1
|
Barnhart S, Shimizu-Albergine M, Kedar E, Kothari V, Shao B, Krueger M, Hsu CC, Tang J, Kanter JE, Kramer F, Djukovic D, Pascua V, Loo YM, Colonna L, Van den Bogaerde SJ, An J, Gale M, Reue K, Fisher EA, Gharib SA, Elkon KB, Bornfeldt KE. Type I IFN induces long-chain acyl-CoA synthetase 1 to generate a phosphatidic acid reservoir for lipotoxic saturated fatty acids. J Lipid Res 2024:100730. [PMID: 39675509 DOI: 10.1016/j.jlr.2024.100730] [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: 09/23/2024] [Revised: 11/22/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024] Open
Abstract
Long-chain acyl-CoA synthetase 1 (ACSL1) catalyzes the conversion of long-chain fatty acids to acyl-CoAs. ACSL1 is required for β-oxidation in tissues that rely on fatty acids as fuel, but no consensus exists on why ACSL1 is induced by inflammatory mediators in immune cells. We used a comprehensive and unbiased approach to investigate the role of ACSL1 induction by interferon type I (IFN-I) in myeloid cells in vitro and in a mouse model of IFN-I overproduction. Our results show that IFN-I induces ACSL1 in macrophages via its interferon-α/β receptor, and consequently that expression of ACSL1 is increased in myeloid cells from individuals with systemic lupus erythematosus (SLE), an autoimmune condition characterized by increased IFN production. Taking advantage of a myeloid cell-targeted ACSL1-deficient mouse model and a series of lipidomics, proteomics, metabolomics and functional analyses, we show that IFN-I leverages induction of ACSL1 to increase accumulation of fully saturated phosphatidic acid species in macrophages. Conversely, ACSL1 induction is not needed for IFN-I's ability to induce the prototypical IFN-stimulated protein signature or to suppress proliferation or macrophage metabolism. Loss of ACSL1 in IFN-I stimulated myeloid cells enhances apoptosis and secondary necrosis in vitro, especially in the presence of increased saturated fatty acid load, and in a mouse model of atherosclerosis associated with IFN overproduction, resulting in larger lesion necrotic cores. We propose that ACSL1 induction is a mechanism used by IFN-I to increase phosphatidic acid saturation while protecting the cells from saturated fatty acid-induced cell death.
Collapse
Affiliation(s)
- Shelley Barnhart
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Masami Shimizu-Albergine
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Eyal Kedar
- Division of Rheumatology, University of Washington, Seattle WA 98109
| | - Vishal Kothari
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Baohai Shao
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Melissa Krueger
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA 98109
| | - Cheng-Chieh Hsu
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Jingjing Tang
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Jenny E Kanter
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Farah Kramer
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109
| | - Danijel Djukovic
- Northwest Metabolomics Research Center and the Department of Anesthesiology and Pain Medicine, University of Washington, Seattle WA 98109
| | - Vadim Pascua
- Northwest Metabolomics Research Center and the Department of Anesthesiology and Pain Medicine, University of Washington, Seattle WA 98109
| | - Yueh-Ming Loo
- Department of Immunology, University of Washington, Seattle WA 98109
| | - Lucrezia Colonna
- Division of Rheumatology, University of Washington, Seattle WA 98109
| | | | - Jie An
- Division of Rheumatology, University of Washington, Seattle WA 98109
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle WA 98109
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016
| | - Sina A Gharib
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA 98109
| | - Keith B Elkon
- Department of Immunology, University of Washington, Seattle WA 98109
| | - Karin E Bornfeldt
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle WA 98109; UW Medicine Diabetes Institute, University of Washington, Seattle WA 98109; Department of Laboratory Medicine and Pathology, University of Washington, Seattle WA 98109.
| |
Collapse
|
2
|
Saurin S, Meineck M, Claßen P, Boedecker-Lips SC, Pautz A, Weinmann-Menke J. Sex-specific differences in SLE - Significance in the experimental setting of inflammation and kidney damage in MRL-Fas lpr mice. Autoimmunity 2024; 57:2377098. [PMID: 39004847 DOI: 10.1080/08916934.2024.2377098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024]
Abstract
Animal models are an important tool in the research of chronic autoimmune diseases, like systemic lupus erythematosus (SLE). MRL-Faslpr mice are one of different lupus models that develop spontaneously an SLE-like disease with autoantibodies and immune complex deposition that leads into damage of different organs. In contrast to human SLE, both sexes of MRL-Faslpr mice develop a similar autoimmune disease. Due to the sex bias in human and the delayed disease progression in male MRL-Faslpr mice, the majority of studies have been performed in female mice. To determine the suitability of male MRL-Faslpr mice for SLE research, especially with regard to the 3 R-principle and animal welfare, analyses of phenotype, inflammation and damage with focus on kidney and spleen were performed in mice of both sexes. Female mice developed lymphadenopathy and skin lesions earlier as males. At an age of 3.5 month, more immune cells infiltrated kidney and spleen in females compared to males. At the age of 5 months, however, substantially less sex-specific differences were detected. Since other studies have shown differences between both sexes on other manifestations like autoimmune pancreatitis and Sjögren syndrome in MRL-Faslpr mice, the use of male mice as part of 3 R-principle and animal welfare must be carefully considered.
Collapse
Affiliation(s)
- Sabrina Saurin
- Department of Nephrology and Rheumatology, Center of Immunotherapy, Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Myriam Meineck
- Department of Nephrology and Rheumatology, Center of Immunotherapy, Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Paul Claßen
- Department of Nephrology and Rheumatology, Center of Immunotherapy, Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Simone Cosima Boedecker-Lips
- Department of Nephrology and Rheumatology, Center of Immunotherapy, Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Andrea Pautz
- Institute of Pharmacology, Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Julia Weinmann-Menke
- Department of Nephrology and Rheumatology, Center of Immunotherapy, Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
3
|
Bogatyreva AI, Gerasimova EV, Kirichenko TV, Markina YV, Popkova TV, Shalygina MV, Tolstik TV, Markin AM, Orekhov AN. Proinflammatory Activation of Monocytes in Patients with Immunoinflammatory Rheumatic Diseases. DOKL BIOCHEM BIOPHYS 2024; 517:228-234. [PMID: 39002011 DOI: 10.1134/s1607672924700959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 07/15/2024]
Abstract
The pathogenesis of immunoinflammatory rheumatic diseases (IRDs) is based on chronic inflammation, one of the key mechanisms of which may be abnormal activation of macrophages, leading to further disruption of the immune system. OBJECTIVE . The objective of this study was to evaluate the proinflammatory activation of circulating monocytes in patients with IRDs. MATERIALS AND METHODS . The study involved 149 participants (53 patients with rheumatoid arthritis (RA), 45 patients with systemic lupus erythematosus (SLE), 34 patients with systemic scleroderma (SSc), and 17 participants without IRDs) 30 to 65 years old. Basal and lipopolysaccharide (LPS)-stimulated secretion of monocytes was studied in a primary culture of monocytes obtained from blood by immunomagnetic separation. Quantitative assessment of the cytokines tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β), as well as the chemokine monocyte chemoattractant protein-1 (MCP-1) was carried out in the culture fluid by ELISA. Proinflammatory activation of monocytes was calculated as the ratio of LPS-stimulated and basal secretions. RESULTS . It was shown that the basal secretion of all studied cytokines was significantly increased in all groups of patients with IRDs, except for the secretion of IL-1β in the SLE group, compared to the control. LPS-stimulated secretion of TNF-α was increased and MCP-1 was decreased in patients with IRDs compared to the control group; LPS-stimulated IL-1β secretion only in the SSc group significantly differed from the control group. In the RA group, monocyte activation was reduced for all cytokines compared to the control; in the SLE group, for TNF-α and MCP-1; in the SSc group, for MCP-1. CONCLUSIONS . The decrease in proinflammatory activation of monocytes in patients with IRDs is due to a high level of basal secretion of cytokines, which can lead to disruption of the adequate immune response in these diseases and is an important link in the pathogenesis of chronic inflammation.
Collapse
Affiliation(s)
- A I Bogatyreva
- Avtsyn Research Institute of Human Morphology, Petrovsky Russian Scientific Center of Surgery, Moscow, Russia
- Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - E V Gerasimova
- Nasonova Research Institute of Rheumatology, Moscow, Russia.
