1
|
Aboulaghras S, Bouyahya A, El Kadri K, Khalid A, Abdalla AN, Hassani R, Lee LH, Bakrim S. Protective and stochastic correlation between infectious diseases and autoimmune disorders. Microb Pathog 2024; 196:106919. [PMID: 39245422 DOI: 10.1016/j.micpath.2024.106919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
A priori, early exposure to a wide range of bacteria, viruses, and parasites appears to fortify and regulate the immune system, potentially reducing the risk of autoimmune diseases. However, improving hygiene conditions in numerous societies has led to a reduction in these microbial exposures, which, according to certain theories, could contribute to an increase in autoimmune diseases. Indeed, molecular mimicry is a key factor triggering immune system reactions; while it seeks pathogens, it can bind to self-molecules, leading to autoimmune diseases associated with microbial infections. On the other hand, a hygiene-based approach aimed at reducing the load of infectious agents through better personal hygiene can be beneficial for such pathologies. This review sheds light on how the evolution of the innate immune system, following the evolution of molecular patterns associated with microbes, contributes to our protection but may also trigger autoimmune diseases linked to microbes. Furthermore, it addresses how hygiene conditions shield us against autoimmune diseases related to microbes but may lead to autoimmune pathologies not associated with microbes.
Collapse
Affiliation(s)
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, 10106, Morocco.
| | - Kawtar El Kadri
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, 10106, Morocco.
| | - Asaad Khalid
- Health Research Centre, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia.
| | - Rym Hassani
- Environment and Nature Research Centre, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia; Biology Department, University College AlDarb, Jazan University, Jazan 45142, Saudi Arabia.
| | - Learn-Han Lee
- Microbiome Research Group, Research Centre for Life Science and Healthcare, Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute (CBI), University of Nottingham Ningbo China, 315000, Ningbo, China; Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor, 47500, Malaysia.
| | - Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir, 80000, Morocco.
| |
Collapse
|
2
|
Pan W, Tsokos MG, Scherlinger M, Li W, Tsokos GC. The PP2A regulatory subunit PPP2R2A controls NAD + biosynthesis to regulate T cell subset differentiation in systemic autoimmunity. Cell Rep 2024; 43:114379. [PMID: 38889006 PMCID: PMC11414414 DOI: 10.1016/j.celrep.2024.114379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 04/03/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
The protein phosphatase 2A (PP2A) regulatory subunit PPP2R2A is involved in the regulation of immune response. We report that lupus-prone mice with T cells deficient in PPP2R2A display less autoimmunity and nephritis. PPP2R2A deficiency promotes NAD+ biosynthesis through the nicotinamide riboside (NR)-directed salvage pathway in T cells. NR inhibits murine Th17 and promotes Treg cell differentiation, in vitro, by PΑRylating histone H1.2 and causing its reduced occupancy in the Foxp3 loci and increased occupancy in the Il17a loci, leading to increased Foxp3 and decreased Il17a transcription. NR treatment suppresses disease in MRL.lpr mice and restores NAD+-dependent poly [ADP-ribose] polymerase 1 (PARP1) activity in CD4 T cells from patients with systemic lupus erythematosus (SLE), while reducing interferon (IFN)-γ and interleukin (IL)-17 production. We conclude that PPP2R2A controls the level of NAD+ through the NR-directed salvage pathway and promotes systemic autoimmunity. Translationally, NR suppresses lupus nephritis in mice and limits the production of proinflammatory cytokines by SLE T cells.
Collapse
Affiliation(s)
- Wenliang Pan
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Marc Scherlinger
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA; Rheumatology Department, Strasbourg University Hospital of Hautepierre, Strasbourg, France
| | - Wei Li
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Fan KQ, Huang T, Yu JS, Li YY, Jin J. The clinical features and potential mechanisms of cognitive disorders in peripheral autoimmune and inflammatory diseases. FUNDAMENTAL RESEARCH 2024; 4:226-236. [PMID: 38933510 PMCID: PMC11197673 DOI: 10.1016/j.fmre.2022.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 10/15/2022] [Accepted: 12/05/2022] [Indexed: 12/26/2022] Open
Abstract
According to a study from World Health Organization's Global Burden of Disease, mental and neurological disorders have accounted for 13% of global diseases in recent years and are on the rise. Neuropsychiatric conditions or neuroinflammatory disorders are linked by the presence of an exaggerated immune response both peripherally and in the central nervous system (CNS). Cognitive dysfunction (CD) encompasses a complex group of diseases and has frequently been described in the field of autoimmune diseases, especially in multiple non-CNS-related autoimmune diseases. Recent studies have provided various hypotheses regarding the occurrence of cognitive impairment in autoimmune diseases, including that abnormally activated immune cells can disrupt the integrity of the blood-brain barrier (BBB) to trigger a central neuroinflammatory response. When the BBB is intact, autoantibodies and pro-inflammatory molecules in peripheral circulation can enter the brain to activate microglia, inducing CNS inflammation and CD. However, the mechanisms explaining the association between the immune system and neural function and their contribution to diseases are uncertain. In this review, we used clinical statistics to illustrate the correlation between CD and autoimmune diseases that do not directly affect the CNS, summarized the clinical features and mechanisms by which autoimmune diseases trigger cognitive impairment, and explored existing knowledge regarding the link between CD and autoimmune diseases from the perspective of the field of neuroimmunology.
Collapse
Affiliation(s)
- Ke-qi Fan
- MOE Laboratory of Biosystem Homeostasis and Protection, and Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- Department of Gastroenterology, Sir Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou 310016, China
| | - Tao Huang
- MOE Laboratory of Biosystem Homeostasis and Protection, and Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- Department of Gastroenterology, Sir Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou 310016, China
| | - Jian-shuai Yu
- MOE Laboratory of Biosystem Homeostasis and Protection, and Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Yi-yuan Li
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing 210096, China
| | - Jin Jin
- MOE Laboratory of Biosystem Homeostasis and Protection, and Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- Department of Gastroenterology, Sir Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
4
|
Little AJ, Chen PM, Vesely MD, Khan RN, Fiedler J, Garritano J, Maisha FI, McNiff JM, Craft J. HIF-1 regulates pathogenic cytotoxic T cells in lupus skin disease. JCI Insight 2023; 8:e166076. [PMID: 37526979 PMCID: PMC10543720 DOI: 10.1172/jci.insight.166076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 07/11/2023] [Indexed: 08/03/2023] Open
Abstract
Cutaneous lupus erythematosus (CLE) is a disfiguring autoimmune skin disease characterized by an inflammatory infiltrate rich in T cells, which are strongly implicated in tissue damage. How these cells adapt to the skin environment and promote tissue inflammation and damage is not known. In lupus nephritis, we previously identified an inflammatory gene program in kidney-infiltrating T cells that is dependent on HIF-1, a transcription factor critical for the cellular and developmental response to hypoxia as well as inflammation-associated signals. In our present studies using a mouse model of lupus skin disease, we find that skin-infiltrating CD4+ and CD8+ T cells also express high levels of HIF-1. Skin-infiltrating T cells demonstrated a strong cytotoxic signature at the transcript and protein levels, and HIF-1 inhibition abrogated skin and systemic diseases in association with decreased T cell cytotoxic activity. We also demonstrate in human CLE tissue that the T cell-rich inflammatory infiltrate exhibited increased amounts of HIF-1 and a cytotoxic signature. Granzyme B-expressing T cells were concentrated at sites of skin tissue damage in CLE, suggesting relevance of this pathway to human disease.
Collapse
Affiliation(s)
| | - Ping-Min Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Institute of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei City, Taiwan
| | | | | | | | | | | | - Jennifer M. McNiff
- Department of Dermatology and
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Joe Craft
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Internal Medicine (Rheumatology)
| |
Collapse
|
5
|
Yuan S, Zeng Y, Li J, Wang C, Li W, He Z, Ye J, Li F, Chen Y, Lin X, Yu N, Cai X. Phenotypical changes and clinical significance of CD4 +/CD8 + T cells in SLE. Lupus Sci Med 2022; 9:9/1/e000660. [PMID: 35732344 PMCID: PMC9226979 DOI: 10.1136/lupus-2022-000660] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 05/28/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVE T cells display significant phenotypical changes and play multiple roles in promoting the immune response in SLE. The frequencies of T cell subpopulations in SLE are still not well understood. To better understanding the phenotypic abnormalities of T cells in SLE will help us to clarify disease immunopathology and to find promising biomarkers for disease monitoring and control. METHODS Peripheral blood CD4+ and CD8+ T cells and their subsets were determined by flow cytometry. Forty-one active SLE patients were selected, including 28 new-onset patients and 13 relapsing patients. One hundred healthy controls (HCs) were enrolled as the control group. The percentages of these cell subsets between patients with SLE and HCs and their relationships with disease activity and autoantibody titers were analysed. Thirteen of 28 new-onset SLE patients were assessed before and after treatment. The changes in the frequencies of these cell subsets and their relationships with renal response were analysed. RESULTS There was a broad range of anomalies in the proportion of T cell subsets in patients with SLE compared with that of the HCs. Compared with the HCs, a higher frequency of memory T cells and a lower frequency of naïve T cells were noted in patients with SLE. In addition, an imbalance of CD28+ and CD28- cells in CD4+ T cells was observed in patients with SLE. We found that the expanded CD4+CD28- T cells did not decrease after treatment in patients who had impaired renal responses. It was very interesting to exhibit a negative correlation in the frequency between the CD4+CD28- T cells and T regulatory (Treg) cells and a positive correlation between the frequency of CD4+CD28+ T cells and Treg cells in this study. Increased CD8+HLADR+ T cell and CD8+CD38+HLADR+ T cell counts were observed in patients with SLE, suggesting an impaired cytotoxic capacity of CD8+ T cells in SLE. Additionally, we found that CD8+CD38+HLADR+ T cells were closely associated with disease activity, autoantibody titres and renal prognosis. CD4+ CXCR5-PD1+ T cells were expanded in patients with SLE in this study and were associated with disease activity in SLE. Th1 (T helper type 1) cells and Treg cells were decreased, but frequencies of T follicular helper (Tfh) cells, Th2 cells, Th17 cells and Tfh17 cells were increased. A strong correlation between Th17 cells and Tregs with renal involvement was observed in this study. CONCLUSION The proportions of CD4+CD28- T cells, CD4+CXCR5-PD1+ T cells, CD8+HLADR+ T cells and CD8+CD38+HLADR+ T cells increased in patients with SLE and could be associated with disease activity and renal prognosis.
Collapse
Affiliation(s)
- Shiwen Yuan
- Department of Rheumatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Yanting Zeng
- Department of Rheumatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Jiawei Li
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, China
| | - Cuicui Wang
- Department of Rheumatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Weinian Li
- Department of Rheumatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Zhixiang He
- Department of Rheumatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Jinghua Ye
- Department of Rheumatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Fangfei Li
- Department of Rheumatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Yi Chen
- Department of Rheumatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Xiaojun Lin
- Department of Rheumatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Na Yu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoyan Cai
- Department of Rheumatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Singh RP, Hahn BH, Bischoff DS. Identification and Contribution of Inflammation-Induced Novel MicroRNA in the Pathogenesis of Systemic Lupus Erythematosus. Front Immunol 2022; 13:848149. [PMID: 35444657 PMCID: PMC9013931 DOI: 10.3389/fimmu.2022.848149] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/24/2022] [Indexed: 02/06/2023] Open
Abstract
Recently microRNAs (miRNAs) have been recognized as powerful regulators of many genes and pathways involved in the pathogenesis of inflammatory diseases including Systemic Lupus Erythematosus (SLE). SLE is an autoimmune disease characterized by production of various autoantibodies, inflammatory immune cells, and dysregulation of epigenetic changes. Several candidate miRNAs regulating inflammation and autoimmunity in SLE are described. In this study, we found significant increases in the expression of miR21, miR25, and miR186 in peripheral blood mononuclear cells (PBMCs) of SLE patients compared to healthy controls. However, miR146a was significantly decreased in SLE patients compared to healthy controls and was negatively correlated with plasma estradiol levels and with SLE disease activity scores (SLEDAI). We also found that protein levels of IL-12 and IL-21 were significantly increased in SLE patients as compared to healthy controls. Further, our data shows that protein levels of IL-12 were positively correlated with miR21 expression and protein levels of IL-21 positively correlated with miR25 and miR186 expression in SLE patients. In addition, we found that levels of miR21, miR25, and miR186 positively correlated with SLEDAI and miR146a was negatively correlated in SLE patients. Thus, our data shows a dynamic interplay between disease pathogenesis and miRNA expression. This study has translational potential and may identify novel therapeutic targets in patients with SLE.
