discuss the potential novel therapeutic drugs. EXPERT OPINION The intensity of activation of different immune cells and their metabolic kinetics vary in different autoimmune diseases; thus, understanding the disease- and cell-specific metabolic mechanisms may help in the development of clinically effective immunometabolism-targeting drugs.
Collapse
Affiliation(s)
- Shigeru Iwata
- The First Department of Internal Medicine, Assistant Professor, University of Occupational and Environmental Health, Japan, School of Medicine, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, Professor and Chairman, Deputy Director, University of Occupational and Environmental Health, Japan, the University Hospital, School of Medicine, Kitakyushu, Japan
| |
Collapse
|
26
|
Sun J, Kumar Panda P, Kumar Samal S, Ahuja R, Ajeganova S, Hafström I, Liu A, Frostegård J. Effects of Atorvastatin on T-Cell Activation and Apoptosis in Systemic Lupus Erythematosus and Novel Simulated Interactions With C-Reactive Protein and Interleukin 6. ACR Open Rheumatol 2021; 3:642-653. [PMID: 34302321 PMCID: PMC8449041 DOI: 10.1002/acr2.11305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/11/2021] [Indexed: 01/22/2023] Open
Abstract
Objective We study activation of T helper 17 (Th17) and regulatory T (Treg) cells and induction of apoptosis in cells from patients with systemic lupus erythematosus (SLE) compared with controls and effects of atorvastatin and its simulated interactions with other compounds. Methods Mononuclear cells from 10 patients with SLE and 10 controls were cultured in conditions that induce Th17 and/or Treg cell polarization and/or apoptosis and were studied by FACScan. Gene expression was determined by quantitative real‐time reverse transcription–polymerase chain reaction. Cytokines in plasma were determined by enzyme‐linked immunosorbent assay. The Search Tool for Interactions of Chemicals (STITCH) was used to retrieve information regarding the binding properties of atorvastatin. Results Among patients with SLE, the proportion of Th17 (CD4+IL17+) cells was higher compared with controls after activation, with Th17 or Treg polarizing cytokines, phorbol myristate acetate, and ionomycin. In contrast, Treg cells (CD4+CD25+CD127dim/−) frequencies were lower. CD95 stimulation induced relatively more apoptosis in Treg cells and less in Th17 cells, as compared with controls. Addition of atorvastatin normalized Th17/Treg cell balance and apoptosis induction. Accordingly, the ratio of RORC/FoxP3 decreased in patients with SLE. Interleukin 17 and interleukin 6 (IL‐6) levels were increased in patients with SLE. Atorvastatin interacted strongly with C‐reactive protein (CRP) and also significantly with IL‐6. Conclusion There is a higher proportion of Th17 cells and a lower proportion of Treg cells in patients with SLE after activation. Th17 cells were more resistant than Treg cells to CD95‐induced apoptosis in SLE. Atorvastatin normalized these effects. Our findings reveal a novel mechanism behind the imbalance of Th17/Treg cells with implications for treatment in SLE. We determine for the first time simulated interaction between atorvastatin, CRP, and IL‐6, implying a novel role of atorvastatin.
Collapse
Affiliation(s)
- Jitong Sun
- Karolinska Institutet, Stockholm, Sweden
| | | | | | - Rajeev Ahuja
- Uppsala University and Royal Institute of Technology, Stockholm, Sweden
| | - Sofia Ajeganova
- Karolinska Institutet, Stockholm, Sweden, and Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ingiäld Hafström
- Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Anquan Liu
- Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
27
|
Bonacina F, Pirillo A, Catapano AL, Norata GD. HDL in Immune-Inflammatory Responses: Implications beyond Cardiovascular Diseases. Cells 2021; 10:cells10051061. [PMID: 33947039 PMCID: PMC8146776 DOI: 10.3390/cells10051061] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022] Open
Abstract
High density lipoproteins (HDL) are heterogeneous particles composed by a vast array of proteins and lipids, mostly recognized for their cardiovascular (CV) protective effects. However, evidences from basic to clinical research have contributed to depict a role of HDL in the modulation of immune-inflammatory response thus paving the road to investigate their involvement in other diseases beyond those related to the CV system. HDL-C levels and HDL composition are indeed altered in patients with autoimmune diseases and usually associated to disease severity. At molecular levels, HDL have been shown to modulate the anti-inflammatory potential of endothelial cells and, by controlling the amount of cellular cholesterol, to interfere with the signaling through plasma membrane lipid rafts in immune cells. These findings, coupled to observations acquired from subjects carrying mutations in genes related to HDL system, have helped to elucidate the contribution of HDL beyond cholesterol efflux thus posing HDL-based therapies as a compelling interventional approach to limit the inflammatory burden of immune-inflammatory diseases.
Collapse
Affiliation(s)
- Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Angela Pirillo
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, 20092 Milan, Italy;
- IRCCS MultiMedica, Sesto S. Giovanni, 20099 Milan, Italy
| | - Alberico L. Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy;
- IRCCS MultiMedica, Sesto S. Giovanni, 20099 Milan, Italy
- Correspondence: (A.L.C.); (G.D.N.)
| | - Giuseppe D. Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy;
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, 20092 Milan, Italy;
- Correspondence: (A.L.C.); (G.D.N.)
| |
Collapse
|
28
|
Immunometabolism in systemic lupus erythematosus: Relevant pathogenetic mechanisms and potential clinical applications. J Formos Med Assoc 2021; 120:1667-1675. [PMID: 33836940 DOI: 10.1016/j.jfma.2021.03.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex, heterogeneous, systemic autoimmune disease involving a wide array of aberrant innate and adaptive immune responses. The immune microenvironment of SLE promotes the metabolic reprogramming of immune cells, leading to immune dyshomeostasis and triggering autoimmune inflammation. Different immune subsets switch from a resting state to a highly metabolic active state by alternating the redox-sensitive signaling pathway and the involved metabolic intermediates to amplify the inflammatory response, which is critical in SLE pathogenesis. In this review, we discuss abnormal metabolic changes in glucose metabolism, tricarboxylic acid cycle, and lipid and amino acid metabolism as well as mitochondrial dysfunction in immune cells in SLE. We also review studies focused on the potential targets for key molecules of metabolic pathways in SLE, such as hypoxia-inducible factor-1α, mammalian target of rapamycin, and AMP-activated protein kinase. We highlight the therapeutic rationale for targeting these pathways in treating SLE and summarize their recent clinical applications in SLE.