| | - T V Kirichenko
- Avtsyn Research Institute of Human Morphology, Petrovsky Russian Scientific Center of Surgery, Moscow, Russia
| | - Yu V Markina
- Avtsyn Research Institute of Human Morphology, Petrovsky Russian Scientific Center of Surgery, Moscow, Russia
| | - T V Popkova
- Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - M V Shalygina
- Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - T V Tolstik
- Avtsyn Research Institute of Human Morphology, Petrovsky Russian Scientific Center of Surgery, Moscow, Russia
| | - A M Markin
- Avtsyn Research Institute of Human Morphology, Petrovsky Russian Scientific Center of Surgery, Moscow, Russia
| | - A N Orekhov
- Avtsyn Research Institute of Human Morphology, Petrovsky Russian Scientific Center of Surgery, Moscow, Russia
| |
Collapse
|
4
|
Ferreté-Bonastre AG, Martínez-Gallo M, Morante-Palacios O, Calvillo CL, Calafell-Segura J, Rodríguez-Ubreva J, Esteller M, Cortés-Hernández J, Ballestar E. Disease activity drives divergent epigenetic and transcriptomic reprogramming of monocyte subpopulations in systemic lupus erythematosus. Ann Rheum Dis 2024; 83:865-878. [PMID: 38413168 DOI: 10.1136/ard-2023-225433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 02/15/2024] [Indexed: 02/29/2024]
Abstract
OBJECTIVES Systemic lupus erythematosus (SLE) is characterised by systemic inflammation involving various immune cell types. Monocytes, pivotal in promoting and regulating inflammation in SLE, differentiate from classic monocytes into intermediate and non-classic monocytes, assuming diverse roles and changing their proportions in inflammation. In this study, we investigated the epigenetic and transcriptomic profiles of these and novel monocyte subsets in SLE in relation to activity and progression. METHODS We obtained the DNA methylomes and transcriptomes of classic, intermediate, non-classic monocytes in patients with SLE (at first and follow-up visits) and healthy donors. We integrated these data with single-cell transcriptomics of SLE and healthy donors and interrogated their relationships with activity and progression. RESULTS In addition to shared DNA methylation and transcriptomic alterations associated with a strong interferon signature, we identified monocyte subset-specific alterations, especially in DNA methylation, which reflect an impact of SLE on monocyte differentiation. SLE classic monocytes exhibited a proinflammatory profile and were primed for macrophage differentiation. SLE non-classic monocytes displayed a T cell differentiation-related phenotype, with Th17-regulating features. Changes in monocyte proportions, DNA methylation and expression occurred in relation to disease activity and involved the STAT pathway. Integration of bulk with single-cell RNA sequencing datasets revealed disease activity-dependent expansion of SLE-specific monocyte subsets, further supported the interferon signature for classic monocytes, and associated intermediate and non-classic populations with exacerbated complement activation. CONCLUSIONS Disease activity in SLE drives a subversion of the epigenome and transcriptome programme in monocyte differentiation, impacting the function of different subsets and allowing to generate predictive methods for activity and progression.
Collapse
Affiliation(s)
| | - Mónica Martínez-Gallo
- Immunology Division, Vall d'Hebron University Hospital and Diagnostic Immunology Research Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | | | - Celia Lourdes Calvillo
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Josep Calafell-Segura
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Javier Rodríguez-Ubreva
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Cancer (CIBERONC), Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain
| | - Josefina Cortés-Hernández
- Rheumatology Department, Hospital Vall d'Hebron and Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
- Epigenetics in Inflammatory and Metabolic Diseases Laboratory, Health Science Center (HSC), East China Normal University (ECNU), Shanghai, China
| |
Collapse
|
5
|
Bruzaite A, Gedvilaite G, Balnyte R, Kriauciuniene L, Liutkeviciene R. Influence of STAT4 Genetic Variants and Serum Levels on Multiple Sclerosis Occurrence in the Lithuanian Population. J Clin Med 2024; 13:2385. [PMID: 38673659 PMCID: PMC11050845 DOI: 10.3390/jcm13082385] [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: 03/20/2024] [Revised: 04/10/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Background: Multiple sclerosis (MS) is an autoimmune disease involving demyelination, inflammation, gliosis, and the loss of neurons. MS is a growing global health problem most likely caused by genetic, immunological, and environmental factors. However, the exact etiology of the disease is still unknown. Since MS is related to a dysregulation of the immune system, it could be linked to signal transducer and activator of transcription 4 (STAT4). To fully comprehend the significance of the STAT4 gene and STAT4 serum levels in MS, further research is required. Methods: A total of 200 MS patients and 200 healthy controls participated in the study. Deoxyribonucleic acid (DNA) was extracted using silica-based membrane technology. Polymerase chain reaction was used in real time for genotyping. Using the ELISA technique, serum levels were measured. Results:STAT4 rs7601754 AA genotype and the A allele were statistically significantly less frequent in MS patients (p = 0.003). Also, rs7601754 was associated with 1.9-fold increased odds of MS occurrence (p = 0.004). The rs7601754 AG genotype was more common in males with MS (p = 0.011) and was associated with 2.5-fold increased odds of MS occurrence in males (p = 0.012). STAT4 serum levels were statistically significantly lower in MS patients compared to the control group (p = 0.007). Conclusions:STAT4 rs7601754 increases the odds of MS occurrence. STAT4 serum levels were statistically significantly lower in MS patients compared to the control group.
Collapse
Affiliation(s)
- Akvile Bruzaite
- Ophthalmology Laboratory, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu Street 2, LT-50161 Kaunas, Lithuania; (G.G.); (L.K.); (R.L.)
| | - Greta Gedvilaite
- Ophthalmology Laboratory, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu Street 2, LT-50161 Kaunas, Lithuania; (G.G.); (L.K.); (R.L.)
| | - Renata Balnyte
- Department of Neurology, Medical Academy, Lithuanian University of Health Sciences, Eiveniu Street 2, LT-50161 Kaunas, Lithuania;
| | - Loresa Kriauciuniene
- Ophthalmology Laboratory, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu Street 2, LT-50161 Kaunas, Lithuania; (G.G.); (L.K.); (R.L.)
| | - Rasa Liutkeviciene
- Ophthalmology Laboratory, Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu Street 2, LT-50161 Kaunas, Lithuania; (G.G.); (L.K.); (R.L.)
| |
Collapse
|
6
|
Játiva S, Torrico S, Calle P, Poch E, Muñoz A, García M, Larque AB, Salido MTT, Hotter G. The phagocytosis dysfunction in lupus nephritis is related to monocyte/macrophage CPT1a. Immunol Lett 2024; 266:106841. [PMID: 38331259 DOI: 10.1016/j.imlet.2024.106841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 12/20/2023] [Accepted: 02/04/2024] [Indexed: 02/10/2024]
Abstract
Macrophages must remove apoptotic cells to shield tissues from the deleterious components of dying cells. The development of chronic inflammation and autoimmune symptoms in systemic lupus is influenced by a deficiency in phagocytosis of apoptotic cells but the underlying mechanism is still unknown. Modifications in monocyte/macrophage phenotype brought on by an increase in their inflammatory phenotype would cause them to decrease the expression of CPT1a, which would reduce their ability to phagocytose, aggravating kidney damage in lupus nephritis. We aim to demonstrate that the deficiency of CPT1A in the immunological system determines lupus. For this purpose, we will monitor CPT1a expression in blood monocytes and phagocytosis and CPT1a expression of macrophages isolated from kidneys and the inflammatory state in kidneys in two experimental models of lupus nephritis such as lupus induced pristane model and in the OVA-IC in vivo model. Additionally, we will test if reestablishing CPT1a expression in tissue macrophages restores the lost phagocytic function. We evidenced that blood monocytes and macrophages isolated from kidneys in the two in vivo models have a reduced expression of CPT1a and a reduced phagocytosis. Phagocytosis could be restored only if macrophage administration leads to an increase in CPT1a expression in kidney macrophages. A new cell therapy to reduce kidney nephritis in lupus could be developed based on these results.
Collapse
Affiliation(s)
- Soraya Játiva
- Department of Experimental Pathology, Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d' Investigacions Biomèdiques August Pi i Sunyer [IIBB-CSIC-IDIBAPS], 08036, Barcelona, Spain; M2rlab-XCELL, 28010, Madrid, Spain
| | - Selene Torrico
- Department of Experimental Pathology, Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d' Investigacions Biomèdiques August Pi i Sunyer [IIBB-CSIC-IDIBAPS], 08036, Barcelona, Spain; M2rlab-XCELL, 28010, Madrid, Spain
| | - Priscila Calle
- Department of Experimental Pathology, Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d' Investigacions Biomèdiques August Pi i Sunyer [IIBB-CSIC-IDIBAPS], 08036, Barcelona, Spain; M2rlab-XCELL, 28010, Madrid, Spain
| | - Esteban Poch
- Nefrologia i Trasplantament Renal, Hospital Clínic, IDIBAPS, Universidad de Barcelona, 08036, Barcelona, Spain
| | - Angeles Muñoz
- Department of Experimental Pathology, Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d' Investigacions Biomèdiques August Pi i Sunyer [IIBB-CSIC-IDIBAPS], 08036, Barcelona, Spain
| | - Miriam García
- Department of Experimental Pathology, Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d' Investigacions Biomèdiques August Pi i Sunyer [IIBB-CSIC-IDIBAPS], 08036, Barcelona, Spain; M2rlab-XCELL, 28010, Madrid, Spain
| | - Ana Belén Larque
- Servicio de Anatomía Patológica, CDB, Hospital Clínic, 08036 Barcelona, Spain
| | - Maria Teresa Torres Salido
- Research coordinator of the cross-sectional of Internal Medicine Department, Quironsalud del Valles Hospital, Passeig de Rubió i Ors, 23, 08203, Sabadell, Barcelona, Spain; Fundacio Catalana del lupus, 08029, Barcelona, Spain
| | - Georgina Hotter
- Department of Experimental Pathology, Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas, Institut d' Investigacions Biomèdiques August Pi i Sunyer [IIBB-CSIC-IDIBAPS], 08036, Barcelona, Spain; CIBER-BBN, Networking Center on Bioengineering, Biomaterials and Nanomedicine, 50018, Zaragoza, Spain.
| |
Collapse
|
7
|
Li X, Huo Y, Wang Z. Screening of potential biomarkers of system lupus erythematosus based on WGCNA and machine learning algorithms. Medicine (Baltimore) 2023; 102:e36243. [PMID: 38013304 PMCID: PMC10681579 DOI: 10.1097/md.0000000000036243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease involving multiple systems. Its recurrent episodes and fluctuating disease courses have a severe impact on patients. Biomarkers to predict disease prognosis and remission are still lacking in SLE. We downloaded the GSE50772 dataset from the Gene Expression Omnibus database and identified differentially expressed genes (DEGs) between SLE and healthy controls. Weighted gene co-expression network analysis was used to identify key gene modules and corresponding genes in SLE. The overlapped genes in DEGs and key modules are used as key genes for subsequent analysis. These key genes were analyzed using 3 machine learning algorithms, including the least absolute shrinkage and selection operator, support vector machine recursive elimination, and random forest algorithms. The overlapped genes were obtained as potential biomarkers for further analysis, investigating and validating the potential biomarkers' possible functions, regulatory mechanisms, diagnostic value, and expression levels. And finally studied the differences between groups in level of immune cell infiltration and explored the relationship between potential biomarkers and immunity. A total of 234 overlapped genes in DEGs and key modules are used as key genes for subsequent analysis. After taking the intersection of the key genes obtained by 3 algorithms, we got 4 potential biomarkers (ARID2, CYSTM1, DDIT3, and RNASE1) with high diagnostic values. Finally, further immune infiltration analysis showed differences in various immune cells in the SLE and healthy control samples. ARID2, CYSTM1, DDIT3, and RNASE1 can affect the immune function of SLE patients. ARID2, CYSTM1, DDIT3, and RNASE1 could be used as immune-related potential biomarkers and therapeutic or diagnostic targets for further research.