Collapse
Affiliation(s)
- Ram P Singh
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Division of Rheumatology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Bevra H Hahn
- Division of Rheumatology, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - David S Bischoff
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
7
|
Singh RP, Hahn BH, Bischoff DS. Interferon Genes Are Influenced by 17β-Estradiol in SLE. Front Immunol 2021; 12:725325. [PMID: 34733276 PMCID: PMC8558410 DOI: 10.3389/fimmu.2021.725325] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/20/2021] [Indexed: 12/23/2022] Open
Abstract
Recent evidence suggests the existence of a nexus between inflammatory pathways and the female sex hormone 17β-estradiol, resulting in increased interferon-stimulated genes (ISGs), autoantibodies, and dysregulation of immune cells in SLE. However, the molecular mechanisms and the effect of estradiol on candidate target genes and their pathways remains poorly understood. Our previous work suggests that female SLE patients have increased estradiol levels compared to healthy controls. In the present study, we explored the effects of 17β-estradiol treatment on expression of IFN (interferons)-stimulated genes and pro-inflammatory cytokines/chemokines. We found significantly increased (5-10-fold) expression of IFN-regulated genes in healthy females. Furthermore, we found significantly increased plasma levels of IL-6, IL-12, IL-17, IL-18, stem cell factor (SCF), and IL-21/IL-23 in SLE patients compared to healthy controls, and those levels positively correlated with the plasma levels of 17β-estradiol. In addition, levels of IL-21 positively correlated with the SLE disease activity index (SLEDAI) score of SLE patients. In vitro treatment of PBMCs from either SLE patients or healthy controls with 17β-estradiol at physiological concentration (~50 pg/ml) also significantly increased secretion of many pro-inflammatory cytokines and chemokines (IL-6, IL-12, IL-17, IL-8, IFN-γ; MIP1α, and MIP1β) in both groups. Further our data revealed that 17β-estradiol significantly increased the percentage of CD3+CD69+ and CD3+IFNγ+ T cells; whereas, simultaneous addition of 17β-estradiol and an ERα inhibitor prevented this effect. Collectively, our findings indicate that 17β-estradiol participates in the induction of pro-inflammatory cytokines and chemokines and further influences interferon genes and pathways.
Collapse
Affiliation(s)
- Ram P Singh
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Division of Rheumatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Bevra H Hahn
- Division of Rheumatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - David S Bischoff
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
8
|
Sete MRC, Carlos JC, Mello-Neto JM, Lira-Junior R, Brito F, Bostrom EA, Sztajnbok FR, Figueredo CM. Impact of chronic gingivitis management on the cytokine and anti-PPAD expressions in juvenile systemic lupus erythematosus: A six-month follow-up. J Periodontal Res 2021; 56:1132-1140. [PMID: 34510434 DOI: 10.1111/jre.12924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/26/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To evaluate how chronic gingivitis treatment impacts the oral and circulating cytokine expressions after six-month follow-up in patients with juvenile systemic lupus erythematosus (jSLE) and also to evaluate the circulating expression of anti-Porphyromonas gingivalis peptidylarginine deiminase antibodies (anti-PPAD) before and after treatment. BACKGROUND Juvenile systemic lupus erythematosus patients present a worse periodontal condition associated with higher gingival crevicular fluid (GCF) levels of interleukin (IL)-1β, IL-8, granulocyte colony-stimulating factor (G-CSF), interferon-γ and monocyte chemoattractant protein (MCP)-1. MATERIALS AND METHODS Twenty-one adolescents with jSLE (mean age: 16.2 ± 1.5 years) were recruited. Participants were rheumatologically and periodontally examined. All individuals were clinically diagnosed with gingival inflammation. Chronic gingivitis treatment consisted of supragingival scaling, prophylaxis and oral hygiene instructions. The cytokine levels were determined by bead-based multiplex assays and the anti-PPAD levels by ELISA. Gingival crevicular fluid (GCF) and serum samples were collected at baseline and 6 months after treatment. RESULTS We observed a reduction in attachment loss, SLE Disease Activity Index (SLEDAI), IL-1β, IL-10 and MCP-1 GCF levels, and the IL-4 and IL-5 serum levels 6 months after periodontal treatment. On the contrary, a significant increase in GCF expression of IL-4, IL-12, IL-17, IFN-γ and serum levels of anti-PPAD antibody was observed. CONCLUSION Juvenile systemic lupus erythematosus patients seem to positively benefit from periodontal treatment by a significantly reduced CAL, a GCF reduction of pro-inflammatory cytokines and an increasing of anti-inflammatory ones. However, an increase in the GCF expression of IL-17 and the serum expression of anti-PPAD antibody 6 months after periodontal treatment might negatively affect the treatment outcome of such patients in the long term.
Collapse
Affiliation(s)
- Manuela Rubim C Sete
- Department of Periodontology, Faculty of Odontology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Juliana C Carlos
- Department of Periodontology, Faculty of Odontology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Joao M Mello-Neto
- Faculty of Medicine and Dentistry, Griffith University, Gold Coast, QLD, Australia
| | - Ronaldo Lira-Junior
- Division of Oral Diagnostics and Rehabilitation, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fernanda Brito
- Department of Periodontology, Faculty of Odontology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Elisabeth A Bostrom
- Division of Oral Diagnostics and Rehabilitation, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Flavio R Sztajnbok
- Unit of Rheumatology, Adolescent Health Studies Center (NESA), Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Carlos Marcelo Figueredo
- Department of Periodontology, Faculty of Odontology, Rio de Janeiro State University, Rio de Janeiro, Brazil.,Faculty of Medicine and Dentistry, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
9
|
Pan W, Nagpal K, Suárez-Fueyo A, Ferretti A, Yoshida N, Tsokos MG, Tsokos GC. The Regulatory Subunit PPP2R2A of PP2A Enhances Th1 and Th17 Differentiation through Activation of the GEF-H1/RhoA/ROCK Signaling Pathway. THE JOURNAL OF IMMUNOLOGY 2021; 206:1719-1728. [PMID: 33762326 DOI: 10.4049/jimmunol.2001266] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/04/2021] [Indexed: 01/04/2023]
Abstract
Protein phosphatase 2A (PP2A) composed of a scaffold subunit, a catalytic subunit, and multiple regulatory subunits is a ubiquitously expressed serine/threonine phosphatase. We have previously shown that the PP2A catalytic subunit is increased in T cells from patients with systemic lupus erythematosus and promotes IL-17 production by enhancing the activity of Rho-associated kinase (ROCK) in T cells. However, the molecular mechanism whereby PP2A regulates ROCK activity is unknown. In this study, we show that the PP2A regulatory subunit PPP2R2A is increased in T cells from people with systemic lupus erythematosus and binds to, dephosphorylates, and activates the guanine nucleotide exchange factor GEF-H1 at Ser885, which in turn increases the levels of RhoA-GTP and the activity of ROCK in T cells. Genetic PPP2R2A deficiency in murine T cells reduced Th1 and Th17, but not regulatory T cell differentiation and mice with T cell-specific PPP2R2A deficiency displayed less autoimmunity when immunized with myelin oligodendrocyte glycoprotein peptide. Our studies indicate that PPP2R2A is the regulatory subunit that dictates the PP2A-directed enhanced Th1 and Th17 differentiation, and therefore, it represents a therapeutic target for pathologies linked to Th1 and Th17 cell expansion.
Collapse
Affiliation(s)
- Wenliang Pan
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Kamalpreet Nagpal
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Abel Suárez-Fueyo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Andrew Ferretti
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Nobuya Yoshida
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
10
|
Chen PM, Tsokos GC. T Cell Abnormalities in the Pathogenesis of Systemic Lupus Erythematosus: an Update. Curr Rheumatol Rep 2021; 23:12. [PMID: 33512577 DOI: 10.1007/s11926-020-00978-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2020] [Indexed: 12/01/2022]
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus is a complex disease with broad spectrum of clinical manifestations. In addition to abnormal B cell responsive leading to autoantibody production, various T cells also play different roles in promoting systemic autoimmunity and end organ damage. We aim to provide a review on recent developments in how abnormalities in different T cells subsets contribute to systemic lupus erythematosus pathogenesis and how they inform the consideration of new promising therapeutics. RECENT FINDINGS Distinct subsets of T cells known as T follicular helper cells enable the production of pathogenic autoantibodies. Detailed understanding of the B cell helping T cell subsets should improve the performance of clinical trials targeting the cognate T:B cell interaction. CD8+ T cells play a role in peripheral tolerance and reversal of its exhausted phenotype could potentially alleviate both systemic autoimmunity and the risk of infection. Research on the abnormal lupus T cell signaling also leads to putative therapeutic targets able to restore interleukin-2 production and suppress the production of the pathogenic IL-17 cytokine. Recently, several studies have focused on dissecting T cell populations located in the damaged organs, aiming to target the pathogenic processes specific to each organ. Numerous T cell subsets play distinct roles in SLE pathogenesis and recent research in understanding abnormal signaling pathways, cellular metabolism, and environmental cues pave the way for the development of novel therapeutics.
Collapse
Affiliation(s)
- Ping-Min Chen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Abstract
Renal inflammation, induced by autoantigen recognition or toxic drugs, leads to renal tissue injury and decline in kidney function. Recent studies have demonstrated the crucial role for regulatory T cells in suppressing pathogenic adaptive but also innate immune responses in the inflamed kidney. However, there is also evidence for other immune cell populations with immunosuppressive function in renal inflammation. This review summarizes mechanisms of immune cell regulation in immune-mediated glomerulonephritis and acute and chronic nephrotoxicity.
Collapse
|
12
|
Zhan CS, Chen J, Chen J, Zhang LG, Liu Y, Du HX, Wang H, Zheng MJ, Yu ZQ, Chen XG, Zhang L, Liang CZ. CaMK4-dependent phosphorylation of Akt/mTOR underlies Th17 excessive activation in experimental autoimmune prostatitis. FASEB J 2020; 34:14006-14023. [PMID: 32862457 DOI: 10.1096/fj.201902910rrr] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 07/27/2020] [Accepted: 08/12/2020] [Indexed: 11/11/2022]
Abstract
Chronic prostatitis and chronic pelvic pain syndrome (CP/CPPS) is a complicated syndrome characterized by genitourinary pain in the absence of bacterial infection. Th17 cell-driven autoimmunity has been proposed as a cause of CP/CPPS. However, the factors that promote Th17-driven autoimmunity in experimental autoimmune prostatitis (EAP) and the molecular mechanisms are still largely unknown. Here, we showed that Th17 cells were excessively activated, and blockade of IL-17A could effectively ameliorate various symptoms in EAP. Furthermore, we revealed that calcium/calmodulin-dependent kinase Ⅳ (CaMK4), especially Thr196 p-CaMK4 was increased in the Th17 cells of the EAP group, which were activated by intracellular cytosolic Ca2+ . Pharmacologic and genetic inhibition of CaMK4 decreased the proportion of Th17 cells, and the protein and mRNA level of IL-17A, IL-22, and RORγt. The phosphorylation of CaMK4 was dependent on the increase in intracellular cytosolic Ca2+ concentration in Th17 cells. A mechanistic study demonstrated that inhibition of CaMK4 reduced IL-17A production by decreasing the phosphorylation of Akt-mTOR, which was well accepted to positively regulate Th17 differentiation. Collectively, our results demonstrated that Ca2+ -CaMK4-Akt/mTOR-IL-17A axis inhibition may serve as a promising therapeutic strategy for CP/CPPS.