Collapse
|
29
|
Lamerton RE, Lightfoot A, Nieves DJ, Owen DM. The Role of Protein and Lipid Clustering in Lymphocyte Activation. Front Immunol 2021; 12:600961. [PMID: 33767692 PMCID: PMC7986720 DOI: 10.3389/fimmu.2021.600961] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 02/12/2021] [Indexed: 12/30/2022] Open
Abstract
Lymphocytes must strike a delicate balance between activating in response to signals from potentially pathogenic organisms and avoiding activation from stimuli emanating from the body's own cells. For cells, such as T or B cells, maximizing the efficiency and fidelity, whilst minimizing the crosstalk, of complex signaling pathways is crucial. One way of achieving this control is by carefully orchestrating the spatiotemporal organization of signaling molecules, thereby regulating the rates of protein-protein interactions. This is particularly true at the plasma membrane where proximal signaling events take place and the phenomenon of protein microclustering has been extensively observed and characterized. This review will focus on what is known about the heterogeneous distribution of proteins and lipids at the cell surface, illustrating how such distributions can influence signaling in health and disease. We particularly focus on nanoscale molecular organization, which has recently become accessible for study through advances in microscope technology and analysis methodology.
Collapse
Affiliation(s)
- Rachel E Lamerton
- Institute of Immunology and Immunotherapy, School of Mathematics and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| | - Abbey Lightfoot
- Institute of Immunology and Immunotherapy, School of Mathematics and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| | - Daniel J Nieves
- Institute of Immunology and Immunotherapy, School of Mathematics and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| | - Dylan M Owen
- Institute of Immunology and Immunotherapy, School of Mathematics and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
30
|
Zhang T, Hu W, Chen W. Plasma Membrane Integrates Biophysical and Biochemical Regulation to Trigger Immune Receptor Functions. Front Immunol 2021; 12:613185. [PMID: 33679752 PMCID: PMC7933204 DOI: 10.3389/fimmu.2021.613185] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/06/2021] [Indexed: 11/23/2022] Open
Abstract
Plasma membrane provides a biophysical and biochemical platform for immune cells to trigger signaling cascades and immune responses against attacks from foreign pathogens or tumor cells. Mounting evidence suggests that the biophysical-chemical properties of this platform, including complex compositions of lipids and cholesterols, membrane tension, and electrical potential, could cooperatively regulate the immune receptor functions. However, the molecular mechanism is still unclear because of the tremendous compositional complexity and spatio-temporal dynamics of the plasma membrane. Here, we review the recent significant progress of dynamical regulation of plasma membrane on immune receptors, including T cell receptor, B cell receptor, Fc receptor, and other important immune receptors, to proceed mechano-chemical sensing and transmembrane signal transduction. We also discuss how biophysical-chemical cues couple together to dynamically tune the receptor's structural conformation or orientation, distribution, and organization, thereby possibly impacting their in-situ ligand binding and related signal transduction. Moreover, we propose that electrical potential could potentially induce the biophysical-chemical coupling change, such as lipid distribution and membrane tension, to inevitably regulate immune receptor activation.
Collapse
Affiliation(s)
- Tongtong Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Hu
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Chen
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory for Biomedical Engineering of Ministry of Education, State Key Laboratory for Modern Optical Instrumentation, College of Biomedical Engineering and Instrument Science, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| |
Collapse
|
31
|
Pathan-Chhatbar S, Drechsler C, Richter K, Morath A, Wu W, OuYang B, Xu C, Schamel WW. Direct Regulation of the T Cell Antigen Receptor's Activity by Cholesterol. Front Cell Dev Biol 2021; 8:615996. [PMID: 33490080 PMCID: PMC7820176 DOI: 10.3389/fcell.2020.615996] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 12/09/2020] [Indexed: 11/14/2022] Open
Abstract
Biological membranes consist of hundreds of different lipids that together with the embedded transmembrane (TM) proteins organize themselves into small nanodomains. In addition to this function of lipids, TM regions of proteins bind to lipids in a very specific manner, but the function of these TM region-lipid interactions is mostly unknown. In this review, we focus on the role of plasma membrane cholesterol, which directly binds to the αβ T cell antigen receptor (TCR), and has at least two opposing functions in αβ TCR activation. On the one hand, cholesterol binding to the TM domain of the TCRβ subunit keeps the TCR in an inactive, non-signaling conformation by stabilizing this conformation. This assures that the αβ T cell remains quiescent in the absence of antigenic peptide-MHC (the TCR's ligand) and decreases the sensitivity of the T cell toward stimulation. On the other hand, cholesterol binding to TCRβ leads to an increased formation of TCR nanoclusters, increasing the avidity of the TCRs toward the antigen, thus increasing the sensitivity of the αβ T cell. In mouse models, pharmacological increase of the cholesterol concentration in T cells caused an increase in TCR clustering, and thereby enhanced anti-tumor responses. In contrast, the γδ TCR does not bind to cholesterol and might be regulated in a different manner. The goal of this review is to put these seemingly controversial findings on the impact of cholesterol on the αβ TCR into perspective.