Collapse
Affiliation(s)
- Xiaojian Li
- Guangxi University of Chinese Medicine, Nan Ning, Guangxi, China
| | - Yun Huo
- Guangxi International Zhuang Medical Hospital, Nan Ning, Guangxi, China
| | - Zhenchang Wang
- Guangxi University of Chinese Medicine, Nan Ning, Guangxi, China
| |
Collapse
|
8
|
Gong H, Qiu X, Li P, Zhao R, Wang B, Zhu L, Huo X. Immune infiltration analysis reveals immune cell signatures in salivary gland tissue of primary Sjögren's syndrome. Front Med (Lausanne) 2023; 10:1033232. [PMID: 36744136 PMCID: PMC9889644 DOI: 10.3389/fmed.2023.1033232] [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: 08/31/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
Introduction Mouse models are the basis for primary Sjögren's syndrome (pSS) research. However, the depth of comparisons between mice and humans in salivary gland (SG) immune cells remains limited. Methods The gene expression profiles of SGs from normal subjects and pSS patients were downloaded from the Gene Expression Comprehensive Database. The proportion of infiltrating immune cell subsets was then assessed by cell type identification by estimating relative subsets of RNA transcripts (CIBERSORT). An experimental Sjögren's syndrome (ESS) mouse model was successfully constructed using SG protein. Based on mouse SG tissue RNA-Seq data, the seq-ImmuCC model was used to quantitatively analyze the compositional ratios of 10 immune cells in pSS patients and mouse model SG tissues. Results Computed and obtained 31 human data samples using the CIBERSORT deconvolution method. The immune cell infiltration results showed that, compared to normal human SG tissue, the content of gamma delta T cells was significantly different from naive CD4+ T cells and significantly increased, while the plasma cell content decreased. Principal component analysis indicated differences in immune cell infiltration between pSS patients and normal subjects. Meanwhile, for ESS model mouse data analysis, we found that the proportion of macrophages increased, while the proportion of CD4+ T cells, B cells, and monocytes decreased. Furthermore, we found that the proportion of monocytes was decreased, while the proportion of macrophages was increased in the SG tissues of pSS patients and model mice. The infiltration of CD4+ T, CD8+ T, and B cells also showed some differences. Discussion We comprehensively analyzed SG immune infiltration in pSS patients and model mice. We demonstrated conserved and nonconserved aspects of the immune system in mice and humans at the level of immune cells to help explain the primary regulation of immune mechanisms during the development of Sjögren's syndrome.
Collapse
Affiliation(s)
- Hongxiao Gong
- Department of Scientific Research, Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China,Department of Otolaryngology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xiaoting Qiu
- Department of Scientific Research, Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China,Department of Otolaryngology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Pingping Li
- Department of Scientific Research, Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China,Department of Otolaryngology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Runzhi Zhao
- Department of Scientific Research, Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China,Department of Otolaryngology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Beijia Wang
- Department of Scientific Research, Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China,Department of Otolaryngology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Ling Zhu
- Department of Otolaryngology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China,Ling Zhu,
| | - Xingxing Huo
- Department of Scientific Research, Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China,*Correspondence: Xingxing Huo,
| |
Collapse
|
9
|
Kraemer AN, Schäfer AL, Sprenger DTL, Sehnert B, Williams JP, Luo A, Riechert L, Al-Kayyal Q, Dumortier H, Fauny JD, Winter Z, Heim K, Hofmann M, Herrmann M, Heine G, Voll RE, Chevalier N. Impact of dietary vitamin D on immunoregulation and disease pathology in lupus-prone NZB/W F1 mice. Front Immunol 2022; 13:933191. [PMID: 36505422 PMCID: PMC9730823 DOI: 10.3389/fimmu.2022.933191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 10/17/2022] [Indexed: 11/27/2022] Open
Abstract
Vitamin D (VD) deficiency is a highly prevalent worldwide phenomenon and is extensively discussed as a risk factor for the development of systemic lupus erythematosus (SLE) and other immune-mediated diseases. In addition, it is now appreciated that VD possesses multiple immunomodulatory effects. This study aims to explore the impact of dietary VD intake on lupus manifestation and pathology in lupus-prone NZB/W F1 mice and identify the underlying immunological mechanisms modulated by VD. Here, we show that low VD intake accelerates lupus progression, reflected in reduced overall survival and an earlier onset of proteinuria, as well higher concentrations of anti-double-stranded DNA autoantibodies. This unfavorable effect gained statistical significance with additional low maternal VD intake during the prenatal period. Among examined immunological effects, we found that low VD intake consistently hampered the adoption of a regulatory phenotype in lymphocytes, significantly reducing both IL-10-expressing and regulatory CD4+ T cells. This goes along with a mildly decreased frequency of IL-10-expressing B cells. We did not observe consistent effects on the phenotype and function of innate immune cells, including cytokine production, costimulatory molecule expression, and phagocytic capacity. Hence, our study reveals that low VD intake promotes lupus pathology, likely via the deviation of adaptive immunity, and suggests that the correction of VD deficiency might not only exert beneficial functions by preventing osteoporosis but also serve as an important module in prophylaxis and as an add-on in the treatment of lupus and possibly other immune-mediated diseases. Further research is required to determine the most appropriate dosage, as too-high VD serum levels may also induce adverse effects, possibly also on lupus pathology.
Collapse
Affiliation(s)
- Antoine N. Kraemer
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna-Lena Schäfer
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dalina T. L. Sprenger
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bettina Sehnert
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Johanna P. Williams
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Aileen Luo
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Laura Riechert
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Qusai Al-Kayyal
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hélène Dumortier
- Centre national de la recherche scientifique (CNRS) UPR3572, Immunology, Immunopathology and Therapeutic Chemistry, Institute of Molecular and Cellular Biology, Strasbourg, France
| | - Jean-Daniel Fauny
- Centre national de la recherche scientifique (CNRS) UPR3572, Immunology, Immunopathology and Therapeutic Chemistry, Institute of Molecular and Cellular Biology, Strasbourg, France
| | - Zoltan Winter
- Institute of Radiology, Preclinical Imaging Platform Erlangen (PIPE), Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Kathrin Heim
- Department of Gastroenterology, Hepatology, Endocrinology and Infectious Diseases, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maike Hofmann
- Department of Gastroenterology, Hepatology, Endocrinology and Infectious Diseases, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3, and Deutsches Zentrum Immuntherapie (DZI), University Medical Center Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Guido Heine
- Division of Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Reinhard E. Voll
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nina Chevalier
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
10
|
Stergioti EM, Manolakou T, Boumpas DT, Banos A. Antiviral Innate Immune Responses in Autoimmunity: Receptors, Pathways, and Therapeutic Targeting. Biomedicines 2022; 10:2820. [PMID: 36359340 PMCID: PMC9687478 DOI: 10.3390/biomedicines10112820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 09/28/2023] Open
Abstract
Innate immune receptors sense nucleic acids derived from viral pathogens or self-constituents and initiate an immune response, which involves, among other things, the secretion of cytokines including interferon (IFN) and the activation of IFN-stimulated genes (ISGs). This robust and well-coordinated immune response is mediated by the innate immune cells and is critical to preserving and restoring homeostasis. Like an antiviral response, during an autoimmune disease, aberrations of immune tolerance promote inflammatory responses to self-components, such as nucleic acids and immune complexes (ICs), leading to the secretion of cytokines, inflammation, and tissue damage. The aberrant immune response within the inflammatory milieu of the autoimmune diseases may lead to defective viral responses, predispose to autoimmunity, or precipitate a flare of an existing autoimmune disease. Herein, we review the literature on the crosstalk between innate antiviral immune responses and autoimmune responses and discuss the pitfalls and challenges regarding the therapeutic targeting of the mechanisms involved.