Collapse
Affiliation(s)
- Chang-Sheng Zhan
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Jia Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
| | - Jing Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
| | - Li-Gang Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
| | - Yi Liu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - He-Xi Du
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
| | - Hui Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
| | - Mei-Juan Zheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zi-Qiang Yu
- Department of Urology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Xian-Guo Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Li Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| | - Chao-Zhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
Elucidating the Pivotal Immunomodulatory and Anti-Inflammatory Potentials of Chloroquine and Hydroxychloroquine. J Immunol Res 2020; 2020:4582612. [PMID: 33062720 PMCID: PMC7533005 DOI: 10.1155/2020/4582612] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/03/2020] [Indexed: 12/30/2022] Open
Abstract
Chloroquine (CQ) and hydroxychloroquine (HCQ) are derivatives of 4-aminoquinoline compounds with over 60 years of safe clinical usage. CQ and HCQ are able to inhibit the production of cytokines such as interleukin- (IL-) 1, IL-2, IL-6, IL-17, and IL-22. Also, CQ and HCQ inhibit the production of interferon- (IFN-) α and IFN-γ and/or tumor necrotizing factor- (TNF-) α. Furthermore, CQ blocks the production of prostaglandins (PGs) in the intact cell by inhibiting substrate accessibility of arachidonic acid necessary for the production of PGs. Moreover, CQ affects the stability between T-helper cell (Th) 1 and Th2 cytokine secretion by augmenting IL-10 production in peripheral blood mononuclear cells (PBMCs). Additionally, CQ is capable of blocking lipopolysaccharide- (LPS-) triggered stimulation of extracellular signal-modulated extracellular signal-regulated kinases 1/2 in human PBMCs. HCQ at clinical levels effectively blocks CpG-triggered class-switched memory B-cells from differentiating into plasmablasts as well as producing IgG. Also, HCQ inhibits cytokine generation from all the B-cell subsets. IgM memory B-cells exhibits the utmost cytokine production. Nevertheless, CQ triggers the production of reactive oxygen species. A rare, but serious, side effect of CQ or HCQ in nondiabetic patients is hypoglycaemia. Thus, in critically ill patients, CQ and HCQ are most likely to deplete all the energy stores of the body leaving the patient very weak and sicker. We advocate that, during clinical usage of CQ and HCQ in critically ill patients, it is very essential to strengthen the CQ or HCQ with glucose infusion. CQ and HCQ are thus potential inhibitors of the COVID-19 cytokine storm.
Collapse
|
14
|
Chetaille Nézondet AL, Poubelle PE, Pelletier M. The evaluation of cytokines to help establish diagnosis and guide treatment of autoinflammatory and autoimmune diseases. J Leukoc Biol 2020; 108:647-657. [PMID: 32040246 DOI: 10.1002/jlb.5mr0120-218rrr] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 12/17/2022] Open
Abstract
Our knowledge of the role of cytokines in pathologic conditions has increased considerably with the emergence of molecular and genetic studies, particularly in the case of autoinflammatory monogenic diseases. Many rare disorders, considered orphan until recently, are directly related to abnormal gene regulation, and the treatment with biologic agents (biologics) targeting cytokine receptors, intracellular signaling or specific cytokines improve the symptoms of an increasing number of chronic inflammatory diseases. As it is currently impossible to systematically conduct genetic studies for all patients with autoinflammatory and autoimmune diseases, the evaluation of cytokines can be seen as a simple, less time consuming, and less expensive alternative. This approach could be especially useful when the diagnosis of syndromes of diseases of unknown etiology remains problematic. The evaluation of cytokines could also help avoid the current trial-and-error approach, which has the disadvantages of exposing patients to ineffective drugs with possible unnecessary side effects and permanent organ damages. In this review, we discuss the various possibilities, as well as the limitations of evaluating the cytokine profiles of patients suffering from autoinflammatory and autoimmune diseases, with methods such as direct detection of cytokines in the plasma/serum or following ex vivo stimulation of PBMCs leading to the production of their cytokine secretome. The patients' secretome, combined with biomarkers ranging from genetic and epigenetic analyses to immunologic biomarkers, may help not only the diagnosis but also guide the choice of biologics for more efficient and rapid treatments.
Collapse
Affiliation(s)
- Anne-Laure Chetaille Nézondet
- Department of Medicine, Faculty of Medicine, Laval University, Québec, Canada.,Reproduction, Mother and Youth Health Axis, CHU de Québec-Université Laval Research Center, Québec, Canada
| | - Patrice E Poubelle
- Department of Medicine, Faculty of Medicine, Laval University, Québec, Canada.,Infectious and Immune Diseases Axis, CHU de Québec-Université Laval Research Center, Québec, Canada
| | - Martin Pelletier
- Infectious and Immune Diseases Axis, CHU de Québec-Université Laval Research Center, Québec, Canada.,Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, Canada.,ARThrite Research Center, Laval University, Québec, Canada
| |
Collapse
|
15
|
Alexander JJ, Jacob A, Chang A, Quigg RJ, Jarvis JN. Double negative T cells, a potential biomarker for systemic lupus erythematosus. PRECISION CLINICAL MEDICINE 2020; 3:34-43. [PMID: 32257532 PMCID: PMC7093895 DOI: 10.1093/pcmedi/pbaa001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/17/2019] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that is a challenge to diagnose and treat. There is an urgent need for biomarkers to help define organ involvement, and more effective therapies. A unique population of T cells, the CD3+CD4−CD8− (DNeg) cells, is significantly increased in lupus patients. Twenty-seven cases (53%) of pediatric SLE patients had elevated DNeg cells in their peripheral blood, which correlated with kidney function (R2 = 0.54). Significant infiltration of DNeg cells was observed in both adult and pediatric lupus kidneys by immunofluorescence. For the first time, this study provides direct evidence that DNeg cells facilitate kidney injury in preclinical 8-week-old MRL/lpr lupus mice. In lupus mice, the increase in DNeg cells tracked with worsening disease and correlated with kidney function (R2 = 0.85). Our results show that DNeg cells per se can cause kidney dysfunction, increase in number with increase in disease pathology, and could serve as a potential biomarker.
Collapse
Affiliation(s)
- Jessy J Alexander
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA.,Departments of Medicine, Pediatrics, Jacobs School of Medicine and Biomedical Sciences and Genetics, Genomics, & Bioinformatics Program, University at Buffalo, Buffalo, NY 14203, USA
| | - Alexander Jacob
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA.,Departments of Medicine, Pediatrics, Jacobs School of Medicine and Biomedical Sciences and Genetics, Genomics, & Bioinformatics Program, University at Buffalo, Buffalo, NY 14203, USA
| | - Anthony Chang
- Departments of Medicine, Pediatrics, Jacobs School of Medicine and Biomedical Sciences and Genetics, Genomics, & Bioinformatics Program, University at Buffalo, Buffalo, NY 14203, USA
| | - Richard J Quigg
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA.,Departments of Medicine, Pediatrics, Jacobs School of Medicine and Biomedical Sciences and Genetics, Genomics, & Bioinformatics Program, University at Buffalo, Buffalo, NY 14203, USA
| | - James N Jarvis
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
16
|
Smith EMD, Lythgoe H, Midgley A, Beresford MW, Hedrich CM. Juvenile-onset systemic lupus erythematosus: Update on clinical presentation, pathophysiology and treatment options. Clin Immunol 2019; 209:108274. [PMID: 31678365 DOI: 10.1016/j.clim.2019.108274] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/12/2019] [Accepted: 10/13/2019] [Indexed: 12/25/2022]
Abstract
Juvenile-onset systemic lupus erythematosus (jSLE) accounts for up to 20% of all SLE patients. Key differences between juvenile- and adult-onset (aSLE) disease include higher disease activity, earlier development of damage, and increased use of immunosuppressive treatment in jSLE suggesting (at least partial) infectivity secondary to variable pathomechanisms. While the exact pathophysiology of jSLE remains unclear, genetic factors, immune complex deposition, complement activation, hormonal factors and immune cell dysregulation are involved to variable extents, promising future patient stratification based on immune phenotypes. Though less effective and potentially toxic, jSLE patients are treated based upon evidence from studies in aSLE cohorts. Here, age-specific clinical features of jSLE, underlying pathomechanisms, treatment options and disease outcomes will be addressed. Future directions to improve the care of jSLE patients, including implementation of the Single Hub and Access point for pediatric Rheumatology in Europe (SHARE) recommendations, biomarkers, treat to target and personalized medicine approaches are discussed.
Collapse
Affiliation(s)
- Eve Mary Dorothy Smith
- Department of Women's & Children's Health, Institution of Translational Medicine, University of Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Eaton Rd, Liverpool L12 2AP, UK.
| | - Hanna Lythgoe
- Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Eaton Rd, Liverpool L12 2AP, UK
| | - Angela Midgley
- Department of Women's & Children's Health, Institution of Translational Medicine, University of Liverpool, UK
| | - Michael William Beresford
- Department of Women's & Children's Health, Institution of Translational Medicine, University of Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Eaton Rd, Liverpool L12 2AP, UK
| | - Christian Michael Hedrich
- Department of Women's & Children's Health, Institution of Translational Medicine, University of Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Eaton Rd, Liverpool L12 2AP, UK.
| |
Collapse
|
17
|
Hofmann SR, Mäbert K, Kapplusch F, Russ S, Northey S, Beresford MW, Tsokos GC, Hedrich CM. cAMP Response Element Modulator α Induces Dual Specificity Protein Phosphatase 4 to Promote Effector T Cells in Juvenile-Onset Lupus. THE JOURNAL OF IMMUNOLOGY 2019; 203:2807-2816. [PMID: 31653682 DOI: 10.4049/jimmunol.1900760] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022]
Abstract
Effector CD4+ T cells with increased IL-17A and reduced IL-2 production contribute to tissue inflammation and organ damage in systemic lupus erythematosus (SLE). Increased expression of the transcription factor cAMP response element modulator (CREM) α promotes altered cytokine expression in SLE. The aim of this study was to investigate CREMα-mediated events favoring effector CD4+ T cells in health and disease. Using CRISPR/Cas9 genome editing and lentiviral transduction, we generated CREMα-deficient and CREMα-overexpressing Jurkat T cells. Gene expression and regulatory events were assessed using luciferase reporter assays and chromatin immunoprecipitation. Interaction between CREMα and p300 was investigated using proximity ligation assays, coimmunoprecipitation, and knockdown of p300. Gene expression profiles of modified cells were compared with CD4+ T cells from patients with juvenile-onset SLE. We show that CREMα induces dual specificity protein phosphatase (DUSP) 4 in effector CD4+ T cells through corecruitment of p300. The transcriptional coactivator p300 mediates histone acetylation at DUSP4, prompting increased gene expression. Using DUSP4 transfection models and genetically modified CREM-deficient and CREMα-overexpressing T cells, we demonstrate the molecular underpinnings by which DUSP4 induces IL-17A while limiting IL-2 expression. We demonstrate that CD4+ T cells from patients with juvenile-onset SLE share phenotypical features with CREMα-overexpressing CD4+ T cells, including increased DUSP4 expression and imbalanced IL-17A and IL-2 production. Taken together, we describe CREMα-mediated mechanisms that involve the transcriptional upregulation of DUSP4, leading to imbalanced cytokine production by effector T cells. Our findings identify the CREMα/DUSP4 axis as a promising candidate in the search for biomarkers and therapeutic targets in SLE.