Collapse
Affiliation(s)
- Salma Pathan-Chhatbar
- Centre for Biological Signalling Studies and Centre for Integrative Biological Signalling Studies, University Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), University of Freiburg, Freiburg, Germany
| | - Carina Drechsler
- Centre for Biological Signalling Studies and Centre for Integrative Biological Signalling Studies, University Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), University of Freiburg, Freiburg, Germany
| | - Kirsten Richter
- Immunology, Infectious Diseases and Ophthalmology Disease Translational Area, Roche Innovation Center Basel, Basel, Switzerland
| | - Anna Morath
- Centre for Biological Signalling Studies and Centre for Integrative Biological Signalling Studies, University Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), University of Freiburg, Freiburg, Germany
| | - Wei Wu
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Bo OuYang
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Chenqi Xu
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Wolfgang W. Schamel
- Centre for Biological Signalling Studies and Centre for Integrative Biological Signalling Studies, University Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), University of Freiburg, Freiburg, Germany
| |
Collapse
|
32
|
Abstract
The immunopathogenesis of rheumatoid arthritis (RA) spans decades, beginning with the production of autoantibodies against post-translationally modified proteins (checkpoint 1). After years of asymptomatic autoimmunity and progressive immune system remodeling, tissue tolerance erodes and joint inflammation ensues as tissue-invasive effector T cells emerge and protective joint-resident macrophages fail (checkpoint 2). The transition of synovial stromal cells into autoaggressive effector cells converts synovitis from acute to chronic destructive (checkpoint 3). The loss of T cell tolerance derives from defective DNA repair, causing abnormal cell cycle dynamics, telomere fragility and instability of mitochondrial DNA. Mitochondrial and lysosomal anomalies culminate in the generation of short-lived tissue-invasive effector T cells. This differentiation defect builds on a metabolic platform that shunts glucose away from energy generation toward the cell building and motility programs. The next frontier in RA is the development of curative interventions, for example, reprogramming T cell defects during the period of asymptomatic autoimmunity.
Collapse
Affiliation(s)
- Cornelia M Weyand
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Jörg J Goronzy
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
33
|
Savas B, Astarita G, Aureli M, Sahali D, Ollero M. Gangliosides in Podocyte Biology and Disease. Int J Mol Sci 2020; 21:E9645. [PMID: 33348903 PMCID: PMC7766259 DOI: 10.3390/ijms21249645] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/07/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Gangliosides constitute a subgroup of glycosphingolipids characterized by the presence of sialic acid residues in their structure. As constituents of cellular membranes, in particular of raft microdomains, they exert multiple functions, some of them capital in cell homeostasis. Their presence in cells is tightly regulated by a balanced expression and function of the enzymes responsible for their biosynthesis, ganglioside synthases, and their degradation, glycosidases. The dysregulation of their abundance results in rare and common diseases. In this review, we make a point on the relevance of gangliosides and some of their metabolic precursors, such as ceramides, in the function of podocytes, the main cellular component of the glomerular filtration barrier, as well as their implications in podocytopathies. The results presented in this review suggest the pertinence of clinical lipidomic studies targeting these metabolites.
Collapse
Affiliation(s)
- Berkan Savas
- INSERM, IMRB, Univ Paris Est Créteil, F-94010 Créteil, France; (B.S.); (D.S.)
| | - Giuseppe Astarita
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, 20007 Washington, DC, USA;
| | - Massimo Aureli
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Milano Italy, 20090 Segrate (Milano), Italy;
| | - Dil Sahali
- INSERM, IMRB, Univ Paris Est Créteil, F-94010 Créteil, France; (B.S.); (D.S.)
- Service Néphrologie, AP-HP, Hôpital Henri Mondor, F-94010 Créteil, France
| | - Mario Ollero
- INSERM, IMRB, Univ Paris Est Créteil, F-94010 Créteil, France; (B.S.); (D.S.)
| |
Collapse
|
34
|
Danolic D, Heffer M, Wagner J, Skrlec I, Alvir I, Mamic I, Susnjar L, Banovic M, Danolić D, Puljiz M. Role of ganglioside biosynthesis genetic polymorphism in cervical cancer development. J OBSTET GYNAECOL 2020; 40:1127-1132. [PMID: 31847655 DOI: 10.1080/01443615.2019.1692801] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cervical cancer is the most common gynaecological cancer in women. Cell mediated immunity plays a significant role in the progression or regression of neoplastic cervical lesions caused by human papilloma virus infection. Engagement of antigen-specific T cell receptors is a prerequisite for T cell activation. The initial events of T cell activation involve the movement of the T cell receptor into specialised microdomains known as lipid rafts. Gangliosides play an active role in the formation, stabilisation and biological functions of lipid rafts. This study aims to determine whether polymorphisms in the genes involved in the biosynthesis of gangliosides represent risk a factor for cervical cancer.Taqman methods for single nucleotide polymorphism genotyping was used. All subjects carried the homozygous wild-type genotypes for all analysed genes (CC for gene B4GALT5, AA for gene ST3GAL5, AA for gene ST8SIA1 and CC for gene B4GALNT1). A χ2 test showed significant differences in genotype failure for B4GALT5 rs138960078 (χ2 = 32.02, df = 1, p = .001) and genotype failure for B4GALNT1 rs144643461 (χ2 = 41.03, df = 1, p = .001) between cervical cancer group and control group. Genotype failures were significantly more frequent in the cervical cancer group. Unknown adjacent SNPs to rs138960078 in gene B4GALT5 and rs144643461 in gene B4GALNT1 could be associated with cervical cancer development.IMPACT STATEMENTWhat is already known on this subject? Individual genetic factors play an important role in the pathogenesis of disease. In recent years, the different SNPs and their potential effects on CC risk have been extensively studied. A large number of single nucleotide genetic variants associated with cervical cancer have been identified.What do the results of this study add? Our results suggest the presence of unknown adjacent SNPs to rs138960078 in gene B4GALT5 and rs144643461 in gene B4GALNT1 that could be associated with cervical cancer development.What are the implications of these findings for clinical practice and/or further research? Better understanding of causal-consequence relationship between ganglioside biosynthesis and TCR mediated activation with consequently cervical cancer development is needed. Our research opens a new possibilities for identification of polymorphisms in the genes involved in the biosynthesis of gangliosides which can be a risk factor for cervical cancer development.