Collapse
Affiliation(s)
- Eirini Maria Stergioti
- Laboratory of Autoimmunity and Inflammation, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, 115 27 Athens, Greece
- School of Medicine, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Theodora Manolakou
- Laboratory of Autoimmunity and Inflammation, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, 115 27 Athens, Greece
- School of Medicine, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Dimitrios T. Boumpas
- Laboratory of Autoimmunity and Inflammation, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, 115 27 Athens, Greece
- 4th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, 124 62 Athens, Greece
| | - Aggelos Banos
- Laboratory of Autoimmunity and Inflammation, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, 115 27 Athens, Greece
| |
Collapse
|
11
|
Hsu CY, Chiu WC, Huang YL, Su YJ. Identify differential inflammatory cellular and serology pathways between children and adult patients in the lupus registry. Medicine (Baltimore) 2022; 101:e29916. [PMID: 35960068 PMCID: PMC9371509 DOI: 10.1097/md.0000000000029916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Age variances in systemic lupus erythematosus (SLE) may reflect different patterns and consequences. Monocyte differentiation is critical, and cytokine and chemokine milieu may be associated with long term outcome and treatment responses. This study aims to evaluate the inflammatory cellular and serology pathways associated with age in our lupus registry. METHODS We included patients with SLE and divided them into 2 groups according to age, ≤18 or >18 years old. We performed flow cytometry analysis to define the peripheral blood monocyte differentiation pattern and phenotypes and used the multiplex method to detect cytokine and chemokine panels. The results were then compared between the 2 subgroups. RESULTS In total, 47 SLE patients were included in this study. Of those, 23 patients were 18 years old or younger, and 24 patients were over the age of 18 years old. An increased distribution of circulating Type 2b macrophage (M2b) subsets was found in patients over 18 years old (P < 0.01), and we found the Type 1 macrophage (M1) to demonstrate a marked increase in those patients ≤18 years old (P = .05). Eotaxin values were significantly higher in patients >18 years old (P = .03), and Macrophage Inflammatory Protein (MIP)-1alpha, MIP-1beta, Interleukine (IL)-1Ra, Interferon (IFN)-alpha2, IL-12, IL-13, IL-17A, IL-1beta, IL-2, IL-4, IL-5, IL-7, IL-9, Monocyte Chemoattractant Protein (MCP)-3, Transforming Growth Factor (TGF)-alpha, and Tumor necrosis factor (TNF)-beta were significantly higher in patients ≤18 years old (all P < .05). CONCLUSIONS We found significant M2b polarization in adult SLE patients, and several cytokines and chemokines were significantly higher in SLE patients ≤ 18 years old. Peripheral blood mononuclear cell differentiation and cytokine milieu could represent composite harm from both Type 2 helper T cells (Th2) and Type 17 helper T cells (Th17) pathways and may thus be a potential therapeutic target in younger SLE patients.
Collapse
Affiliation(s)
- Chung-Yuan Hsu
- Departments of Rheumatology, Allergy and Immunology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Wen-Chan Chiu
- Departments of Rheumatology, Allergy and Immunology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Ling Huang
- Departments of Rheumatology, Allergy and Immunology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Jih Su
- Departments of Rheumatology, Allergy and Immunology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- *Correspondence: Yu-Jih Su, Departments of Rheumatology, Allergy and Immunology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan, No. 123, Ta Pei Road, Niao Sung Hsiang, Kaohsiung City 833, Taiwan (e-mail: )
| |
Collapse
|
12
|
Bose M, Jefferies C. Sex bias in systemic lupus erythematosus: a molecular insight. IMMUNOMETABOLISM (COBHAM, SURREY) 2022; 4:e00004. [PMID: 35966636 PMCID: PMC9358995 DOI: 10.1097/in9.0000000000000004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/09/2022] [Indexed: 12/24/2022]
Abstract
Acknowledging sex differences in immune response is particularly important when we consider the differences between men and women in the incidence of disease. For example, over 80% of autoimmune disease occurs in women, whereas men have a higher incidence of solid tumors compared to women. In general women have stronger innate and adaptive immune responses than men, explaining their ability to clear viral and bacterial infections faster, but also contributing to their increased susceptibility to autoimmune disease. The autoimmune disease systemic lupus erythematosus (SLE) is the archetypical sexually dimorphic disease, with 90% of patients being women. Various mechanisms have been suggested to account for the female prevalence of SLE, including sex hormones, X-linked genes, and epigenetic regulation of gene expression. Here, we will discuss how these mechanisms contribute to pathobiology of SLE and how type I interferons work with them to augment sex specific disease pathogenesis in SLE.
Collapse
Affiliation(s)
- Moumita Bose
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Caroline Jefferies
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
13
|
Giuliani AL, Sarti AC, Di Virgilio F. Ectonucleotidases in Acute and Chronic Inflammation. Front Pharmacol 2021; 11:619458. [PMID: 33613285 PMCID: PMC7887318 DOI: 10.3389/fphar.2020.619458] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
Ectonucleotidases are extracellular enzymes with a pivotal role in inflammation that hydrolyse extracellular purine and pyrimidine nucleotides, e.g., ATP, UTP, ADP, UDP, AMP and NAD+. Ectonucleotidases, expressed by virtually all cell types, immune cells included, either as plasma membrane-associated or secreted enzymes, are classified into four main families: 1) nucleoside triphosphate diphosphohydrolases (NTPDases), 2) nicotinamide adenine dinucleotide glycohydrolase (NAD glycohydrolase/ADP-ribosyl cyclase/cyclic ADP-ribose hydrolase 1), 3) ecto-5′-nucleotidase (NT5E), and 4) ecto-nucleotide pyrophosphatase/phosphodiesterases (NPPs). Concentration of ATP, UTP and NAD+ can be increased in the extracellular space thanks to un-regulated, e.g., cell damage or cell death, or regulated processes. Regulated processes include secretory exocytosis, connexin or pannexin hemichannels, ATP binding cassette (ABC) transporters, calcium homeostasis modulator (CALMH) channels, the ATP-gated P2X7 receptor, maxi-anion channels (MACs) and volume regulated ion channels (VRACs). Hydrolysis of extracellular purine nucleotides generates adenosine, an important immunosuppressant. Extracellular nucleotides and nucleosides initiate or dampen inflammation via P2 and P1 receptors, respectively. All these agents, depending on their level of expression or activation and on the agonist concentration, are potent modulators of inflammation and key promoters of host defences, immune cells activation, pathogen clearance, tissue repair and regeneration. Thus, their knowledge is of great importance for a full understanding of the pathophysiology of acute and chronic inflammatory diseases. A selection of these pathologies will be briefly discussed here.
Collapse
Affiliation(s)
- Anna Lisa Giuliani
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Alba Clara Sarti
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Francesco Di Virgilio
- Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
14
|
Jiao Y, Wu L, Huntington ND, Zhang X. Crosstalk Between Gut Microbiota and Innate Immunity and Its Implication in Autoimmune Diseases. Front Immunol 2020; 11:282. [PMID: 32153586 PMCID: PMC7047319 DOI: 10.3389/fimmu.2020.00282] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
The emerging concept of microbiota contributing to local mucosal homeostasis has fueled investigation into its specific role in immunology. Gut microbiota is mostly responsible for maintaining the balance between host defense and immune tolerance. Dysbiosis of gut microbiota has been shown to be related to various alterations of the immune system. This review focuses on the reciprocal relationship between gut microbiota and innate immunity compartment, with emphasis on gut-associated lymphoid tissue, innate lymphoid cells, and phagocytes. From a clinical perspective, the review gives a possible explanation of how the “gut microbiota—innate immunity” axis might contribute to the pathogenesis of autoimmune diseases like rheumatoid arthritis, spondyloarthritis, and systemic lupus erythematosus.
Collapse
Affiliation(s)
- Yuhao Jiao
- The Ministry of Education Key Laboratory, Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,School of Medicine, Tsinghua University, Beijing, China
| | - Li Wu
- Institute for Immunology, Tsinghua University, Beijing, China.,Tsinghua-Peking Joint Centre for Life Sciences, Beijing, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Nicholas D Huntington
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Xuan Zhang
- The Ministry of Education Key Laboratory, Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Clinical Immunology Centre, Medical Epigenetics Research Centre, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
15
|
The epigenetic face of lupus: Focus on antigen-presenting cells. Int Immunopharmacol 2020; 81:106262. [PMID: 32045873 DOI: 10.1016/j.intimp.2020.106262] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/23/2020] [Accepted: 01/27/2020] [Indexed: 12/19/2022]
Abstract
In recent years, epigenetic mechanisms became widely known due to their ability to regulate and maintain physiological processes such as cell growth, development, differentiation and genomic stability. When dysregulated, epigenetic mechanisms, may introduce gene expression changes and disturbance in immune homeostasis leading to autoimmune diseases. Systemic lupus erythematosus (SLE), the most extensively studied autoimmune disorder, has already been correlated with epigenetic modifications, especially in T cells. Since these cell rely on antigen presentation, it may be assumed that erroneous activity of antigen-presenting cells (APCs), culminates in T cell abnormalities. In this review we summarize and discuss the epigenetic modifications in SLE affected APCs, with the focus on dendritic cells (DCs), B cells and monocytes. Unravelling this aspect of SLE pathogenesis, might result in identification of new disease biomarkers and putative therapeutic approaches.