Collapse
Affiliation(s)
- Sigrun R Hofmann
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, TU Dresden, D01307 Dresden, Germany
| | - Katrin Mäbert
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, TU Dresden, D01307 Dresden, Germany
| | - Franz Kapplusch
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool L14 5AB, United Kingdom
| | - Susanne Russ
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, TU Dresden, D01307 Dresden, Germany
| | - Sarah Northey
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool L14 5AB, United Kingdom
| | - Michael W Beresford
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool L14 5AB, United Kingdom.,Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool L14 5AB, United Kingdom.,National Institute for Health Research Alder Hey Clinical Research Facility, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool L14 5AB, United Kingdom; and
| | - George C Tsokos
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Christian M Hedrich
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, TU Dresden, D01307 Dresden, Germany; .,Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool L14 5AB, United Kingdom.,Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool L14 5AB, United Kingdom.,National Institute for Health Research Alder Hey Clinical Research Facility, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool L14 5AB, United Kingdom; and
| |
Collapse
|
18
|
Yan L, Wu P, Gao DM, Hu J, Wang Q, Chen NF, Tong SQ, Rao L, Liu J. The Impact of Vitamin D on Cognitive Dysfunction in Mice with Systemic Lupus Erythematosus. Med Sci Monit 2019; 25:4716-4722. [PMID: 31281179 PMCID: PMC6607939 DOI: 10.12659/msm.915355] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Background A growing body of evidence suggests that systemic lupus erythematosus (SLE) may result in reversible cognitive dysfunction. Vitamin D is considered important for neurons. The therapeutic effect of vitamin D was evaluated in a rat model of SLE. Material/Methods There were 20 male MRL/lpr mice randomly divided into the SLE model group and the vitamin D group, in addition, 10 male C57BL 6J mice were used as the control (CON) group. Vitamin D was administered intraperitoneally (2 μg/kg) for 4 weeks. After 4 weeks of continuing intervention, we tested the cognitive function using the Morris water maze. The expression of vitamin D receptor (VDR), amyloid-β, caspase-3, and Bcl-2 were detected by western blot analysis. Results In the present study, we observed that vitamin D treatment alleviated neurobehavioral deficits in the mice with SLE. At the molecular levels, administration of vitamin D activated the expression of VDR and reduced the number of dead cells in the CA1 region of the hippocampus as well as regulated caspase-3 and Bcl-2 expression. Conclusions In conclusion, our results indicated that vitamin D played a protective role by suppressing inflammatory cytokines, thereby ultimately inhibiting the progression of apoptosis in a mouse model of SLE. Vitamin D may be promising as a protective intervention in SLE with cognitive dysfunction, and more and more experiments are warranted for its clinical testing in the near future.
Collapse
Affiliation(s)
- Lijun Yan
- Department of Rheumatology, Tangshan Gongren Hospita, Tangshan, Hebei, China (mainland)
| | - Ping Wu
- Department of Rheumatology, Tangshan Gongren Hospital, Tangshan, Hebei, China (mainland)
| | - Dong-Mei Gao
- Department of Rheumatology, Tangshan Gongren Hospital, Tangshan, Hebei, China (mainland)
| | - Jie Hu
- Department of Rheumatology, Tangshan Gongren Hospital Group Rehabilitation Hospital, Tangshan, Hebei, China (mainland)
| | - Qian Wang
- Department of Rheumatology, Tangshan Gongren Hospital Group Rehabilitation Hospital, Tangshan, Hebei, China (mainland)
| | - Nan-Fang Chen
- Department of Rheumatology, Tangshan Gongren Hospital Group Rehabilitation Hospital, Tangshan, Hebei, China (mainland)
| | - Sheng-Quan Tong
- Department of Rheumatology, Tangshan Gongren Hospital, Tangshan, Hebei, China (mainland)
| | - Li Rao
- Department of Rheumatology, Tangshan Gongren Hospital, Tangshan, Hebei, China (mainland)
| | - Jing Liu
- Department of Rheumatology, Tangshan Gongren Hospital, Tangshan, Hebei, China (mainland)
| |
Collapse
|
19
|
Yazdani R, Shapoori S, Rezaeepoor M, Sanaei R, Ganjalikhani-Hakemi M, Azizi G, Rae W, Aghamohammadi A, Rezaei N. Features and roles of T helper 9 cells and interleukin 9 in immunological diseases. Allergol Immunopathol (Madr) 2019; 47:90-104. [PMID: 29703631 DOI: 10.1016/j.aller.2018.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 01/28/2018] [Accepted: 02/09/2018] [Indexed: 02/08/2023]
Abstract
T helper 9 (TH9) cells are considered as newly classified helper T cells that have an important role in the regulation of immune responses. Since these cells preferentially produce IL-9, these cells are termed TH9 cells. Recently, the role of TH9 and its signature cytokine (IL-9) has been investigated in a wide range of diseases, including autoimmunity, allergy, infections, cancer and immunodeficiency. Herein, we review the most recent data concerning TH9 cells and IL-9 as well as their roles in disease. These insights suggest that TH9 cells are a future target for therapeutic intervention.
Collapse
Affiliation(s)
- R Yazdani
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - S Shapoori
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - M Rezaeepoor
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - R Sanaei
- Department of Immunology, School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - M Ganjalikhani-Hakemi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - G Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Laboratory Medicine, Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - W Rae
- Department of Immunology, MP8, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton, Hampshire SO16 6YD, UK
| | - A Aghamohammadi
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - N Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Science, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
20
|
La/SSB chimeric autoantibody receptor modified NK92MI cells for targeted therapy of autoimmune disease. Clin Immunol 2018; 192:40-49. [PMID: 29673902 DOI: 10.1016/j.clim.2018.04.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 03/07/2018] [Accepted: 04/09/2018] [Indexed: 11/21/2022]
Abstract
It has been long sought to specifically eliminate B-cell clones that generate autoreactive antibodies, while sparing the immune system when combating autoimmune disease. Although it was impossible to achieve this goal before, newly developed techniques have made it feasible today. Autoantibodies against La/SSB were involved in several autoimmune diseases. Here, we aimed to introduce La/SSB epitope-based chimeric autoantibody receptors (CAAR) into NK92MI cells enabled it to destroy the corresponding La/SSB-specific B cell receptor (BCR) -bearing lymphoma cells (LaA-BCR-Romas, LaA-BCR-Maver-1, and LaA-BCR-Jurkat cells). Such cell lines could eliminate a part of the B-cells in the blood of patients positive for anti-La/SSB antibodies. The CAAR we used in this study was constructed by fusing fragments from the nucleus protein, La/SSB, with the TCR signaling molecules, CD28, CD137, and CD3ζ. Thus, this method could specifically destroy the La/SSB autoreactive B-cell clones. Our results might provide a new strategy to combat antibody-mediated autoimmune diseases.
Collapse
|
21
|
Luo XM, Edwards MR, Mu Q, Yu Y, Vieson MD, Reilly CM, Ahmed SA, Bankole AA. Gut Microbiota in Human Systemic Lupus Erythematosus and a Mouse Model of Lupus. Appl Environ Microbiol 2018; 84:e02288-17. [PMID: 29196292 PMCID: PMC5795066 DOI: 10.1128/aem.02288-17] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 11/24/2017] [Indexed: 01/11/2023] Open
Abstract
Gut microbiota dysbiosis has been observed in a number of autoimmune diseases. However, the role of the gut microbiota in systemic lupus erythematosus (SLE), a prototypical autoimmune disease characterized by persistent inflammation in multiple organs of the body, remains elusive. Here we report the dynamics of the gut microbiota in a murine lupus model, NZB/W F1, as well as intestinal dysbiosis in a small group of SLE patients with active disease. The composition of the gut microbiota changed markedly before and after the onset of lupus disease in NZB/W F1 mice, with greater diversity and increased representation of several bacterial species as lupus progressed from the predisease stage to the diseased stage. However, we did not control for age and the cage effect. Using dexamethasone as an intervention to treat SLE-like signs, we also found that a greater abundance of a group of lactobacilli (for which a species assignment could not be made) in the gut microbiota might be correlated with more severe disease in NZB/W F1 mice. Results of the human study suggest that, compared to control subjects without immune-mediated diseases, SLE patients with active lupus disease possessed an altered gut microbiota that differed in several particular bacterial species (within the genera Odoribacter and Blautia and an unnamed genus in the family Rikenellaceae) and was less diverse, with increased representation of Gram-negative bacteria. The Firmicutes/Bacteroidetes ratios did not differ between the SLE microbiota and the non-SLE microbiota in our human cohort.IMPORTANCE SLE is a complex autoimmune disease with no known cure. Dysbiosis of the gut microbiota has been reported for both mice and humans with SLE. In this emerging field, however, more studies are required to delineate the roles of the gut microbiota in different lupus-prone mouse models and people with diverse manifestations of SLE. Here, we report changes in the gut microbiota in NZB/W F1 lupus-prone mice and a group of SLE patients with active disease.
Collapse
Affiliation(s)
- Xin M Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Michael R Edwards
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Qinghui Mu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Yang Yu
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Miranda D Vieson
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | | | - S Ansar Ahmed
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | | |
Collapse
|
22
|
Manni M, Ricker E, Pernis AB. Regulation of systemic autoimmunity and CD11c + Tbet + B cells by SWEF proteins. Cell Immunol 2017; 321:46-51. [PMID: 28780965 DOI: 10.1016/j.cellimm.2017.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 05/10/2017] [Indexed: 12/18/2022]
Abstract
Recent studies have revealed the existence of a T-bet dependent subset of B cells, which expresses unique phenotypic and functional characteristics including high levels of CD11c and CD11b. In the murine system this B cell subset has been termed Age/autoimmune-associated B cells (ABCs) since it expands with age in non-autoimmune mice and it prematurely accumulates in autoimmune-prone strains. The molecular mechanisms that promote the expansion and function of ABCs are largely unknown. This review will focus on the SWEF proteins, a small family of Rho GEFs comprised of SWAP-70 and its homolog DEF6, a newly identified risk variant for human SLE. We will first provide an overview of the SWEF proteins and then discuss the complex array of biological processes that they control and the autoimmune phenotypes that spontaneously develop in their absence, highlighting the emerging involvement of these proteins in regulating ABCs. A better understanding of the pathways controlled by the SWEF proteins could help provide new insights into the mechanisms responsible for the expansion of ABCs in autoimmunity and potentially guide the design of novel therapeutic approaches.