Collapse
Affiliation(s)
- Damir Danolic
- Department of Gynaecologic Oncology, University Hospital for Tumors, Clinical Hospital Centre "Sestre Milosrdnice", Zagreb, Croatia
| | - Marija Heffer
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Osijek, Osijek, Croatia
| | - Jasenka Wagner
- Department of Biology, Faculty of Dental Medicine and Health, J. J. Strossmayer University of Osijek, Osijek, Croatia
| | - Ivana Skrlec
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Osijek, Osijek, Croatia
- Department of Biology, Faculty of Dental Medicine and Health, J. J. Strossmayer University of Osijek, Osijek, Croatia
| | - Ilija Alvir
- Department of Gynaecologic Oncology, University Hospital for Tumors, Clinical Hospital Centre "Sestre Milosrdnice", Zagreb, Croatia
| | - Ivica Mamic
- Department of Gynaecologic Oncology, University Hospital for Tumors, Clinical Hospital Centre "Sestre Milosrdnice", Zagreb, Croatia
| | - Lucija Susnjar
- Department of Gynaecologic Oncology, University Hospital for Tumors, Clinical Hospital Centre "Sestre Milosrdnice", Zagreb, Croatia
| | - Marija Banovic
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | | | - Mario Puljiz
- Department of Gynaecologic Oncology, University Hospital for Tumors, Clinical Hospital Centre "Sestre Milosrdnice", Zagreb, Croatia
| |
Collapse
|
35
|
IgD-Fc-Ig fusion protein, a new biological agent, inhibits T cell function in CIA rats by inhibiting IgD-IgDR-Lck-NF-κB signaling pathways. Acta Pharmacol Sin 2020; 41:800-812. [PMID: 31937932 PMCID: PMC7470893 DOI: 10.1038/s41401-019-0337-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/18/2019] [Indexed: 12/15/2022] Open
Abstract
IgD-Fc-Ig fusion protein, a new biological agent, is constructed by linking a segment of human IgD-Fc with a segment of human IgG1-Fc, which specifically blocks the IgD-IgDR pathway and selectively inhibits the abnormal proliferation, activation, and differentiation of T cells. In this study we investigated whether IgD-Fc-Ig exerted therapeutic effects in collagen-induced arthritis (CIA) rats. CIA rats were treated with IgD-Fc-Ig (1, 3, and 9 mg/kg) or injected with biological agents etanercept (3 mg/kg) once every 3 days for 40 days. In the PBMCs and spleen lymphocytes of CIA rats, both T and B cells exhibited abnormal proliferation; the percentages of CD3+ total T cells, CD3+CD4+ Th cells, CD3+CD4+CD25+-activated Th cells, Th1(CD4+IFN-γ+), and Th17(CD4+IL-17+) were significantly increased, whereas the Treg (CD4+CD25+Foxp3+) cell percentage was decreased. IgD-Fc-Ig administration dose-dependently decreased the indicators of arthritis; alleviated the histopathology of spleen and joint; reduced serum inflammatory cytokines levels; decreased the percentages of CD3+ total T cells, CD3+CD4+ Th cells, CD3+CD4+CD25+-activated Th cells, Th1 (CD4+IFN-γ+), and Th17(CD4+IL-17+); increased Treg (CD4+CD25+Foxp3+) cell percentage; and down-regulated the expression of key molecules in IgD-IgDR-Lck-NF-κB signaling (p-Lck, p-ZAP70, p-P38, p-NF-κB65). Treatment of normal T cells with IgD (9 μg/mL) in vitro promoted their proliferation. Co-treatment with IgD-Fc-Ig (0.1–10 μg/mL) dose-dependently decreased IgD-stimulated T cell subsets percentages and down-regulated the IgD-IgDR-Lck-NF-κB signaling. In summary, this study demonstrates that IgD-Fc-Ig alleviates CIA and regulates the functions of T cells through inhibiting IgD-IgDR-Lck-NF-κB signaling.
Collapse
|
36
|
Shan J, Jin H, Xu Y. T Cell Metabolism: A New Perspective on Th17/Treg Cell Imbalance in Systemic Lupus Erythematosus. Front Immunol 2020; 11:1027. [PMID: 32528480 PMCID: PMC7257669 DOI: 10.3389/fimmu.2020.01027] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/29/2020] [Indexed: 12/20/2022] Open
Abstract
The Th17/T-regulatory (Treg) cell imbalance is involved in the occurrence and development of organ inflammation in systemic lupus erythematosus (SLE). Metabolic pathways can regulate T cell differentiation and function, thus contributing to SLE inflammation. Increasingly, data have shown metabolism influences and reprograms the Th17/Treg cell balance, and the metabolic pattern of T cells is different in SLE. Notably, metabolic characteristics of SLE T cells, such as enhanced glycolysis, lipid synthesis, glutaminolysis, and highly activated mTOR, all favored Th17 differentiation and function, which underlie the Th17/Treg cell imbalance in SLE patients. Targeting metabolic pathways to reverse Th17/Treg imbalance offer a promising method for SLE therapy.