Collapse
|
16
|
Cuitino L, Obreque J, Gajardo-Meneses P, Villarroel A, Crisóstomo N, San Francisco IF, Valenzuela RA, Méndez GP, Llanos C. Heme-Oxygenase-1 Is Decreased in Circulating Monocytes and Is Associated With Impaired Phagocytosis and ROS Production in Lupus Nephritis. Front Immunol 2019; 10:2868. [PMID: 31921135 PMCID: PMC6923251 DOI: 10.3389/fimmu.2019.02868] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 11/22/2019] [Indexed: 12/18/2022] Open
Abstract
Lupus nephritis (LN) is one of the most serious manifestations of systemic lupus erythematosus (SLE). Based on studies showing the potential role of heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme and has anti-inflammatory properties in SLE development, we decided to explore HO-1 in LN. Accordingly, we evaluated HO-1 levels and function in circulating and infiltrating monocytes and neutrophils of LN patients. HO-1 levels were assessed in peripheral monocytes of LN patients and controls by flow cytometry and immunofluorescence microscopy. Phagocytosis and the production of reactive oxygen species (ROS) were evaluated to determine the effect of HO-1 in monocyte function. In addition, renal biopsies with proliferative LN were used to identify HO-1 in infiltrating cells and renal tissue by immunofluorescence and immunohistochemistry. Biopsies of healthy controls (HC) and patients who underwent nephrectomy were included as controls. Circulating pro-inflammatory monocytes and activated neutrophils were increased in LN patients. HO-1 levels were decreased in all subsets of monocytes and in activated neutrophils. LN monocytes showed increased phagocytosis and higher production of ROS than those of HC. When HO-1 was induced, phagocytosis and ROS levels became similar to those of HC. HO-1 was mostly expressed in renal tubular epithelial cells (RTEC). Renal tissue of LN patients showed lower levels of HO-1 than HC, whereas infiltrating immune cells of LN showed lower levels of HO-1 than biopsies of patients who had renal surgery. HO-1 is decreased in circulating monocytes and activated neutrophils of LN patients. HO-1 levels modulate the phagocytosis of LN monocytes and ROS production. HO-1 expression in RTEC might be an attempt of self-protection from inflammation.
Collapse
Affiliation(s)
- Loreto Cuitino
- Departamento de Inmunología Clínica y Reumatología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Javiera Obreque
- Departamento de Inmunología Clínica y Reumatología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricia Gajardo-Meneses
- Departamento de Inmunología Clínica y Reumatología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra Villarroel
- Departamento de Anatomía Patológica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Natalia Crisóstomo
- Departamento de Inmunología Clínica y Reumatología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ignacio F San Francisco
- Departamento de Urología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo A Valenzuela
- Departamento de Ciencias Químicas y Biológicas, Facultad de Salud, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Gonzalo P Méndez
- Departamento de Anatomía Patológica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina Llanos
- Departamento de Inmunología Clínica y Reumatología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
17
|
Leptin: an unappreciated key player in SLE. Clin Rheumatol 2019; 39:305-317. [PMID: 31707542 DOI: 10.1007/s10067-019-04831-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/18/2019] [Accepted: 10/23/2019] [Indexed: 02/08/2023]
Abstract
Leptin is the forerunner of the adipokine superfamily and plays a key role in regulating energy expenditure and neuroendocrine function. Researches into leptin put emphasize not only on the metabolic role but also its immunoregulatory effect on immune response through immunocyte activation and cytokine secretion. Leptin acts on receptors that are widespread throughout the body and that are expressed across many tissue types. As a consequence, the abnormal expression of leptin has been found to correlate with a number of diseases, including cancers, autoimmune diseases, and cardiovascular diseases. The significance of leptin in the development of autoimmune diseases is becoming increasingly prominent. Systemic lupus erythematosus (SLE) is a severe atypical autoimmune disease that causes damage to multiple organ systems. It is characterised by the following: impaired clearance of apoptotic cells, loss of tolerance to self-antigens, aberrant activation of T cells and B cells, and chronic inflammation. The heightened immunocyte response in SLE means that these physiological systems are particularly vulnerable to regulation by leptin in addition to being of great significance to the research field. Our current review provides insight into the regulatory roles that leptin plays on immune effector cells in SLE.
Collapse
|
18
|
Identification of U11snRNA as an endogenous agonist of TLR7-mediated immune pathogenesis. Proc Natl Acad Sci U S A 2019; 116:23653-23661. [PMID: 31694883 PMCID: PMC6876158 DOI: 10.1073/pnas.1915326116] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The activation of innate immune receptors by pathogen-associated molecular patterns (PAMPs) is central to host defense against infections. On the other hand, these receptors are also activated by immunogenic damage-associated molecular patterns (DAMPs), typically released from dying cells, and the activation can evoke chronic inflammatory or autoimmune disorders. One of the best known receptors involved in the immune pathogenesis is Toll-like receptor 7 (TLR7), which recognizes RNA with single-stranded structure. However, the causative DAMP RNA(s) in the pathogenesis has yet to be identified. Here, we first developed a chemical compound, termed KN69, that suppresses autoimmunity in several established mouse models. A subsequent search for KN69-binding partners led to the identification of U11 small nuclear RNA (U11snRNA) as a candidate DAMP RNA involved in TLR7-induced autoimmunity. We then showed that U11snRNA robustly activated the TLR7 pathway in vitro and induced arthritis disease in vivo. We also found a correlation between high serum level of U11snRNA and autoimmune diseases in human subjects and established mouse models. Finally, by revealing the structural basis for U11snRNA's ability to activate TLR7, we developed more potent TLR7 agonists and TLR7 antagonists, which may offer new therapeutic approaches for autoimmunity or other immune-driven diseases. Thus, our study has revealed a hitherto unknown immune function of U11snRNA, providing insight into TLR7-mediated autoimmunity and its potential for further therapeutic applications.
Collapse
|
19
|
Kong Y, Rastogi D, Seoighe C, Greally JM, Suzuki M. Insights from deconvolution of cell subtype proportions enhance the interpretation of functional genomic data. PLoS One 2019; 14:e0215987. [PMID: 31022271 PMCID: PMC6483354 DOI: 10.1371/journal.pone.0215987] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/11/2019] [Indexed: 02/07/2023] Open
Abstract
Cell subtype proportion variability between samples contributes significantly to the variation of functional genomic properties such as gene expression or DNA methylation. Although the impact of the variation of cell subtype composition on measured genomic quantities is recognized, and some innovative tools have been developed for the analysis of heterogeneous samples, most functional genomics studies using samples with mixed cell types still ignore the influence of cell subtype proportion variation, or just deal with it as a nuisance variable to be eliminated. Here we demonstrate how harvesting information about cell subtype proportions from functional genomics data can provide insights into cellular changes associated with phenotypes. We focused on two types of mixed cell populations, human blood and mouse kidney. Cell type prediction is well developed in the former, but not currently in the latter. Estimating the cellular repertoire is easier when a reference dataset from purified samples of all cell types in the tissue is available, as is the case for blood. However, reference datasets are not available for most other tissues, such as the kidney. In this study, we showed that the proportion of alterations attributable to changes in the cellular composition varies strikingly in the two disorders (asthma and systemic lupus erythematosus), suggesting that the contribution of cell subtype proportion changes to functional genomic properties can be disease-specific. We also showed that a reference dataset from a single-cell RNA-seq study successfully estimated the cell subtype proportions in mouse kidney and allowed us to distinguish altered cell subtype differences between two different knock-out mouse models, both of which had reported a reduced number of glomeruli compared to their wild-type counterparts. These findings demonstrate that testing for changes in cell subtype proportions between conditions can yield important insights in functional genomics studies.
Collapse
Affiliation(s)
- Yu Kong
- Department of Genetics and Center for Epigenomics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Deepa Rastogi
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Cathal Seoighe
- School of Mathematics, Statistics and Applied Mathematics, National University of Ireland Galway, University Road, Galway, Ireland
| | - John M. Greally
- Department of Genetics and Center for Epigenomics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Masako Suzuki
- Department of Genetics and Center for Epigenomics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
20
|
Abstract
The Interferon regulatory factors (IRFs) are a family of transcription factors that play pivotal roles in many aspects of the immune response, including immune cell development and differentiation and regulating responses to pathogens. Three family members, IRF3, IRF5, and IRF7, are critical to production of type I interferons downstream of pathogen recognition receptors that detect viral RNA and DNA. A fourth family member, IRF9, regulates interferon-driven gene expression. In addition, IRF4, IRF8, and IRF5 regulate myeloid cell development and phenotype, thus playing important roles in regulating inflammatory responses. Thus, understanding how their levels and activity is regulated is of critical importance given that perturbations in either can result in dysregulated immune responses and potential autoimmune disease. This review will focus the role of IRF family members in regulating type I IFN production and responses and myeloid cell development or differentiation, with particular emphasis on how regulation of their levels and activity by ubiquitination and microRNAs may impact autoimmune disease.
Collapse
Affiliation(s)
- Caroline A Jefferies
- Department of Medicine, Division of Rheumatology and Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
21
|
Horuluoglu B, Bayik D, Kayraklioglu N, Goguet E, Kaplan MJ, Klinman DM. PAM3 supports the generation of M2-like macrophages from lupus patient monocytes and improves disease outcome in murine lupus. J Autoimmun 2019; 99:24-32. [PMID: 30679006 DOI: 10.1016/j.jaut.2019.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 01/01/2023]
Abstract
Systematic Lupus Erythematosus (SLE) is an autoimmune syndrome of unclear etiology. While T and B cell abnormalities contribute to disease pathogenesis, recent work suggests that inflammatory M1-like macrophages also play a role. Previous work showed that the TLR2/1 agonist PAM3CSK4 (PAM3) could stimulate normal human monocytes to preferentially differentiate into immunosuppressive M2-like rather than inflammatory M1-like macrophages. This raised the possibility of PAM3 being used to normalize the M1:M2 ratio in SLE. Consistent with that possibility, monocytes from lupus patients differentiated into M2-like macrophages when treated with PAM3 in vitro. Furthermore, lupus-prone NZB x NZW F1 mice responded similarly to weekly PAM3 treatment. Normalization of the M2 macrophage frequency was associated with delayed disease progression, decreased autoantibody and inflammatory cytokine synthesis, reduced proteinuria and prolonged survival in NZB x NZW F1 mice. The ability of PAM3 to bias monocyte differentiation in favor of immunosuppressive macrophages may represent a novel approach to the therapy of SLE.