Collapse
Affiliation(s)
- Michela Manni
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY, USA
| | - Edd Ricker
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY, USA; Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Alessandra B Pernis
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY, USA; Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA; David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA.
| |
Collapse
|
23
|
Rozo C, Chinenov Y, Maharaj RK, Gupta S, Leuenberger L, Kirou KA, Bykerk VP, Goodman SM, Salmon JE, Pernis AB. Targeting the RhoA-ROCK pathway to reverse T-cell dysfunction in SLE. Ann Rheum Dis 2017; 76:740-747. [PMID: 28283529 PMCID: PMC5839171 DOI: 10.1136/annrheumdis-2016-209850] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 08/04/2016] [Accepted: 10/09/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Deregulated production of interleukin (IL)-17 and IL-21 contributes to the pathogenesis of autoimmune disorders such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Production of IL-17 and IL-21 can be regulated by ROCK2, one of the two Rho kinases. Increased ROCK activation was previously observed in an SLE cohort. Here, we evaluated ROCK activity in a new SLE cohort, and an RA cohort, and assessed the ability of distinct inhibitors of the ROCK pathway to suppress production of IL-17 and IL-21 by SLE T cells or human Th17 cells. METHODS ROCK activity in peripheral blood mononuclear cells (PBMCs) from 29 patients with SLE, 31 patients with RA and 28 healthy controls was determined by ELISA. SLE T cells or in vitro-differentiated Th17 cells were treated with Y27632 (a pan-ROCK inhibitor), KD025 (a selective ROCK2 inhibitor) or simvastatin (which inhibits RhoA, a major ROCK activator). ROCK activity and IL-17 and IL-21 production were assessed. The transcriptional profile altered by ROCK inhibitors was evaluated by NanoString technology. RESULTS ROCK activity levels were significantly higher in patients with SLE and RA than healthy controls. Th17 cells exhibited high ROCK activity that was inhibited by Y27632, KD025 or simvastatin; each also decreased IL-17 and IL-21 production by purified SLE T cells or Th17 cells. Immune profiling revealed both overlapping and distinct effects of the different ROCK inhibitors. CONCLUSIONS ROCK activity is elevated in PBMCs from patients with SLE and RA. Production of IL-17 and IL-21 by SLE T cells or Th17 cells can furthermore be inhibited by targeting the RhoA-ROCK pathway via both non-selective and selective approaches.
Collapse
Affiliation(s)
- Cristina Rozo
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, NY, NY, USA
| | - Yurii Chinenov
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, NY, NY, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, NY, NY, USA
| | | | - Sanjay Gupta
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, NY, NY, USA
| | - Laura Leuenberger
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, NY, NY, USA
| | - Kyriakos A. Kirou
- Department of Rheumatology, Hospital for Special Surgery, NY, NY, USA
| | - Vivian P. Bykerk
- Department of Rheumatology, Hospital for Special Surgery, NY, NY, USA
- Department of Medicine, Weill Cornell Medical College, Cornell University, NY, NY, USA
| | - Susan M. Goodman
- Department of Rheumatology, Hospital for Special Surgery, NY, NY, USA
- Department of Medicine, Weill Cornell Medical College, Cornell University, NY, NY, USA
| | - Jane E. Salmon
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, NY, NY, USA
- Department of Rheumatology, Hospital for Special Surgery, NY, NY, USA
- Department of Medicine, Weill Cornell Medical College, Cornell University, NY, NY, USA
| | - Alessandra B. Pernis
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, NY, NY, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, NY, NY, USA
- Department of Medicine, Weill Cornell Medical College, Cornell University, NY, NY, USA
| |
Collapse
|
24
|
Balomenos D, Shokri R, Daszkiewicz L, Vázquez-Mateo C, Martínez-A C. On How Fas Apoptosis-Independent Pathways Drive T Cell Hyperproliferation and Lymphadenopathy in lpr Mice. Front Immunol 2017; 8:237. [PMID: 28344578 PMCID: PMC5344898 DOI: 10.3389/fimmu.2017.00237] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 02/20/2017] [Indexed: 01/20/2023] Open
Abstract
Fas induces massive apoptosis in T cells after repeated in vitro T cell receptor (TCR) stimulation and is critical for lymphocyte homeostasis in Fas-deficient (lpr) mice. Although the in vitro Fas apoptotic mechanism has been defined, there is a large conceptual gap between this in vitro phenomenon and the pathway that leads to in vivo development of lymphadenopathy and autoimmunity. A striking abnormality in lpr mice is the excessive proliferation of CD4+ and CD8+ T cells, and more so of the double-negative TCR+CD4−CD8−B220+ T cells. The basis of lpr T cell hyperproliferation remains elusive, as it cannot be explained by Fas-deficient apoptosis. T cell-directed p21 overexpression reduces hyperactivation/hyperproliferation of all lpr T cell subtypes and lymphadenopathy in lpr mice. p21 controls expansion of repeatedly stimulated T cells without affecting apoptosis. These results confirm a direct link between hyperactivation/hyperproliferation, autoreactivity, and lymphadenopathy in lpr mice and, with earlier studies, suggest that Fas apoptosis-independent pathways control lpr T cell hyperproliferation. lpr T cell hyperproliferation could be an indirect result of the defective apoptosis of repeatedly stimulated lpr T cells. Nonetheless, in this perspective, we argue for an alternative setting, in which lack of Fas would directly cause lpr T cell hyperactivation/hyperproliferation in vivo. We propose that Fas/Fas ligand (FasL) acts as an activation inhibitor of recurrently stimulated T cells, and that its disruption causes overexpansion of T cells in lpr mice. Research to define the underlying mechanism of this Fas/FasL effect could resolve the phenotype of lpr mice and lead to therapeutics for related human syndromes.
Collapse
Affiliation(s)
- Dimitrios Balomenos
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Rahman Shokri
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Lidia Daszkiewicz
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Cristina Vázquez-Mateo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Carlos Martínez-A
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| |
Collapse
|
25
|
Miossec P. Update on interleukin-17: a role in the pathogenesis of inflammatory arthritis and implication for clinical practice. RMD Open 2017; 3:e000284. [PMID: 28243466 PMCID: PMC5318575 DOI: 10.1136/rmdopen-2016-000284] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/16/2017] [Accepted: 01/17/2017] [Indexed: 01/05/2023] Open
Abstract
Interleukin-17 (IL-17A) is a cytokine critical for the acute defence against extracellular bacterial and fungal infections. Excess production during chronic inflammation has been associated with many inflammatory and autoimmune disorders. The present review describes the key molecules of the IL-17 pathway, which are or could be targeted for treatment. Since targeting of IL-17A may affect defence mechanisms, the pathogenesis of such possible adverse events is analysed. Then the contributions of IL-17 to bone changes in various forms of arthritis are discussed. Finally, the results of current inhibitors of the IL-17 pathway in clinical trials are detailed. IL-17A inhibition has been first registered for the treatment of psoriasis, psoriatic arthritis and ankylosing spondylitis. Other therapeutic options are now tested in a long list of diseases.
Collapse
Affiliation(s)
- Pierre Miossec
- Immunogenomics and Inflammation Research Unit EA 4130, Department of Immunology and Rheumatology , University of Lyon , Lyon , France
| |
Collapse
|
26
|
Yoshida N, Comte D, Mizui M, Otomo K, Rosetti F, Mayadas TN, Crispín JC, Bradley SJ, Koga T, Kono M, Karampetsou MP, Kyttaris VC, Tenbrock K, Tsokos GC. ICER is requisite for Th17 differentiation. Nat Commun 2016; 7:12993. [PMID: 27680869 PMCID: PMC5056420 DOI: 10.1038/ncomms12993] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 08/24/2016] [Indexed: 12/25/2022] Open
Abstract
Inducible cAMP early repressor (ICER) has been described as a transcriptional repressor isoform of the cAMP response element modulator (CREM). Here we report that ICER is predominantly expressed in Th17 cells through the IL-6–STAT3 pathway and binds to the Il17a promoter, where it facilitates the accumulation of the canonical enhancer RORγt. In vitro differentiation from naive ICER/CREM-deficient CD4+ T cells to Th17 cells is impaired but can be rescued by forced overexpression of ICER. Consistent with a role of Th17 cells in autoimmune and inflammatory diseases, ICER/CREM-deficient B6.lpr mice are protected from developing autoimmunity. Similarly, both anti-glomerular basement membrane-induced glomerulonephritis and experimental encephalomyelitis are attenuated in ICER/CREM-deficient mice compared with their ICER/CREM-sufficient littermates. Importantly, we find ICER overexpressed in CD4+ T cells from patients with systemic lupus erythematosus. Collectively, our findings identify a unique role for ICER, which affects both organ-specific and systemic autoimmunity in a Th17-dependent manner. ICER is a CREM splice variant that represses CREM/CREB signalling. Here the authors use human cells and mouse models of various autoimmune diseases to show that ICER is central to pathogenic Th17 cell differentiation in autoimmunity.
Collapse
Affiliation(s)
- Nobuya Yoshida
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Denis Comte
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA.,Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland
| | - Masayuki Mizui
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Kotaro Otomo
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Florencia Rosetti
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Tanya N Mayadas
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - José C Crispín
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Sean J Bradley
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Tomohiro Koga
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Michihito Kono
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Maria P Karampetsou
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Vasileios C Kyttaris
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Klaus Tenbrock
- Department of Pediatrics, Division of Allergology and Immunology, RWTH University of Aachen, 52056 Aachen, Germany
| | - George C Tsokos
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
27
|
Vitamin D supplementation effects on FoxP3 expression in T cells and FoxP3+/IL-17A ratio and clinical course in systemic lupus erythematosus patients: a study in a Portuguese cohort. Immunol Res 2016; 65:197-206. [DOI: 10.1007/s12026-016-8829-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
28
|
Yuan L, Xiao ZT, Huang XZ, Wu MJ, Shi H, Liu AF. Human embryonic mesenchymal stem cells alleviate pathologic changes of MRL/Lpr mice by regulating Th7 cell differentiation. Ren Fail 2016; 38:1432-1440. [PMID: 27416851 DOI: 10.3109/0886022x.2015.1136894] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Recent evidence indicates that mesenchymal stem cells (MSC) derived from early embryonic tissues have better therapeutic ability as compared with adult tissue-derived stem cells. In the present study, we transplanted human early embryonic MSC (hMSC) into MRL/Lpr mice via tail vein injection to observe the therapeutic efficacy of hMSC and their impact on T helper 17 (Th17) cell differentiation in MRL/Lpr mice. Animals in hMSC treatment group received hMSC (1 × 106/200 μL) via the tail vein at the age of 16 and 19 weeks. We found that hMSC treatment prolonged the survival of MRL/Lpr mice without inducing tumorigenesis, reduced urine protein, and alleviated the renal pathologic changes. In addition, it reduced the proportion of Th17 cells in the spleen of MRL/Lpr mice and the serum interleukin 17 (IL-17) concentration. Our in vitro experiment also demonstrated that hMSC could secrete Th17 differentiation-related cytokines of PGE2, IL-10 and TGF-β, and IFN-γ stimulation up-regulated the secretion of these immune regulating factors. The results of the present study suggest that hMSC therapy could alleviate systemic and local renal lesions in MRL/Lpr mice, probably by secreting immune regulating factors and regulating Th17 cell differentiation in MRL/Lpr mice.