Collapse
Affiliation(s)
- Juan Shan
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Hong Jin
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Yan Xu
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| |
Collapse
|
37
|
Abstract
Rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) are relatively common autoimmune diseases, often considered prototypic examples for how protective immunity switches to destructive immunity. The autoantigens recognized in RA and SLE are distinct, clinical manifestations are partially overlapping. A shared feature is the propensity of the adaptive immune system to respond inappropriately, with T cell hyper-responsiveness a pinnacle pathogenic defect. Upon antigen recognition, T cells mobilize a multi-pranged metabolic program, enabling them to massively expand and turn into highly mobile effector cells. Current evidence supports that T cells from patients with RA or SLE adopt metabolic programs different from healthy T cells, in line with the concept that autoimmune effector functions rely on specified pathways of energy sensing, energy generation and energy utilization. Due to misrouting of the energy sensor AMPK, RA T cells have a defect in balancing catabolic and anabolic processes and deviate towards a cell-building program. They supply biosynthetic precursors by shunting glucose away from glycolytic breakdown towards the pentose phosphate pathway and upregulate lipogenesis, enabling cellular motility and tissue invasiveness. Conversely, T cells from SLE patients are committed to high glycolytic flux, overusing the mitochondrial machinery and imposing oxidative stress. Typically, disease-relevant effector functions in SLE are associated with inappropriate activation of the key metabolic regulator mTORC1. Taken together, disease-specific metabolic signatures in RA and SLE represent vulnerabilities that are therapeutically targetable to suppress pathogenic immune responses.
Collapse
Affiliation(s)
- Bowen Wu
- School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Jörg J. Goronzy
- School of Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA 94304, USA
| | - Cornelia M. Weyand
- School of Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA 94304, USA
| |
Collapse
|
38
|
Zhang CX, Wang HY, Yin L, Mao YY, Zhou W. Immunometabolism in the pathogenesis of systemic lupus erythematosus. J Transl Autoimmun 2020; 3:100046. [PMID: 32743527 PMCID: PMC7388408 DOI: 10.1016/j.jtauto.2020.100046] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/07/2020] [Accepted: 03/08/2020] [Indexed: 12/25/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a typical autoimmune disease characterized by chronic inflammation and pathogenic auto-antibodies. Apart from B cells, dysregulation of other immune cells also plays an essential role in the pathogenesis and development of the disease including CD4+T cells, dendritic cells, macrophages and neutrophils. Since metabolic programs control immune cell fate and function, they are critical checkpoints in an effective immune response and are involved in the etiology of autoimmune disease. In addition, mitochondria and oxidative stress are both involved in cellular metabolism and is also essential in immune response. In this review, apart from the disturbed immune system, we will discuss mitochondrial dysfunction, oxidative stress, abnormal metabolism (including glucose, lipid and amino acid metabolism) of immune cells as well as epigenetic control of metabolism reprogramming to elucidate the underlying pathogenic mechanisms of systemic lupus erythematosus. Mitochondria plays a vital role in cellular metabolism and is involved in immune response. There are alterations in glucose, lipid and amino acid metabolism of various immune cells in SLE patients. Epigenetic status is influenced by the presence of metabolic intermediates and certain autoimmunity-related genes are hypomethylated in CD4+T cells, CD19+ B cells as well as CD14+ monocytes of SLE.
Collapse
Affiliation(s)
- Chen-Xing Zhang
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China
| | - Hui-Yu Wang
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, 48149, Muenster, Germany
| | - Lei Yin
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China
| | - You-Ying Mao
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China
| | - Wei Zhou
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China
| |
Collapse
|
39
|
T cell metabolism: new insights in systemic lupus erythematosus pathogenesis and therapy. Nat Rev Rheumatol 2020; 16:100-112. [PMID: 31949287 DOI: 10.1038/s41584-019-0356-x] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2019] [Indexed: 12/12/2022]
Abstract
T cell subsets are critically involved in the development of systemic autoimmunity and organ inflammation in systemic lupus erythematosus (SLE). Each T cell subset function (such as effector, helper, memory or regulatory function) is dictated by distinct metabolic pathways requiring the availability of specific nutrients and intracellular enzymes. The activity of these enzymes or nutrient transporters influences the differentiation and function of T cells in autoimmune responses. Data are increasingly emerging on how metabolic processes control the function of various T cell subsets and how these metabolic processes are altered in SLE. Specifically, aberrant glycolysis, glutaminolysis, fatty acid and glycosphingolipid metabolism, mitochondrial hyperpolarization, oxidative stress and mTOR signalling underwrite the known function of T cell subsets in patients with SLE. A number of medications that are used in the care of patients with SLE affect cell metabolism, and the development of novel therapeutic approaches to control the activity of metabolic enzymes in T cell subsets represents a promising endeavour in the search for effective treatment of systemic autoimmune diseases.
Collapse
|
40
|
Crispin JC, Hedrich CM, Suárez-Fueyo A, Comte D, Tsokos GC. SLE-Associated Defects Promote Altered T Cell Function. Crit Rev Immunol 2019; 37:39-58. [PMID: 29431078 DOI: 10.1615/critrevimmunol.2018025213] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease linked to profound defects in the function and phenotype of T lymphocytes. Here, we describe abnormal signaling pathways that have been documented in T cells from patients with SLE and discuss how they impact gene expression and immune function, in order to understand how they contribute to disease development and progression.