Collapse
Affiliation(s)
- Begum Horuluoglu
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD 21720, USA; Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Defne Bayik
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD 21720, USA
| | - Neslihan Kayraklioglu
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD 21720, USA
| | - Emilie Goguet
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD 21720, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892, USA
| | - Dennis M Klinman
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick, MD 21720, USA.
| |
Collapse
|
22
|
Torres-Ruiz J, Mejía-Domínguez NR, Zentella-Dehesa A, Ponce-de-León A, Morales-Padilla SR, Vázquez-Rodríguez R, Alvarado-Lara MR, Reyna-de-la-Garza RA, Tapia-Rodríguez M, Juárez-Vega G, Merayo-Chalico J, Barrera-Vargas A, Alcocer-Varela JC, Gómez-Martín D. The Systemic Lupus Erythematosus Infection Predictive Index (LIPI): A Clinical-Immunological Tool to Predict Infections in Lupus Patients. Front Immunol 2019; 9:3144. [PMID: 30692998 PMCID: PMC6340073 DOI: 10.3389/fimmu.2018.03144] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/19/2018] [Indexed: 12/16/2022] Open
Abstract
Among autoimmune diseases, systemic lupus erythematosus (SLE) patients have a unique predisposition to develop infections, which represents one of their main causes of morbidity and mortality. Many infections occur at disease diagnosis in the absence of immunosuppressive therapy, suggesting that the immunological abnormalities in SLE patients might be fundamental for the development of this complication. The aim of this study was to address the main clinical and immunological features associated with the development of infection and to create and validate a compound clinical-immunological infection predictive index in a cohort of SLE patients. We included 55 SLE patients with < 5 years since diagnosis. The clinical and immunological features were evaluated periodically and patients were followed-up during 1 year, searching for the development of infection. Immunophenotyping was performed by multiparametric flow cytometry and neutrophil extracellular traps (NETs) were assessed by confocal microscopy. Eighteen patients (32.7%) presented 19 infectious events, 5 (26.3%) were severe. For the construction of the index, we performed a logistic regression analysis and the cutoff points were determined with ROC curves. Increased numbers of peripheral Th17 cells, B cell lymphopenia, and lower TLR2 expression in monocytes, as well as the use of cyclophosphamide were the major risk factors for the development of infection and thus were included in the index. Besides, patients that developed infection were characterized by increased numbers of low-density granulocytes (LDGs) and higher expression of LL-37 in NETs upon infection. Finally, we validated the index retrospectively in a nested case-control study. A score >1.5 points was able to predict infection in the following year (AUC = 0.97; LR- = 0.001, specificity 100%, P = 0.0003). Our index encompasses novel immunological features able to prospectively predict the risk of infection in SLE patients.
Collapse
Affiliation(s)
- Jiram Torres-Ruiz
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Emergency Medicine Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Nancy R Mejía-Domínguez
- Bioinformatics, Biostatistics and Computational Biology Unit, Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alejandro Zentella-Dehesa
- Department of Genomic Medicine and Environmental Toxicology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alfredo Ponce-de-León
- Department of Infectology and Microbiology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sandra Rubí Morales-Padilla
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Ricardo Vázquez-Rodríguez
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Mario René Alvarado-Lara
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Roberto Adrián Reyna-de-la-Garza
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Miguel Tapia-Rodríguez
- Microscopy Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Guillermo Juárez-Vega
- Flow Cytometry Unit, Red de Apoyo a la Investigación, Coordinación de Investigación Científica, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Javier Merayo-Chalico
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Ana Barrera-Vargas
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jorge C Alcocer-Varela
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Diana Gómez-Martín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Flow Cytometry Unit, Red de Apoyo a la Investigación, Coordinación de Investigación Científica, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
23
|
Smith S, Wu PW, Seo JJ, Fernando T, Jin M, Contreras J, Montano EN, Gabhann JN, Cunningham K, Widaa A, McCarthy EM, Molloy ES, Kearns G, Murphy CC, Kong W, Björkbacka H, Kornfeld H, Forbess L, Venuturupalli S, Ishimori M, Wallace D, Weisman MH, Jefferies CA. IL-16/miR-125a axis controls neutrophil recruitment in pristane-induced lung inflammation. JCI Insight 2018; 3:120798. [PMID: 30089723 DOI: 10.1172/jci.insight.120798] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/26/2018] [Indexed: 12/20/2022] Open
Abstract
Severe lung inflammation and alveolar hemorrhage can be life-threatening in systemic lupus erythematosus (SLE) patients if not treated early and aggressively. Neutrophil influx is the driver key of this pathology, but little is known regarding the molecular events regulating this recruitment. Here, we uncover a role for IL-16/mir-125a in this pathology and show not only that IL-16 is a target for miR-125a but that reduced miR-125a expression in SLE patients associates with lung involvement. Furthermore, in the pristane model of acute "SLE-like" lung inflammation and alveolar hemorrhage, we observed reduced pulmonary miR-125a and enhanced IL-16 expression. Neutrophil infiltration was markedly reduced in the peritoneal lavage of pristane-treated IL-16-deficient mice and elevated following i.n. delivery of IL-16. Moreover, a miR-125a mimic reduced pristane-induced IL-16 expression and neutrophil recruitment and rescued lung pathology. Mechanistically, IL-16 acts directly on the pulmonary epithelium and markedly enhances neutrophil chemoattractant expression both in vitro and in vivo, while the miR-125a mimic can prevent this. Our results reveal a role for miR-125a/IL-16 in regulating lung inflammation and suggest this axis may be a therapeutic target for management of acute lung injury in SLE.
Collapse
Affiliation(s)
- Siobhan Smith
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Pei Wen Wu
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jane J Seo
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Thilini Fernando
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Mengyao Jin
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jorge Contreras
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Erica N Montano
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Joan Ní Gabhann
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kyle Cunningham
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Amro Widaa
- Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Eamonn S Molloy
- Department of Rheumatology, St. Vincent's University Hospital, Dublin, Ireland
| | - Grainne Kearns
- Department of Rheumatology, Beaumont Hospital, Dublin, Ireland
| | - Conor C Murphy
- Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Weiping Kong
- China-Japan Friendship Hospital, Chaoyang District, Beijing, China
| | - Harry Björkbacka
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Hardy Kornfeld
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Lindsy Forbess
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Swamy Venuturupalli
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Mariko Ishimori
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Daniel Wallace
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Michael H Weisman
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Caroline A Jefferies
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
24
|
Arts RJW, Joosten LAB, Netea MG. The Potential Role of Trained Immunity in Autoimmune and Autoinflammatory Disorders. Front Immunol 2018. [PMID: 29515591 PMCID: PMC5826224 DOI: 10.3389/fimmu.2018.00298] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
During induction of trained immunity, monocytes and macrophages undergo a functional and transcriptional reprogramming toward increased activation. Important rewiring of cellular metabolism of the myeloid cells takes place during induction of trained immunity, including a shift toward glycolysis induced through the mTOR pathway, as well as glutaminolysis and cholesterol synthesis. Subsequently, this leads to modulation of the function of epigenetic enzymes, resulting in important changes in chromatin architecture that enables increased gene transcription. However, in addition to the beneficial effects of trained immunity as a host defense mechanism, we hypothesize that trained immunity also plays a deleterious role in the induction and/or maintenance of autoimmune and autoinflammatory diseases if inappropriately activated.
Collapse
Affiliation(s)
- Rob J W Arts
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| |
Collapse
|
25
|
Liu L, Allman WR, Coleman AS, Takeda K, Lin TL, Akkoyunlu M. Delayed onset of autoreactive antibody production and M2-skewed macrophages contribute to improved survival of TACI deficient MRL-Fas/Lpr mouse. Sci Rep 2018; 8:1308. [PMID: 29358664 PMCID: PMC5778001 DOI: 10.1038/s41598-018-19827-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/09/2018] [Indexed: 12/24/2022] Open
Abstract
Anti-B cell activating factor belonging to TNF-family (BAFF) antibody therapy is indicated for the treatment of patients with active systemic lupus erythematosus (SLE). We hypothesized that the BAFF receptor, transmembrane activator and calcium-modulator and cyclophilin interactor (TACI) may be responsible for the generation of antibody secreting plasma cells in SLE. To test this hypothesis, we generated TACI deficient MRL-Fas/Lpr (LPR-TACI−/−) mouse. TACI deficiency resulted in improved survival of MRL-Fas/Lpr mice and delayed production of anti-dsDNA and anti-SAM/RNP antibodies. There was also a delay in the onset of proteinuria and the accumulation of IgG and inflammatory macrophages (Mϕs) in the glomeruli of young LPR-TACI−/− mice compared to wild-type mice. Underscoring the role of TACI in influencing Mϕ phenotype, the transfer of Mϕs from 12-week-old LPR-TACI−/− mice to age-matched sick wild-type animals led to a decrease in proteinuria and improvement in kidney pathology. The fact that, in LPR-TACI−/− mouse a more pronounced delay was in IgM and IgG3 autoreactive antibody isotypes and the kinetics of follicular helper T (Tfh) cell-development was comparable between the littermates suggest a role for TACI in T cell-independent autoantibody production in MRL-Fas/Lpr mouse prior to the onset of T cell-dependent antibody production.