Collapse
Affiliation(s)
- Li Yuan
- a Division of Nephrology , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| | - Zhuo-Tao Xiao
- a Division of Nephrology , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| | - Xin-Zhong Huang
- a Division of Nephrology , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| | - Min-Juan Wu
- b Research Center of Developmental Biology and Department of Histology and Embryology , Second Military Medical University , Shanghai , China
| | - Hui Shi
- a Division of Nephrology , Affiliated Hospital of Nantong University , Nantong , Jiangsu Province , China
| | - Ai-Fen Liu
- c Renal Group, Basic Medical Research Centre, Medical College of Nantong University , Nantong , Jiangsu Province , China
| |
Collapse
|
29
|
Affiliation(s)
- Alessandra B. Pernis
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021; , ,
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065;
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY 10021
| | - Edd Ricker
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021; , ,
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065;
| | - Chien-Huan Weng
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021; , ,
- Graduate Program in Biochemistry Cell and Molecular Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065;
| | - Cristina Rozo
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021; , ,
| | - Woelsung Yi
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY 10021; , ,
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021
| |
Collapse
|
30
|
Araújo JAP, Mesquita D, Cruvinel WDM, Salmazi KI, Kallás EG, Andrade LEC. Linfócitos Th17 e linfócitos T CD4+ multifuncionais em pacientes com lúpus eritematoso sistêmico. REVISTA BRASILEIRA DE REUMATOLOGIA 2016. [DOI: 10.1016/j.rbr.2015.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
31
|
Yin Y, Choi SC, Xu Z, Zeumer L, Kanda N, Croker BP, Morel L. Glucose Oxidation Is Critical for CD4+ T Cell Activation in a Mouse Model of Systemic Lupus Erythematosus. THE JOURNAL OF IMMUNOLOGY 2015; 196:80-90. [PMID: 26608911 DOI: 10.4049/jimmunol.1501537] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/27/2015] [Indexed: 12/26/2022]
Abstract
We have previously shown that CD4(+) T cells from B6.Sle1Sle2.Sle3 lupus mice and patients present a high cellular metabolism, and a treatment combining 2-deoxy-D-glucose, which inhibits glucose metabolism, and metformin, which inhibits oxygen consumption, normalized lupus T cell functions in vitro and reverted disease in mice. We obtained similar results with B6.lpr mice, another model of lupus, and showed that a continuous treatment is required to maintain the beneficial effect of metabolic inhibitors. Further, we investigated the relative roles of glucose oxidation and pyruvate reduction into lactate in this process. Treatments of B6.Sle1Sle2.Sle3 mice with either 2-deoxy-D-glucose or metformin were sufficient to prevent autoimmune activation, whereas their combination was necessary to reverse the process. Treatment of B6.Sle1Sle2.Sle3 mice with dichloroacetate, an inhibitor of lactate production, failed to effectively prevent or reverse autoimmune pathology. In vitro, CD4(+) T cell activation upregulated the expression of genes that favor oxidative phosphorylation. Blocking glucose oxidation inhibited both IFN-γ and IL-17 production, which could not be achieved by blocking pyruvate reduction. Overall, our data show that targeting glucose oxidation is required to prevent or reverse lupus development in mice, which cannot be achieved by simply targeting the pyruvate-lactate conversion.
Collapse
Affiliation(s)
- Yiming Yin
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Seung-Chul Choi
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Zhiwei Xu
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Leilani Zeumer
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Nathalie Kanda
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Byron P Croker
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Laurence Morel
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| |
Collapse
|
32
|
Araújo JAP, Mesquita D, de Melo Cruvinel W, Salmazi KI, Kallás EG, Andrade LEC. Th17 cells and CD4(+) multifunctional T cells in patients with systemic lupus erythematosus. REVISTA BRASILEIRA DE REUMATOLOGIA 2015; 56:28-36. [PMID: 27267331 DOI: 10.1016/j.rbre.2015.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 08/22/2015] [Indexed: 10/22/2022] Open
Abstract
INTRODUCTION/OBJECTIVE Recent evidence suggests that abnormalities involving Th17 lymphocytes are associated with the pathophysiology of systemic lupus erythematosus (SLE). In addition, multifunctional T cells (MFT), i.e., those producing multiple cytokines simultaneously, are present in the inflammatory milieu and may be implicated in the autoimmune process observed in SLE. In the present study, we aimed to characterize the functional status of CD4(+) T cells in SLE by simultaneously determining the concentration of IL-2, IFN-γ and IL-17 in lymphocyte cultures under exogenous and self-antigenic stimuli. PATIENTS AND METHODS Eighteen patients with active disease, 18 with inactive disease, and 14 healthy controls had functional status of CD4(+) T cells analyzed. RESULTS We found that SLE patients presented a decreased number of total CD4(+) cells, an increased number of activated T cells, and an increased frequency of Th17 cells compared to healthy controls (HC). MFT cells had increased frequency in SLE patients and there was an increased frequency of tri-functional MFT in patients with active SLE compared with those with inactive SLE. Interestingly, MTF cells produced larger amounts of IFNγ than mono-functional T cells in patients and controls. CONCLUSION Taken together these data indicate the participation of recently activated Th17 cells and MTF cells in the SLE pathophysiology.
Collapse
Affiliation(s)
| | - Danilo Mesquita
- Department of Rheumatology, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Wilson de Melo Cruvinel
- Department of Rheumatology, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil; Department of Biomedicine, Pontifícia Universidade Católica de Goiás (PUC-GO), Goiânia, GO, Brazil
| | - Karina Inácio Salmazi
- Department of Clinical Immunology and Allergy, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Esper Georges Kallás
- Department of Clinical Immunology and Allergy, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | | |
Collapse
|
33
|
Cao AT, Yao S, Gong B, Nurieva RI, Elson CO, Cong Y. Interleukin (IL)-21 promotes intestinal IgA response to microbiota. Mucosal Immunol 2015; 8:1072-82. [PMID: 25586558 PMCID: PMC4501922 DOI: 10.1038/mi.2014.134] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 12/13/2014] [Indexed: 02/04/2023]
Abstract
Commensal microbiota-specific T helper type 17 (Th17) cells are enriched in the intestines, which can convert into T follicular helper (Tfh) in Peyer's patches, and are crucial for production of intestinal immunoglobulin A (IgA) against microbiota; however, the role of Th17 and Tfh cytokines in regulating the mucosal IgA response to enteric microbiota is still not completely known. In this study, we found that intestinal IgA was impaired in mice deficient in interleukin (IL)-17 or IL-21 signaling. IL-21, but not IL-17, is able to augment B-cell differentiation to IgA(+) cells as mediated by transforming growth factor β1 (TGFβ1) and accelerate IgA class switch recombination (CSR). IL-21 and retinoic acid (RA) induce IgA(+) B-cell development and IgA production and drives autocrine TGFβ1 production to initiate IgA CSR. Repletion of T-cell-deficient TCRβxδ(-/-) mice with Th17 cells specific for commensal bacterial antigen increased the levels of IgA(+) B cells and IgA production in the intestine, which was blocked by neutralizing IL-21. Thus IL-21 functions to strongly augment IgA production under intestinal environment. Furthermore, IL-21 promotes intestinal B-cell homing through α4β7 expression, alone or with TGFβ and RA. Together, IL-21 from microbiota-specific Th17 and/or Tfh cells contributes to robust intestinal IgA levels by enhancing IgA(+) CSR, IgA production and B-cell trafficking into the intestine.
Collapse
Affiliation(s)
- Anthony T. Cao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Suxia Yao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555
| | - Roza I. Nurieva
- Department of Immunology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030
| | - Charles O. Elson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555,Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
34
|
Ryu H, Chung Y. Regulation of IL-17 in atherosclerosis and related autoimmunity. Cytokine 2015; 74:219-27. [DOI: 10.1016/j.cyto.2015.03.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 12/14/2022]
|
35
|
Rodgers DT, McGrath MA, Pineda MA, Al-Riyami L, Rzepecka J, Lumb F, Harnett W, Harnett MM. The parasitic worm product ES-62 targets myeloid differentiation factor 88-dependent effector mechanisms to suppress antinuclear antibody production and proteinuria in MRL/lpr mice. Arthritis Rheumatol 2015; 67:1023-35. [PMID: 25546822 PMCID: PMC4409857 DOI: 10.1002/art.39004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 12/16/2014] [Indexed: 12/30/2022]
Abstract
Objective The hygiene hypothesis suggests that parasitic helminths (worms) protect against the development of autoimmune disease via a serendipitous side effect of worm-derived immunomodulators that concomitantly promote parasite survival and limit host pathology. The aim of this study was to investigate whether ES-62, a phosphorylcholine-containing glycoprotein secreted by the filarial nematode Acanthocheilonema viteae, protects against kidney damage in an MRL/lpr mouse model of systemic lupus erythematosus (SLE). Methods MRL/lpr mice progressively produce high levels of autoantibodies, and the resultant deposition of immune complexes drives kidney pathology. The effects of ES-62 on disease progression were assessed by measurement of proteinuria, assessment of kidney histology, determination of antinuclear antibody (ANA) production and cytokine levels, and flow cytometric analysis of relevant cellular populations. Results ES-62 restored the disrupted balance between effector and regulatory B cells in MRL/lpr mice by inhibiting plasmablast differentiation, with a consequent reduction in ANA production and deposition of immune complexes and C3a in the kidneys. Moreover, by reducing interleukin-22 production, ES-62 may desensitize downstream effector mechanisms in the pathogenesis of kidney disease. Highlighting the therapeutic importance of resetting B cell responses, adoptive transfer of purified splenic B cells from ES-62–treated MRL/lpr mice mimicked the protection afforded by the helminth product. Mechanistically, this reflects down-regulation of myeloid differentiation factor 88 expression by B cells and also kidney cells, resulting in inhibition of pathogenic cross-talk among Toll-like receptor–, C3a-, and immune complex–mediated effector mechanisms. Conclusion This study provides the first demonstration of protection against kidney pathology by a parasitic worm–derived immunomodulator in a model of SLE and suggests therapeutic potential for drugs based on the mechanism of action of ES-62.
Collapse
|
36
|
Biswas PS, Aggarwal R, Levesque MC, Maers K, Ramani K. Type I interferon and T helper 17 cells co-exist and co-regulate disease pathogenesis in lupus patients. Int J Rheum Dis 2015; 18:646-53. [DOI: 10.1111/1756-185x.12636] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Partha S. Biswas
- Division of Rheumatology and Clinical Immunology; Department of Medicine; University of Pittsburgh; Pittsburgh PA USA
| | - Rohit Aggarwal
- Division of Rheumatology and Clinical Immunology; Department of Medicine; University of Pittsburgh; Pittsburgh PA USA
| | - Marc C. Levesque
- Division of Rheumatology and Clinical Immunology; Department of Medicine; University of Pittsburgh; Pittsburgh PA USA
| | - Kelly Maers
- Division of Rheumatology and Clinical Immunology; Department of Medicine; University of Pittsburgh; Pittsburgh PA USA
| | - Kritika Ramani
- Division of Rheumatology and Clinical Immunology; Department of Medicine; University of Pittsburgh; Pittsburgh PA USA
| |
Collapse
|
37
|
de Oliveira Peliçari K, Postal M, Sinicato NA, Peres FA, Fernandes PT, Marini R, Costallat LTL, Appenzeller S. Serum interleukin-17 levels are associated with nephritis in childhood-onset systemic lupus erythematosus. Clinics (Sao Paulo) 2015; 70:313-7. [PMID: 26039945 PMCID: PMC4449477 DOI: 10.6061/clinics/2015(05)01] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/27/2015] [Accepted: 01/27/2015] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES To determine the serum interleukin-17 (IL-17) levels in childhood-onset systemic lupus erythematosus patients and to evaluate the association between IL-17 and clinical manifestations, disease activity, laboratory findings and treatment. METHODS We included 67 consecutive childhood-onset systemic lupus erythematosus patients [61 women; median age 18 years (range 11-31)], 55 first-degree relatives [50 women; median age 40 years (range 29-52)] and 47 age- and sex-matched healthy controls [42 women; median age 19 years (range 6-30)]. The childhood-onset systemic lupus erythematosus patients were assessed for clinical and laboratory systemic lupus erythematosus manifestations, disease activity [Systemic Lupus Erythematosus Disease Activity Index (SLEDAI)], cumulative damage [Systemic Lupus International Collaborating Clinics/American College of Rheumatology (ACR) Damage Index] and current drug use. Serum IL-17 levels were measured by an enzyme-linked immunosorbent assay using commercial kits. RESULTS The median serum IL-17 level was 36.3 (range 17.36-105.92) pg/mL in childhood-onset systemic lupus erythematosus patients and 29.47 (15.16-62.17) pg/mL in healthy controls (p=0.009). We observed an association between serum IL-17 levels and active nephritis (p=0.01) and migraines (p=0.03). Serum IL-17 levels were not associated with disease activity (p=0.32), cumulative damage (p=0.34), or medication use (p=0.63). CONCLUSION IL-17 is increased in childhood-onset systemic lupus erythematosus and may play a role in the pathogenesis of neuropsychiatric and renal manifestations. Longitudinal studies are necessary to determine the role of IL-17 in childhood-onset systemic lupus erythematosus.