Collapse
Affiliation(s)
- Jose C Crispin
- Departamento de Inmunologia y Reumatologia, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Christian M Hedrich
- Department of Women's & Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK
| | - Abel Suárez-Fueyo
- Department of Rheumatology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Denis Comte
- Divisions of Immunology and Allergy, Lausanne University Hospital, Lausanne, Switzerland
| | - George C Tsokos
- Department of Rheumatology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
41
|
Comte D, Karampetsou MP, Humbel M, Tsokos GC. Signaling lymphocyte activation molecule family in systemic lupus erythematosus. Clin Immunol 2018; 204:57-63. [PMID: 30415085 DOI: 10.1016/j.clim.2018.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 11/04/2018] [Accepted: 11/05/2018] [Indexed: 01/09/2023]
Abstract
Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease characterized by a breakdown in immune tolerance leading to the development of auto-reactive lymphocytes and autoantibodies. Recent findings have provided new insight on the role of the signaling lymphocytic activation molecule family (SLAMF) receptors, a group of nine co-regulatory molecules involved in the activation of hematopoietic cells, and their downstream protein SLAM-associated protein (SAP), into the pathogenesis of SLE. This review summarizes the current knowledge on SLAMF in human SLE immunopathogenesis, and the importance of SLAMF molecules as new therapeutic targets.
Collapse
Affiliation(s)
- Denis Comte
- Divisions of Immunology and Allergy, Lausanne University Hospital, Lausanne, Switzerland.
| | | | - Morgane Humbel
- Divisions of Immunology and Allergy, Lausanne University Hospital, Lausanne, Switzerland
| | | |
Collapse
|
42
|
Nakayama H, Nagafuku M, Suzuki A, Iwabuchi K, Inokuchi JI. The regulatory roles of glycosphingolipid-enriched lipid rafts in immune systems. FEBS Lett 2018; 592:3921-3942. [PMID: 30320884 DOI: 10.1002/1873-3468.13275] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 01/04/2023]
Abstract
Lipid rafts formed by glycosphingolipids (GSLs) on cellular membranes play important roles in innate and adaptive immunity. Lactosylceramide (LacCer) forms lipid rafts on plasma and granular membranes of human neutrophils. These LacCer-enriched lipid rafts bind directly to pathogenic components, such as pathogenic fungi-derived β-glucan and Mycobacteria-derived lipoarabinomannan via carbohydrate-carbohydrate interactions, and mediate innate immune responses to these pathogens. In contrast, a-series and o-series gangliosides form distinct rafts on CD4+ and CD8+ T cell subsets, respectively, contributing to the respective functions of these cells and stimulating adaptive immune responses through T cell receptors. These findings suggest that gangliosides play indispensable roles in T cell selection and activation. This Review introduces the involvement of GSL-enriched lipid rafts in innate and adaptive immunity.
Collapse
Affiliation(s)
- Hitoshi Nakayama
- Laboratory of Biochemistry, Juntendo University Faculty of Health Care and Nursing, Urayasu, Japan.,Institute for Environmental and Gender-specific Medicine, Juntendo University Graduate School of Medicine, Urayasu, Japan
| | - Masakazu Nagafuku
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Akemi Suzuki
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kazuhisa Iwabuchi
- Laboratory of Biochemistry, Juntendo University Faculty of Health Care and Nursing, Urayasu, Japan.,Institute for Environmental and Gender-specific Medicine, Juntendo University Graduate School of Medicine, Urayasu, Japan.,Infection Control Nursing, Juntendo University Graduate School of Health Care and Nursing, Urayasu, Japan
| | - Jin-Ichi Inokuchi
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
43
|
Sawaf M, Fauny JD, Felten R, Sagez F, Gottenberg JE, Dumortier H, Monneaux F. Defective BTLA functionality is rescued by restoring lipid metabolism in lupus CD4+ T cells. JCI Insight 2018; 3:99711. [PMID: 29997289 DOI: 10.1172/jci.insight.99711] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/29/2018] [Indexed: 12/11/2022] Open
Abstract
Coinhibitory receptors play an important role in the prevention of autoimmune diseases, such as systemic lupus erythematosus (SLE), by limiting T cell activation. B and T lymphocyte attenuator (BTLA) is an inhibitory receptor, similar to cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed death 1 (PD1), that negatively regulates the immune response. The role of BTLA in the pathogenesis of autoimmune diseases in humans and, more specifically, in SLE is largely unknown. We investigated BTLA expression on various T cell subsets, and we did not observe significant variations of BTLA expression between lupus patients and healthy controls. However, the enhancement of BTLA expression after activation was significantly lower in SLE patients compared with that in healthy controls. Furthermore, we found an impaired capacity of BTLA to inhibit T cell activation in SLE due to a poor BTLA recruitment to the immunological synapse following T cell stimulation. Finally, we demonstrated that defective BTLA function can be corrected by restoring intracellular trafficking and by normalizing the lipid metabolism in lupus CD4+ T cells. Collectively, our results evidence that the BTLA signaling pathway is altered in SLE T cells and highlight the potential of targeting this pathway for the development of new therapeutic strategies in lupus.