Collapse
Affiliation(s)
- Lunhua Liu
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Silver Spring, MD, 20993, United States of America
| | - Windy Rose Allman
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Silver Spring, MD, 20993, United States of America
| | - Adam Steven Coleman
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Silver Spring, MD, 20993, United States of America
| | - Kazuyo Takeda
- Microscopy and Imaging Core Facility, Division of Viral Products, Silver Spring, MD, 20993, United States of America
| | - Tsai-Lien Lin
- Vaccine Evaluation Branch, Division of Biostatistics, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, United States of America
| | - Mustafa Akkoyunlu
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Silver Spring, MD, 20993, United States of America.
| |
Collapse
|
26
|
Upregulation of CD16- monocyte subsets in systemic lupus erythematous patients. Clin Rheumatol 2017; 36:2281-2287. [PMID: 28821990 DOI: 10.1007/s10067-017-3787-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 06/23/2017] [Accepted: 08/09/2017] [Indexed: 01/16/2023]
Abstract
Monocytes are an important component in the innate immune system. However, studies to date have failed to conclude whether their levels are altered in patients with systemic lupus erythematosus (SLE). We applied the cytodiff counting method and comprehensively measured the circulating levels of distinct white blood cell (WBC) subsets, including CD16+, CD16-, and total monocytes, in 61 SLE patients as well as in 203 age-matched healthy controls (HCs). The absolute number of CD16- monocytes, total monocytes, immature granulocytes, mature neutrophils, total neutrophils, and T cell blasts was significantly higher, that of non-cytotoxic T lymphocytes, cytotoxic T + NK lymphocytes, T + NK lymphocytes, total lymphocytes, basophils, and eosinophils significantly lower (all p < 0.05), but that of CD16+ monocytes, B lymphocytes, B cell blasts, non-B and non-T cell blasts, and total blasts was not statistically different in SLE patients, as compared to HC. Specifically, among all subsets examined, the percentage of CD16- monocytes and total monocytes was the only one that could discriminate active SLE from quiescent SLE (p = 0.033 and 0.026, respectively). SLE patients with lupus nephritis were also associated with higher levels of circulating CD16- monocytes and total monocytes, in comparison with that of controls (both p < 0.0001). This study suggests the significance of distinct WBC subsets, particularly the differential regulations of monocyte subsets, in the pathogenesis and development of SLE.
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Recent discoveries relay commensal gut microbiota as a relevant factor in the maintenance of intestinal homeostasis. RECENT FINDINGS Alterations in the composition of the intestinal microbiota have been reported in patients with systemic lupus erythematosus and many other inflammatory and autoimmune conditions. However, the mechanisms by which the intestinal microbiota can influence systemic immunity in these situations remain to be elucidated. The inappropriate immune responses of patients with systemic lupus erythematosus could originate a breakdown of tolerance towards the microbiota, leading to the expansion and/or contraction of specific bacterial groups that may culminate in a dysbiotic state. Conversely, an altered composition of the intestinal microbiome in genetically predisposed individuals could influence systemic immunity by several mechanisms, leading to a breakdown of tolerance to self-antigens. Moreover, humoral immune responses can be affected by specific bacterial groups in these individuals. SUMMARY Recent findings support an important role for the crosstalk between bacteria and immune cells to maintain an intestinal homeostasis crucial to sustain tolerance toward self-antigens and intestinal microbiota.
Collapse
|
28
|
|
29
|
Guo C, Huang Y, Yu J, Liu L, Gong X, Huang M, Jiang C, Liao Y, Huang L, Yang G, Li J. The impacts of single nucleotide polymorphisms in genes of cell cycle and NF-kB pathways on the efficacy and acute toxicities of radiotherapy in patients with nasopharyngeal carcinoma. Oncotarget 2017; 8:25334-25344. [PMID: 28445979 PMCID: PMC5421934 DOI: 10.18632/oncotarget.15835] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/06/2017] [Indexed: 12/12/2022] Open
Abstract
Radiotherapy is one of the primary choices for the treatment of nasopharyngeal carcinoma (NPC) and may result in severe radiotoxicities on normal tissues. Single nucleotide polymorphisms (SNPs) in genes of cell cycle and NF-κB pathways have been linked with the prognoses of various cancers. The aim of this study was to explore whether SNPs of genes involved in cell cycle and NF-κB pathways are associated with responses to radiotherapy in NPC patients. We selected 3 SNPs in cell cycle pathway and 5 SNPs in NF-κB pathway and genotyped them in 154 NPC patients treated with radiotherapy. Multivariate logistic regression was used to determine the association of these 8 SNPs with the responses to radiotherapy. We observed that cyclin-dependent kinase inhibitor gene CDKN2A rs3088440 was significantly related with a poorer treatment efficacy on the primary tumor and cervical lymph node after radiotherapy, and also with a decreased risk of grade 3-4 acute radiation-induced myelosuppression. In some subgroups, cyclin D1 gene CCND1 rs9344 and inhibitor of κB kinase gene IKBKB rs12676482 were related with the grade 3-4 acute radiation-induced myelosuppression, and CCND1 rs9344 was also associated with grade 3-4 acute radiation-induced oral mucositis. The current results reveal that SNPs in genes of cell cycle pathwayand NF-κB pathway have the potential to predict the clinical responses to radiotherapy for NPC patients.
Collapse
Affiliation(s)
- Chengxian Guo
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Yuling Huang
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| | - Jingjing Yu
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Lijuan Liu
- Department of Pharmacy, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| | - Xiaochang Gong
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| | - Min Huang
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| | - Chunling Jiang
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| | - Yulu Liao
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| | - Lihua Huang
- Center for Medical Experiments, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Guoping Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Jingao Li
- Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi 330029, China
| |
Collapse
|
30
|
Laria A, Lurati A, Marrazza M, Mazzocchi D, Re KA, Scarpellini M. The macrophages in rheumatic diseases. J Inflamm Res 2016; 9:1-11. [PMID: 26929657 PMCID: PMC4755472 DOI: 10.2147/jir.s82320] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Macrophages belong to the innate immune system giving us protection against pathogens. However it is known that they are also involved in rheumatic diseases. Activated macrophages have two different phenotypes related to different stimuli: M1 (classically activated) and M2 (alternatively activated). M1 macrophages release high levels of pro-inflammatory cytokines, reactive nitrogen and oxygen intermediates killing microorganisms and tumor cells; while M2 macrophages are involved in resolution of inflammation through phagocytosis of apoptotic neutrophils, reduced production of pro-inflammatory cytokines, and increased synthesis of mediators important in tissue remodeling, angiogenesis, and wound repair. The role of macrophages in the different rheumatic diseases is different according to their M1/M2 macrophages phenotype.
Collapse
|
31
|
Nakazawa D, Shida H, Kusunoki Y, Miyoshi A, Nishio S, Tomaru U, Atsumi T, Ishizu A. The responses of macrophages in interaction with neutrophils that undergo NETosis. J Autoimmun 2016; 67:19-28. [DOI: 10.1016/j.jaut.2015.08.018] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Revised: 08/23/2015] [Accepted: 08/26/2015] [Indexed: 10/23/2022]
|
32
|
Son M, Diamond B. C1q-mediated repression of human monocytes is regulated by leukocyte-associated Ig-like receptor 1 (LAIR-1). Mol Med 2015; 20:559-68. [PMID: 25247291 DOI: 10.2119/molmed.2014.00185] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 09/17/2014] [Indexed: 01/28/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by abnormal function of both the innate and the adaptive immune system, leading to a loss of tolerance to self-antigens. Monocytes are a key component of the innate immune system and are efficient producers of multiple cytokines. In SLE, inappropriate activation of monocytes is thought to contribute to the loss of self-tolerance. In this study, we demonstrate that type 1 interferon (IFN) production by CpG-challenged monocytes can be suppressed by C1q through activating leukocyte-associated Ig-like receptor-1 (LAIR-1), which contains immunoreceptor tyrosine-based inhibition motifs (ITIMs). The phosphorylation of LAIR-1 and the interaction of LAIR-1 with SH2 domain-containing protein tyrosine phosphatase-1 (SHP-1) were enhanced after LAIR-1 engagement by C1q. Moreover, engagement of LAIR-1 by C1q inhibited nuclear translocation of interferon regulatory factor (IRF)-3 and IRF5 in CpG-stimulated monocytes. These data suggest a model in which LAIR-1 engagement by C1q helps maintain monocyte tolerance, specifically with respect to Toll-like receptor-9-mediated monocyte activation.