Collapse
Affiliation(s)
- Karina de Oliveira Peliçari
- Rheumatology Laboratory, Universidade Estadual de Campinas (UNICAMP), Faculty of Medical Sciences, Campinas/SP, Brazil
| | - Mariana Postal
- Rheumatology Laboratory, Universidade Estadual de Campinas (UNICAMP), Faculty of Medical Sciences, Campinas/SP, Brazil
| | - Nailú Angelica Sinicato
- Rheumatology Laboratory, Universidade Estadual de Campinas (UNICAMP), Faculty of Medical Sciences, Campinas/SP, Brazil
| | - Fernando Augusto Peres
- Rheumatology Laboratory, Universidade Estadual de Campinas (UNICAMP), Faculty of Medical Sciences, Campinas/SP, Brazil
| | - Paula Teixeira Fernandes
- Department of Sport Sciences, Universidade Estadual de Campinas (UNICAMP), Faculty of Physical Education, Campinas/SP, Brazil
| | - Roberto Marini
- Pediatric Rheumatology Unit, Universidade Estadual de Campinas (UNICAMP), Faculty of Medical Science, Department of Pediatrics, Campinas/SP, Brazil
| | - Lilian Tereza Lavras Costallat
- Rheumatology Unit, Universidade Estadual de Campinas (UNICAMP), Faculty of Medical Science Department of Medicine, Campinas/SP, Brazil
| | - Simone Appenzeller
- Rheumatology Laboratory, Universidade Estadual de Campinas (UNICAMP), Faculty of Medical Sciences, Campinas/SP, Brazil
- Rheumatology Unit, Universidade Estadual de Campinas (UNICAMP), Faculty of Medical Science Department of Medicine, Campinas/SP, Brazil
| |
Collapse
|
38
|
Yang J, Li Q, Yang X, Li M. Interleukin-9 Is Associated with Elevated Anti-Double-Stranded DNA Antibodies in Lupus-Prone Mice. Mol Med 2015; 21:364-70. [PMID: 25902303 DOI: 10.2119/molmed.2014.00237] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 04/15/2015] [Indexed: 11/06/2022] Open
Abstract
Interleukin (IL)-9, which is produced mainly by CD4(+) T cells, is implicated in mast cell-related allergic diseases, although its involvement in systemic lupus erythematosus (SLE) pathogenesis remains unclear. Thus, we investigated the presence of IL-9 in lupus-prone MRL/Mp-lpr/lpr (MRL/lpr) mice and examined the role of IL-9 in lupus pathogenesis. Increased levels of IL-9(+) lymphocytes were detected in the spleens and kidneys of MRL/lpr mice and increased IL-9 levels in the spleen correlated with PNA(+) germinal center (GC) cell expansion. The percentage of CD4(+)IL-9(+) (Th9) cells was increased in MRL/lpr mice and serum IL-9 levels were elevated and closely related to the production of antibodies against double-stranded DNA (dsDNA). IL-9 appears to promote B-cell proliferation and immunoglobulin production, which could be blocked by inhibition of signal transducer and activator of transcription 3 (STAT3). Treatment with neutralizing anti-IL-9 antibody in vivo decreased serum anti-dsDNA-antibody titers and alleviated lupus nephritis in MRL/lpr mice. Our findings indicate expansion of Th9 cells in lupus-prone MRL/lpr mice and the correlation of IL-9 with B-cell proliferation and autoantibody production. These findings suggest that IL-9 is a potential therapeutic target for SLE.
Collapse
Affiliation(s)
- Ji Yang
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiao Li
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xue Yang
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Ming Li
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
39
|
Daszkiewicz L, Vázquez-Mateo C, Rackov G, Ballesteros-Tato A, Weber K, Madrigal-Avilés A, Di Pilato M, Fotedar A, Fotedar R, Flores JM, Esteban M, Martínez-A C, Balomenos D. Distinct p21 requirements for regulating normal and self-reactive T cells through IFN-γ production. Sci Rep 2015; 5:7691. [PMID: 25573673 PMCID: PMC4287747 DOI: 10.1038/srep07691] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/01/2014] [Indexed: 12/30/2022] Open
Abstract
Self/non-self discrimination characterizes immunity and allows responses against pathogens but not self-antigens. Understanding the principles that govern this process is essential for designing autoimmunity treatments. p21 is thought to attenuate autoreactivity by limiting T cell expansion. Here, we provide direct evidence for a p21 role in controlling autoimmune T cell autoreactivity without affecting normal T cell responses. We studied C57BL/6, C57BL/6/lpr and MRL/lpr mice overexpressing p21 in T cells, and showed reduced autoreactivity and lymphadenopathy in C57BL/6/lpr, and reduced mortality in MRL/lpr mice. p21 inhibited effector/memory CD4(+) CD8(+) and CD4(-)CD8(-) lpr T cell accumulation without altering defective lpr apoptosis. This was mediated by a previously non-described p21 function in limiting T cell overactivation and overproduction of IFN-γ, a key lupus cytokine. p21 did not affect normal T cell responses, revealing differential p21 requirements for autoreactive and normal T cell activity regulation. The underlying concept of these findings suggests potential treatments for lupus and autoimmune lymphoproliferative syndrome, without compromising normal immunity.
Collapse
Affiliation(s)
- Lidia Daszkiewicz
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Cristina Vázquez-Mateo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Gorjana Rackov
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - André Ballesteros-Tato
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Kathrin Weber
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Adrián Madrigal-Avilés
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Mauro Di Pilato
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Arun Fotedar
- Cancer Cell Biology Program, Sidney Kimmel Cancer Center, San Diego, CA, USA
| | - Rati Fotedar
- Sanford-Burnham Medical Research Institute, San Diego, CA, USA
| | - Juana M Flores
- Animal Biology Department, School of Veterinary Medicine, Universidad Complutense, Madrid, Spain
| | - Mariano Esteban
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Carlos Martínez-A
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Dimitrios Balomenos
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, UAM Campus de Cantoblanco, E-28049 Madrid, Spain
| |
Collapse
|
40
|
Clarke EV, Weist BM, Walsh CM, Tenner AJ. Complement protein C1q bound to apoptotic cells suppresses human macrophage and dendritic cell-mediated Th17 and Th1 T cell subset proliferation. J Leukoc Biol 2015; 97:147-60. [PMID: 25381385 PMCID: PMC4377823 DOI: 10.1189/jlb.3a0614-278r] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/15/2014] [Accepted: 09/01/2014] [Indexed: 12/17/2022] Open
Abstract
A complete genetic deficiency of the complement protein C1q results in SLE with nearly 100% penetrance in humans, but the molecular mechanisms responsible for this association have not yet been fully determined. C1q opsonizes ACs for enhanced ingestion by phagocytes, such as Mϕ and iDCs, avoiding the extracellular release of inflammatory DAMPs upon loss of the membrane integrity of the dying cell. We previously showed that human monocyte-derived Mϕ and DCs ingesting autologous, C1q-bound LALs (C1q-polarized Mϕ and C1q-polarized DCs), enhance the production of anti-inflammatory cytokines, and reduce proinflammatory cytokines relative to Mϕ or DC ingesting LAL alone. Here, we show that C1q-polarized Mϕ have elevated PD-L1 and PD-L2 and suppressed surface CD40, and C1q-polarized DCs have higher surface PD-L2 and less CD86 relative to Mϕ or DC ingesting LAL alone, respectively. In an MLR, C1q-polarized Mϕ reduced allogeneic and autologous Th17 and Th1 subset proliferation and demonstrated a trend toward increased Treg proliferation relative to Mϕ ingesting LAL alone. Moreover, relative to DC ingesting AC in the absence of C1q, C1q-polarized DCs decreased autologous Th17 and Th1 proliferation. These data demonstrate that a functional consequence of C1q-polarized Mϕ and DC is the regulation of Teff activation, thereby "sculpting" the adaptive immune system to avoid autoimmunity, while clearing dying cells. It is noteworthy that these studies identify novel target pathways for therapeutic intervention in SLE and other autoimmune diseases.
Collapse
Affiliation(s)
- Elizabeth V Clarke
- Department of Molecular Biology and Biochemistry, Institute for Immunology, University of California-Irvine, Irvine, California, USA; and
| | - Brian M Weist
- Department of Molecular & Cell Biology, University of California-Berkeley, Berkeley, California, USA
| | - Craig M Walsh
- Department of Molecular Biology and Biochemistry, Institute for Immunology, University of California-Irvine, Irvine, California, USA; and
| | - Andrea J Tenner
- Department of Molecular Biology and Biochemistry, Institute for Immunology, University of California-Irvine, Irvine, California, USA; and
| |
Collapse
|
41
|
Moutsopoulos NM, Konkel J, Sarmadi M, Eskan MA, Wild T, Dutzan N, Abusleme L, Zenobia C, Hosur KB, Abe T, Uzel G, Chen W, Chavakis T, Holland SM, Hajishengallis G. Defective neutrophil recruitment in leukocyte adhesion deficiency type I disease causes local IL-17-driven inflammatory bone loss. Sci Transl Med 2014; 6:229ra40. [PMID: 24670684 DOI: 10.1126/scitranslmed.3007696] [Citation(s) in RCA: 236] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Leukocyte adhesion deficiency type I (LAD-I), a disease syndrome associated with frequent microbial infections, is caused by mutations on the CD18 subunit of β₂ integrins. LAD-I is invariably associated with severe periodontal bone loss, which historically has been attributed to the lack of neutrophil surveillance of the periodontal infection. We provide an alternative mechanism by showing that the cytokine interleukin-17 (IL-17) plays a major role in the oral pathology of LAD-I. Defective neutrophil recruitment in LAD-I patients or in LFA-1 (CD11a/CD18)-deficient mice--which exhibit the LAD-I periodontal phenotype--was associated with excessive production of predominantly T cell-derived IL-17 in the periodontal tissue, although innate lymphoid cells also contributed to pathological IL-17 elevation in the LFA-1-deficient mice. Local treatment with antibodies to IL-17 or IL-23 in LFA-1-deficient mice not only blocked inflammatory periodontal bone loss but also caused a reduction in the total bacterial burden, suggesting that the IL-17-driven pathogenesis of LAD-I periodontitis leads to dysbiosis. Therefore, our findings support an IL-17-targeted therapy for periodontitis in LAD-I patients.
Collapse
Affiliation(s)
- Niki M Moutsopoulos
- National Institute of Dental and Craniofacial Research, Oral Immunity and Infection Unit, Oral and Pharyngeal Cancer Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Amarilyo G, Lourenço EV, Shi FD, La Cava A. IL-17 promotes murine lupus. THE JOURNAL OF IMMUNOLOGY 2014; 193:540-3. [PMID: 24920843 DOI: 10.4049/jimmunol.1400931] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The proinflammatory activity of IL-17-producing Th17 cells has been associated with the pathogenesis of several autoimmune diseases. In this article, we provide direct evidence for a role of IL-17 in the pathogenesis of systemic lupus erythematosus (SLE). The induction of SLE by pristane in IL-17-sufficient wild-type mice did not occur in IL-17-deficient mice, which were protected from development of lupus autoantibodies and glomerulonephritis. The protection from SLE in IL-17-deficient mice was associated with a reduced frequency of CD3(+)CD4(-)CD8(-) double-negative T cells and an expansion of CD4(+) regulatory T cells, and did not depend on Stat-1 signaling. These data affirm the key role of IL-17 in the pathogenesis of SLE and strengthen the support for IL-17 blockade in the therapy of SLE.