Collapse
Affiliation(s)
- Matthieu Sawaf
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France
| | - Jean-Daniel Fauny
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France
| | - Renaud Felten
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France.,Rheumatology Department, Strasbourg University Hospital, National Reference Center for Autoimmune Diseases, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Flora Sagez
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France.,Rheumatology Department, Strasbourg University Hospital, National Reference Center for Autoimmune Diseases, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Jacques-Eric Gottenberg
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France.,Rheumatology Department, Strasbourg University Hospital, National Reference Center for Autoimmune Diseases, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Hélène Dumortier
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France
| | - Fanny Monneaux
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunologie, Immunopathologie et Chimie Thérapeutique, Strasbourg, France
| |
Collapse
|
44
|
Andersen CJ. Impact of Dietary Cholesterol on the Pathophysiology of Infectious and Autoimmune Disease. Nutrients 2018; 10:E764. [PMID: 29899295 PMCID: PMC6024721 DOI: 10.3390/nu10060764] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/02/2018] [Accepted: 06/11/2018] [Indexed: 01/02/2023] Open
Abstract
Cellular cholesterol metabolism, lipid raft formation, and lipoprotein interactions contribute to the regulation of immune-mediated inflammation and response to pathogens. Lipid pathways have been implicated in the pathogenesis of bacterial and viral infections, whereas altered lipid metabolism may contribute to immune dysfunction in autoimmune diseases, such as systemic lupus erythematosus, multiple sclerosis, and rheumatoid arthritis. Interestingly, dietary cholesterol may exert protective or detrimental effects on risk, progression, and treatment of different infectious and autoimmune diseases, although current findings suggest that these effects are variable across populations and different diseases. Research evaluating the effects of dietary cholesterol, often provided by eggs or as a component of Western-style diets, demonstrates that cholesterol-rich dietary patterns affect markers of immune inflammation and cellular cholesterol metabolism, while additionally modulating lipoprotein profiles and functional properties of HDL. Further, cholesterol-rich diets appear to differentially impact immunomodulatory lipid pathways across human populations of variable metabolic status, suggesting that these complex mechanisms may underlie the relationship between dietary cholesterol and immunity. Given the Dietary Guidelines for Americans 2015⁻2020 revision to no longer include limitations on dietary cholesterol, evaluation of dietary cholesterol recommendations beyond the context of cardiovascular disease risk is particularly timely. This review provides a comprehensive and comparative analysis of significant and controversial studies on the role of dietary cholesterol and lipid metabolism in the pathophysiology of infectious disease and autoimmune disorders, highlighting the need for further investigation in this developing area of research.
Collapse
|
45
|
Katsuyama T, Tsokos GC, Moulton VR. Aberrant T Cell Signaling and Subsets in Systemic Lupus Erythematosus. Front Immunol 2018; 9:1088. [PMID: 29868033 PMCID: PMC5967272 DOI: 10.3389/fimmu.2018.01088] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/01/2018] [Indexed: 12/20/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic multi-organ debilitating autoimmune disease, which mainly afflicts women in the reproductive years. A complex interaction of genetics, environmental factors and hormones result in the breakdown of immune tolerance to "self" leading to damage and destruction of multiple organs, such as the skin, joints, kidneys, heart and brain. Both innate and adaptive immune systems are critically involved in the misguided immune response against self-antigens. Dendritic cells, neutrophils, and innate lymphoid cells are important in initiating antigen presentation and propagating inflammation at lymphoid and peripheral tissue sites. Autoantibodies produced by B lymphocytes and immune complex deposition in vital organs contribute to tissue damage. T lymphocytes are increasingly being recognized as key contributors to disease pathogenesis. CD4 T follicular helper cells enable autoantibody production, inflammatory Th17 subsets promote inflammation, while defects in regulatory T cells lead to unchecked immune responses. A better understanding of the molecular defects including signaling events and gene regulation underlying the dysfunctional T cells in SLE is necessary to pave the path for better management, therapy, and perhaps prevention of this complex disease. In this review, we focus on the aberrations in T cell signaling in SLE and highlight therapeutic advances in this field.
Collapse
Affiliation(s)
| | | | - Vaishali R. Moulton
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
46
|
Inokuchi JI, Inamori KI, Kabayama K, Nagafuku M, Uemura S, Go S, Suzuki A, Ohno I, Kanoh H, Shishido F. Biology of GM3 Ganglioside. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 156:151-195. [PMID: 29747813 DOI: 10.1016/bs.pmbts.2017.10.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Since the successful molecular cloning in 1998 of GM3 synthase (GM3S, ST3GAL5), the enzyme responsible for initiating biosynthesis of all complex gangliosides, the efforts of our research group have been focused on clarifying the physiological and pathological implications of gangliosides, particularly GM3. We have identified isoforms of GM3S proteins having distinctive lengths of N-terminal cytoplasmic tails, and found that these cytoplasmic tails define subcellular localization, stability, and in vivo activity of GM3S isoforms. Our studies of the molecular pathogenesis of type 2 diabetes, focused on interaction between insulin receptor and GM3 in membrane microdomains, led to a novel concept: type 2 diabetes and certain other lifestyle-related diseases are membrane microdomain disorders resulting from aberrant expression of gangliosides. This concept has enhanced our understanding of the pathophysiological roles of GM3 and related gangliosides in various diseases involving chronic inflammation, such as insulin resistance, leptin resistance, and T-cell function and immune disorders (e.g., allergic asthma). We also demonstrated an essential role of GM3 in murine and human auditory systems; a common pathological feature of GM3S deficiency is deafness. This is the first direct link reported between gangliosides and auditory functions.