Collapse
Affiliation(s)
- Myoungsun Son
- Center for Autoimmune and Musculoskeletal Diseases, Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Betty Diamond
- Center for Autoimmune and Musculoskeletal Diseases, Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| |
Collapse
|
33
|
Basophil count, a marker for disease activity in systemic lupus erythematosus. Clin Rheumatol 2014; 34:891-6. [PMID: 25403252 DOI: 10.1007/s10067-014-2822-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 11/04/2014] [Accepted: 11/04/2014] [Indexed: 01/02/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic multisystem autoimmune disease, with frequent flares amid remissions. Basophils contribute to the immunopathogenesis of SLE. This retrospective clinical study evaluated blood basophil count as a potential marker of SLE activity. This study included 213 patients with SLE, 70 with non-SLE chronic kidney disease (CKD), and 100 healthy volunteers. SLE disease activity was scored using the SLE Disease Activity Index (SLEDAI). Baseline and post-immunosuppressant bioparameters were compared in patients with active SLE, with second samples taken at total SLEDAI ≤4. Blood basophil counts and other conventional biomarkers were compared among the groups. Among the 213 SLE patients (192 women, 21 men; mean age 33.0 ± 12.0 years), 149 had active disease. Basophil counts were significantly lower in patients with SLE than in patients with non-SLE CKD and healthy controls (0.009 ± 0.010 vs. 0.025 ± 0.015 vs. 0.022 ± 0.010 × 10(9)/L, p <0.001), and lower in patients with active than inactive SLE (0.008 ± 0.009 vs. 0.014 ± 0.012 × 10(9)/L, p <0.001). Basophil counts in SLE patients were significantly higher after than before immunosuppressive treatment (0.021 ± 0.017 vs. 0.008 ± 0.008 × 10(9)/L, p <0.001) and correlated with total SLEDAI score (r = -0.30, p <0.001). Receiver operator curve analysis showed that basophil counts were similar to conventional markers (leukocytes, platelets, and double-stranded (ds) DNA IgG) in differentiating active from inactive SLE. These findings indicate that blood basophil counts may be a useful biomarker in evaluating SLE activity.
Collapse
|
34
|
Abstract
Innate immune detection and subsequent immune responses rely on the initial recognition of pathogen specific molecular motifs. Foreign nucleic acids are key structures recognised by the immune system, recognition of which occurs mainly through the use of nucleic acid receptors including members of the Toll-like receptors, AIM2-like receptors, RIG-I-like receptors and intracellular DNA receptors. While the immune system is critically important in protecting the host from infection, it is of utmost importance that it is tightly regulated, in order to prevent recognition of self-nucleic acids and the subsequent development of autoimmunity. Defects in the mechanisms regulating such pathways, for example mutations in endonucleases that clear DNA, altered expression of nucleic acid sensors and defects in negative regulators of these signalling pathways involved in RNA/DNA sensing, have all been implicated in promoting the generation of autoimmune responses. This evidence, as reviewed here, suggests that novel therapeutics targeting these sensors and their downstream pathways may be of use in the treatment of patients with autoimmune diseases such as systemic lupus erythematosus, rheumatoid arthritis and primary Sjögren's syndrome.
Collapse
Affiliation(s)
- Siobhán Smith
- Molecular and Cellular Therapeutics and RCSI Research Institute, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - Caroline Jefferies
- Molecular and Cellular Therapeutics and RCSI Research Institute, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland.
| |
Collapse
|
35
|
Zhong H, Bao W, Friedman D, Yazdanbakhsh K. Hemin controls T cell polarization in sickle cell alloimmunization. THE JOURNAL OF IMMUNOLOGY 2014; 193:102-10. [PMID: 24879794 DOI: 10.4049/jimmunol.1400105] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Patients with sickle cell disease (SCD) often require transfusions to treat and prevent worsening anemia and other SCD complications. However, transfusions can trigger alloimmunization against transfused RBCs with serious clinical sequelae. Risk factors for alloimmunization in SCD remain poorly understood. We recently reported altered regulatory T cell (Treg) and Th responses with higher circulating Th1 (IFN-γ(+)) cytokines in chronically transfused SCD patients with alloantibodies as compared with those without alloantibodies. Because monocytes play a critical role in polarization of T cell subsets and participate in clearance of transfused RBCs, we tested the hypothesis that in response to the RBC breakdown product hemin, monocyte control of T cell polarization will differ between alloimmunized and non-alloimmunized SCD patients. Exogenous hemin induced Treg polarization in purified T cell/monocyte cocultures from healthy volunteers through the monocyte anti-inflammatory heme-degrading enzyme heme oxygenase-1. Importantly, hemin primarily through its effect on CD16+ monocytes induced an anti-inflammatory (higher Treg/lower Th1) polarization state in the non-alloimmunized SCD group, whereas it had little effect in the alloimmunized group. Non-alloimmunized SCD CD16+ monocytes expressed higher basal levels of heme oxygenase-1. Furthermore, IL-12, which contributed to a proinflammatory polarization state (low Treg/high Th1) in SCD, was dampened in hemin-treated stimulated monocytes from non-alloimmunized SCD patients, but not in the alloimmunized group. These data suggest that unlike alloimmunized patients, non-alloimmunized SCD CD16+ monocytes in response to transfused RBC breakdown products promote an anti-inflammatory state that is less conducive to alloimmunization.
Collapse
Affiliation(s)
- Hui Zhong
- Laboratory of Complement Biology, New York Blood Center, New York, NY 10065
| | - Weili Bao
- Laboratory of Complement Biology, New York Blood Center, New York, NY 10065
| | - David Friedman
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; and Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Karina Yazdanbakhsh
- Laboratory of Complement Biology, New York Blood Center, New York, NY 10065;
| |
Collapse
|
36
|
Lancemaside A from Codonopsis lanceolata modulates the inflammatory responses mediated by monocytes and macrophages. Mediators Inflamm 2014; 2014:405158. [PMID: 24782593 PMCID: PMC3981472 DOI: 10.1155/2014/405158] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 02/15/2014] [Accepted: 02/17/2014] [Indexed: 12/20/2022] Open
Abstract
In this study, we aimed to examine the cellular and molecular mechanisms of lancemaside A from Codonopsis lanceolata (Campanulaceae) in the inflammatory responses of monocytes (U937 cells) and macrophages (RAW264.7 cells). Lancemaside A significantly suppressed the inflammatory functions of lipopolysaccharide- (LPS-) treated RAW264.7 cells by suppressing the production of nitric oxide (NO), the expression of the NO-producing enzyme inducible NO synthase (iNOS), the upregulation of the costimulatory molecule CD80, and the morphological changes induced by LPS exposure. In addition, lancemaside A diminished the phagocytic activity of RAW264.7 cells and boosted the neutralizing capacity of these cells when treated with the radical generator sodium nitroprusside (SNP). Interestingly, lancemaside A strongly blocked the adhesion activity of RAW264.7 cells to plastic culture plates, inhibited the cell-cell and cell-fibronectin (FN) adhesion of U937 cells that was triggered by treatment with an anti-β1-integrin (CD29) antibody and immobilized FN, respectively. By evaluating the activation of various intracellular signaling pathways and the levels of related nuclear transcription factors, lancemaside A was found to block the activation of inhibitor of κB kinase (IKK) and p65/nuclear factor- (NF-) κB. Taken together, our findings strongly suggest that the anti-inflammatory function of lancemaside A is the result of its strong antioxidative and IKK/NF-κB inhibitory activities.
Collapse
|
37
|
Pereira-Lopes S, Celhar T, Sans-Fons G, Serra M, Fairhurst AM, Lloberas J, Celada A. The exonuclease Trex1 restrains macrophage proinflammatory activation. THE JOURNAL OF IMMUNOLOGY 2013; 191:6128-35. [PMID: 24218451 DOI: 10.4049/jimmunol.1301603] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The three-prime repair exonuclease 1 (TREX1) is the most abundant exonuclease in mammalian cells. Mutations in Trex1 gene are being linked to the development of Aicardi-Goutières syndrome, an inflammatory disease of the brain, and systemic lupus erythematosus. In clinical cases and in a Trex1-deficient murine model, chronic production of type I IFN plays a pathogenic role. In this study, we demonstrate that Trex1(-/-) mice present inflammatory signatures in many different organs, including the brain. Trex1 is highly induced in macrophages in response to proinflammatory stimuli, including TLR7 and TLR9 ligands. Our findings show that, in the absence of Trex1, macrophages displayed an exacerbated proinflammatory response. More specifically, following proinflammatory stimulation, Trex1(-/-) macrophages exhibited an increased TNF-α and IFN-α production, higher levels of CD86, and increased Ag presentation to CD4(+) T cells, as well as an impaired apoptotic T cell clearance. These results evidence an unrevealed function of the Trex1 as a negative regulator of macrophage inflammatory activation and demonstrate that macrophages play a major role in diseases associated with Trex1 mutations, which contributes to the understanding of inflammatory signature in these diseases.
Collapse
Affiliation(s)
- Selma Pereira-Lopes
- Grupo Biología del Macrófago, Departamento de Fisiología e Inmunología, Universitat de Barcelona, 08028 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
38
|
Altered immune regulation in type 1 diabetes. Clin Dev Immunol 2013; 2013:254874. [PMID: 24285974 PMCID: PMC3763577 DOI: 10.1155/2013/254874] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 06/19/2013] [Accepted: 07/04/2013] [Indexed: 02/05/2023]
Abstract
Research in genetics and immunology was going on separate strands for a long time. Type 1 diabetes mellitus might not be characterized with a single pathogenetic factor. It develops when a susceptible individual is exposed to potential triggers in a given sequence and timeframe that eventually disarranges the fine-tuned immune mechanisms that keep autoimmunity under control in health. Genomewide association studies have helped to understand the congenital susceptibility, and hand-in-hand with the immunological research novel paths of immune dysregulation were described in central tolerance, apoptotic pathways, or peripheral tolerance mediated by regulatory T-cells. Epigenetic factors are contributing to the immune dysregulation. The interplay between genetic susceptibility and potential triggers is likely to play a role at a very early age and gradually results in the loss of balanced autotolerance and subsequently in the development of the clinical disease. Genetic susceptibility, the impaired elimination of apoptotic β-cell remnants, altered immune regulatory functions, and environmental factors such as viral infections determine the outcome. Autoreactivity might exist under physiologic conditions and when the integrity of the complex regulatory process is damaged the disease might develop. We summarized the immune regulatory mechanisms that might have a crucial role in disease pathology and development.
Collapse
|