Collapse
Affiliation(s)
- Gil Amarilyo
- Division of Rheumatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095; and
| | - Elaine V Lourenço
- Division of Rheumatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095; and
| | - Fu-Dong Shi
- Division of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013
| | - Antonio La Cava
- Division of Rheumatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095; and
| |
Collapse
|
43
|
Mohammadzadeh A, Pourfathollah AA, Shahrokhi S, Hashemi SM, Moradi SLA, Soleimani M. Immunomodulatory effects of adipose-derived mesenchymal stem cells on the gene expression of major transcription factors of T cell subsets. Int Immunopharmacol 2014; 20:316-21. [DOI: 10.1016/j.intimp.2014.03.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 02/15/2014] [Accepted: 03/05/2014] [Indexed: 01/01/2023]
|
44
|
Koga T, Hedrich CM, Mizui M, Yoshida N, Otomo K, Lieberman LA, Rauen T, Crispín JC, Tsokos GC. CaMK4-dependent activation of AKT/mTOR and CREM-α underlies autoimmunity-associated Th17 imbalance. J Clin Invest 2014; 124:2234-45. [PMID: 24667640 DOI: 10.1172/jci73411] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 01/23/2014] [Indexed: 12/17/2022] Open
Abstract
Tissue inflammation in several autoimmune diseases, including SLE and MS, has been linked to an imbalance of IL-17-producing Th (Th17) cells and Tregs; however, the factors that promote Th17-driven autoimmunity are unclear. Here, we present evidence that the calcium/calmodulin-dependent protein kinase IV (CaMK4) is increased and required during Th17 cell differentiation. Isolation of naive T cells from a murine model of lupus revealed increased levels of CaMK4 following stimulation with Th17-inducing cytokines but not following Treg, Th1, or Th2 induction. Furthermore, naive T cells from mice lacking CaMK4 did not produce IL-17. Genetic or pharmacologic inhibition of CaMK4 decreased the frequency of IL-17-producing T cells and ameliorated EAE and lupus-like disease in murine models. Inhibition of CaMK4 reduced Il17 transcription through decreased activation of the cAMP response element modulator α (CREM-α) and reduced activation of the AKT/mTOR pathway, which is known to enhance Th17 differentiation. Importantly, silencing CaMK4 in T cells from patients with SLE and healthy individuals inhibited Th17 differentiation through reduction of IL17A and IL17F mRNA. Collectively, our results suggest that CaMK4 inhibition has potential as a therapeutic strategy for Th17-driven autoimmune diseases.
Collapse
|
45
|
Figueroa FE, Cuenca Moreno J, La Cava A. Novel approaches to lupus drug discovery using stem cell therapy. Role of mesenchymal-stem-cell-secreted factors. Expert Opin Drug Discov 2014; 9:555-66. [PMID: 24655067 DOI: 10.1517/17460441.2014.897692] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Patients with systemic lupus erythematosus (SLE) are at increased risk for premature death, particularly among young adults, and present dilemmas regarding drug efficacy versus toxicity. Novel therapeutic strategies have included the use of mesenchymal stem cell (MSC) therapies that are promising but still have limitations. In several disease models, it has become apparent that MSCs do not necessarily replace diseased tissues but rather exert complex paracrine effects that are mediated by their extracellular-secreted products. AREAS COVERED In this review, the authors highlight the data on MSC treatment of SLE and related mechanisms of actions. This data includes the recent evidence that MSC-secreted factors such as extracellular microvesicles (MVs) are important mediators of MSC therapy. Among MVs, the authors delineate the role of exosomes as triggers of regenerative effects in target cells, mediated by transfer of proteins, mRNAs or microRNAs. The authors also outline some of the biological and regulatory restraints encountered by MSC therapy, in contrast to the potential advantages of MSC-derived exosomes as new therapeutic tools in SLE. EXPERT OPINION There is concern about reproducible data on the use of MSC therapy in rheumatic diseases and specifically SLE. Although most experts consider MSCs to be safe, there are still worries over donor variability, immune-mediated rejection, culture-induced senescence, loss of functional properties and genetic instability or eventual malignant transformation. MSC-released factors could avoid most limiting factors associated with cell therapy and are therefore expected to provide a new and safe therapeutic option at an affordable cost.
Collapse
Affiliation(s)
- Fernando E Figueroa
- Universidad de los Andes, Centro de Investigaciones Biomédicas, Facultad de Medicina , Santiago de , Chile
| | | | | |
Collapse
|
46
|
de Araújo MF, Filho AFL, da Silva GP, de Melo MLR, Napimoga MH, Rodrigues DBR, Alves PM, de Lima Pereira SA. Evaluation of peri-implant mucosa: clinical, histopathological and immunological aspects. Arch Oral Biol 2014; 59:470-8. [PMID: 24631629 DOI: 10.1016/j.archoralbio.2014.01.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 12/13/2013] [Accepted: 01/28/2014] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The aim was to compare the inflammatory response in peri-implant mucosa between patients with peri-implantitis (PP-group) and patients with healthy peri-implant tissues (HP-group). MATERIALS AND METHODS Two fragments of peri-implant mucosa of 18 patients were collected and serial sections were performed for histological and immunohistochemical analysis. RESULTS When compared with HP-group, PP-group showed higher immunostained cell density for TGF-β, IL-17 and CD31, beyond greater density of red cells, leukocytes, mast cells chymase (MCC) and mast cell tryptase (MCT). HP-group patients showed higher IL-13 expression and increased amount of collagen fibres when compared with PP-group. In PP-group there was significant positive correlation between MCT density and density of blood vessels immunostained, and between MCC density and density of blood vessels immunostained. There was significant negative correlation between the IL-17 density and collagen percentage. CONCLUSIONS This study demonstrated that in patients with peri-implantitis there was higher of TGF-β and IL-17, indicating that these cytokines are directly involved in the inflammatory process. Thus, understanding the influence of cytokines in the peri-implantitis installation, new therapies could be developed in order to inhibit the synthesis of IL-17 and induce synthesis of IL-13 in peri-implant tissue, contributing to increase the longevity of the implant.
Collapse
Affiliation(s)
| | | | | | | | - Marcelo Henrique Napimoga
- Laboratory of Immunology and Molecular Biology, São Leopoldo Mandic Institute and Research Center, Brazil
| | - Denise Bertulucci Rocha Rodrigues
- Laboratory of Biopathology and Molecular Biology, University of Uberaba (UNIUBE), Brazil; Cefores, Federal University of Triangulo Mineiro (UFTM), Brazil
| | | | - Sanivia Aparecida de Lima Pereira
- Laboratory of Biopathology and Molecular Biology, University of Uberaba (UNIUBE), Brazil; Cefores, Federal University of Triangulo Mineiro (UFTM), Brazil.
| |
Collapse
|
47
|
Shin JI. Induction of severe systemic lupus erythematosus by TNF blockade and response to an anti-IL-6 strategy: Role of IL-17? J Allergy Clin Immunol 2013; 132:1257. [DOI: 10.1016/j.jaci.2013.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 08/13/2013] [Indexed: 10/26/2022]
|
48
|
Le Coz C, Joublin A, Pasquali JL, Korganow AS, Dumortier H, Monneaux F. Circulating TFH subset distribution is strongly affected in lupus patients with an active disease. PLoS One 2013; 8:e75319. [PMID: 24069401 PMCID: PMC3777901 DOI: 10.1371/journal.pone.0075319] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 08/13/2013] [Indexed: 02/06/2023] Open
Abstract
Follicular helper T cells (TFH) represent a distinct subset of CD4+ T cells specialized in providing help to B lymphocytes, which may play a central role in autoimmune diseases having a major B cell component such as systemic lupus erythematosus. Recently, TFH subsets that share common phenotypic and functional characteristics with TFH cells from germinal centers, have been described in the peripheral blood from healthy individuals. The aim of this study was to analyze the distribution of such populations in lupus patients. Circulating TFH cell subsets were defined by multicolor flow cytometry as TFH17 (CXCR3-CCR6+), TFH1 (CXCR3 + CCR6-) or TFH2 (CXCR3-CCR6-) cells among CXCR5 + CD45RA-CD4+ T cells in the peripheral blood of 23 SLE patients and 23 sex and age-matched healthy controls. IL-21 receptor expression by B cells was analyzed by flow cytometry and the serum levels of IL-21 and Igs were determined by ELISA tests. We found that the TFH2 cell subset frequency is strongly and significantly increased in lupus patients with an active disease (SLEDAI score>8), while the TFH1 cell subset percentage is greatly decreased. The TFH2 and TFH1 cell subset frequency alteration is associated with the presence of high Ig levels and autoantibodies in patient’s sera. Moreover, the TFH2 cell subset enhancement correlates with an increased frequency of double negative memory B cells (CD27-IgD-CD19+ cells) expressing the IL-21R. Finally, we found that IgE levels in lupus patients’ sera correlate with disease activity and seem to be associated with high TFH2 cell subset frequency. In conclusion, our study describes for the first time the distribution of circulating TFH cell subsets in lupus patients. Interestingly, we found an increased frequency of TFH2 cells, which correlates with disease activity. Our results suggest that this subset might play a key role in lupus pathogenesis.
Collapse
Affiliation(s)
- Carole Le Coz
- CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunopathologie et Chimie Thérapeutique/Laboratory of excellence MEDALIS, Strasbourg, France
| | | | | | | | | | | |
Collapse
|
49
|
Liu CC, Kao AH, Manzi S, Ahearn JM. Biomarkers in systemic lupus erythematosus: challenges and prospects for the future. Ther Adv Musculoskelet Dis 2013; 5:210-33. [PMID: 23904865 DOI: 10.1177/1759720x13485503] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The search for lupus biomarkers to diagnose, monitor, stratify, and predict individual response to therapy is currently more intense than ever before. This effort is essential for several reasons. First, epidemic overdiagnosis and underdiagnosis of lupus, even by certified rheumatologists, leads to errors in therapy with concomitant side effects which may be more serious than the disease itself. Second, identification of lupus flares remains as much an art as it is a science. Third, the capacity to stratify patients so as to predict those who will develop specific patterns of organ involvement is not currently possible but would potentially lead to preventive therapeutic strategies. Fourth, only one new drug for the treatment of lupus has been approved by the US Food and Drug Administration in over 50 years. A major obstacle in this pipeline is the dearth of biomarkers available to prove a patient has responded to an experimental therapeutic intervention. This review will summarize the challenges faced in the discovery and validation of lupus biomarkers, the most promising lupus biomarkers identified to date, and the promise of future directions.
Collapse
Affiliation(s)
- Chau-Ching Liu
- Allegheny Singer Research Institute,Temple University School of Medicine,320 East North Avenue Pittsburgh, PA 15212, USA
| | | | | | | |
Collapse
|
50
|
Interrelationship of dendritic cells, type 1 interferon system, regulatory T cells and toll-like receptors and their role in lichen planus and lupus erythematosus -- a literature review. Arch Oral Biol 2013; 58:1532-40. [PMID: 23845753 DOI: 10.1016/j.archoralbio.2013.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 05/28/2013] [Accepted: 06/17/2013] [Indexed: 01/21/2023]
Abstract
There is evidence that the activation of some receptors of the toll-like family (TLRs) of the innate immune system, and also changes in expression levels of forkhead box p3 (Foxp3) protein, which is found in regulatory T cells (Tregs), could be involved in the development of autoimmunity. We present here a literature review focusing on the interrelationship of dendritic cells, TLRs, Tregs and type 1 interferon in autoimmune diseases, with special interest in lichen planus and lupus erythematosus. Understanding the specific role of each of these factors would help elucidate the obscure aetiology of such diseases and open new perspectives for their management and treatment.
Collapse
|