Collapse
Affiliation(s)
- Jin-Ichi Inokuchi
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
| | - Kei-Ichiro Inamori
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | | | - Masakazu Nagafuku
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Satoshi Uemura
- Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Shinji Go
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Akemi Suzuki
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Isao Ohno
- Center for Medical Education, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Hirotaka Kanoh
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Fumi Shishido
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| |
Collapse
|
47
|
Li W, Sivakumar R, Titov AA, Choi SC, Morel L. Metabolic Factors that Contribute to Lupus Pathogenesis. Crit Rev Immunol 2017; 36:75-98. [PMID: 27480903 DOI: 10.1615/critrevimmunol.2016017164] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease in which organ damage is mediated by pathogenic autoantibodies directed against nucleic acids and protein complexes. Studies in SLE patients and in mouse models of lupus have implicated virtually every cell type in the immune system in the induction or amplification of the autoimmune response as well as the promotion of an inflammatory environment that aggravates tissue injury. Here, we review the contribution of CD4+ T cells, B cells, and myeloid cells to lupus pathogenesis and then discuss alterations in the metabolism of these cells that may contribute to disease, given the recent advances in the field of immunometabolism.
Collapse
Affiliation(s)
- Wei Li
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610; Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology, Beijing Key Laboratory, Beijing Normal University, Beijing 100875, People's Republic of China
| | - Ramya Sivakumar
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Anton A Titov
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Seung-Chul Choi
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| |
Collapse
|
48
|
Bietz A, Zhu H, Xue M, Xu C. Cholesterol Metabolism in T Cells. Front Immunol 2017; 8:1664. [PMID: 29230226 PMCID: PMC5711771 DOI: 10.3389/fimmu.2017.01664] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/13/2017] [Indexed: 01/10/2023] Open
Abstract
Compartmentalization and spatial control of biochemical reactions is the foundation of cell-based life on earth. The lipid bilayer system employed by eukaryote cells not only keeps them separate from the environment but also provides a platform for key receptors to sense and interact with outside factors. Arguably one of the cell types most reliant on interactions of this kind, immune cells depend on their membrane to keep functioning properly. In this review, the influence of variation in cholesterol levels, a key component of lipid bilayer stability, on T cells will be discussed in detail. In comparison to other cells, T cells must be able to undergo rapid activation followed by proliferation. Furthermore, receptor colocalization is an important mechanism in this activation process. The impact of cholesterol availability on the processes of T cell proliferation and receptor sensitivity, as well as its potential for immunomodulation in disease treatment will be considered.
Collapse
Affiliation(s)
- Andreas Bietz
- State Key Laboratory of Molecular Biology, Chinese Academy Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,University of Heidelberg, Heidelberg, Germany
| | - Hengyu Zhu
- State Key Laboratory of Molecular Biology, Chinese Academy Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Manman Xue
- State Key Laboratory of Molecular Biology, Chinese Academy Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Chenqi Xu
- State Key Laboratory of Molecular Biology, Chinese Academy Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
49
|
Karampetsou MP, Comte D, Kis-Toth K, Kyttaris VC, Tsokos GC. Expression patterns of signaling lymphocytic activation molecule family members in peripheral blood mononuclear cell subsets in patients with systemic lupus erythematosus. PLoS One 2017; 12:e0186073. [PMID: 29020082 PMCID: PMC5636110 DOI: 10.1371/journal.pone.0186073] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 09/25/2017] [Indexed: 12/15/2022] Open
Abstract
Genome-wide linkage analysis studies (GWAS) studies in systemic lupus erythematosus (SLE) identified the 1q23 region on human chromosome 1, containing the Signaling Lymphocytic Activation Molecule Family (SLAMF) cluster of genes, as a lupus susceptibility locus. The SLAMF molecules (SLAMF1-7) are immunoregulatory receptors expressed predominantly on hematopoietic cells. Activation of cells of the adaptive immune system is aberrant in SLE and dysregulated expression of certain SLAMF molecules has been reported. We examined the expression of SLAMF1-7 on peripheral blood T cells, B cells, monocytes, and their respective differentiated subsets, in patients with SLE and healthy controls in a systematic manner. SLAMF1 levels were increased on both T cell and B cells and their differentiated subpopulations in patients with SLE. SLAMF2 was increased on SLE CD4+ and CD8+ T cells. The frequency of SLAMF4+ and SLAMF7+ central memory and effector memory CD8+ T cells was reduced in SLE patients. Naïve CD4+ and CD8+ SLE T cells showed a slight increase in SLAMF3 levels. No differences were seen in the expression of SLAMF5 and SLAMF6 among SLE patients and healthy controls. Overall, the expression of various SLAMF receptors is dysregulated in SLE and may contribute to the immunopathogenesis of the disease.
Collapse
Affiliation(s)
- Maria P. Karampetsou
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Denis Comte
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Katalin Kis-Toth
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Vasileios C. Kyttaris
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - George C. Tsokos
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
50
|
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease mediated by pathogenic autoantibodies directed against nucleoprotein complexes. Beyond the activation of autoreactive B cells, this process involves dysregulation in many other types of immune cells, including CD4+ T cells, dendritic cells, macrophages and neutrophils. Metabolic substrate utilization and integration of cues from energy sensors are critical checkpoints of effector functions in the immune system, with common as well as cell-specific programmes. Patients with SLE and lupus-prone mice present with activated metabolism of CD4+ T cells, and the use of metabolic inhibitors to normalize these features is associated with therapeutic effects. Far less is known about the metabolic requirements of B cells and myeloid cells in SLE. This article reviews current knowledge of the alterations in metabolism of immune cells in patients with SLE and mouse models of lupus in the context of what is known about the metabolic regulation of these cells during normal immune responses. How these alterations might contribute to lupus pathogenesis and how they can be targeted therapeutically are also discussed.
Collapse
Affiliation(s)
- Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida 32610, USA
| |
Collapse
|