1
|
Hao J, Jie Y, Lu Z, Ye T, Meng J, Liu C, Yan J, Zheng Y, Dong Z, Gu Z. Temporal changes in the transcriptome profile of Macrobrachium rosenbergii in response to decapod iridescent virus 1 infection. Front Immunol 2025; 16:1575476. [PMID: 40276510 PMCID: PMC12018387 DOI: 10.3389/fimmu.2025.1575476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
The farming of Macrobrachium rosenbergii faces significant challenges due to infections caused by Decapod iridovirus 1 (DIV1). To gain deeper insights into the dynamic immune regulatory processes of M. rosenbergii in response to DIV1 infection, RNA sequencing (RNA-seq) was employed to profile the transcriptome in the hepatopancreas at 24, 48, 72, and 96 hours post-infection (hpi). Time-course analysis revealed 3,339 differentially expressed genes (DEGs), which exhibited distinct expression patterns across various stages of infection. At 24 hpi and 48 hpi, the top 20 enriched pathways included 3 immunity-related pathways (Lysosome, Phagosome, C-type lectin receptor signaling) and 7 metabolism-related pathways at 24 hpi, and 5 metabolism-related pathways at 48 hpi. In contrast, in the later stages of infection (72 hpi), 13 of the top 17 enriched pathways associated with DEGs were metabolism-related, including those involved in antioxidant defense, such as the Peroxisome, Cysteine and methionine metabolism, and Glutathione metabolism. At 96 hpi, pathways related to ECM-receptor interaction, Purine metabolism, and Lysosome were significantly enriched. Among the DEGs, a total of 16 genes were consistently identified across all time points, with 14 of these genes, including alpha-2-macroglobulin-like, alpha-amylase 1-like, putative aldolase class 2 protein PA3430, platelet-derived growth factor subunit B-like, serum amyloid A-5 protein-like, phenoloxidase-activating enzyme-like, pantetheinase-like, and perlucin-like protein, demonstrating sustained upregulation at all time points. In contrast, the gene encoding rhodanese domain-containing protein CG4456-like was consistantly downregulated. Additionally, weighted gene co-expression network analysis (WGCNA) indicated several hub genes that were tightly connected to intercellular communication, such as innexin shaking-B-like and innexin inx3-like, and endochitinase A1-like. The gene expression changes varied over time, exhibiting a dynamic, time-dependent pattern that underscores the complexity of host-pathogen interactions. These results provide new insights into the cellular mechanisms influenced by DIV1 throughout the infection process, offering valuable knowledge for developing virus control strategies in shrimp aquaculture.
Collapse
Affiliation(s)
- Jingwen Hao
- Xianghu Laboratory, Hangzhou, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Yukun Jie
- Xianghu Laboratory, Hangzhou, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Zhibin Lu
- Xianghu Laboratory, Hangzhou, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | | | | | - Cui Liu
- Xianghu Laboratory, Hangzhou, China
| | | | | | - Zaijie Dong
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | | |
Collapse
|
2
|
Van Damme J, Struyf S, Proost P, Opdenakker G, Gouwy M. Functional Interactions Between Recombinant Serum Amyloid A1 (SAA1) and Chemokines in Leukocyte Recruitment. Int J Mol Sci 2025; 26:2258. [PMID: 40076881 PMCID: PMC11900440 DOI: 10.3390/ijms26052258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The acute phase response is a hallmark of all inflammatory reactions and acute phase reactants, such as C-reactive protein (CRP) and serum amyloid A (SAA) proteins, are among the most useful plasma and serum markers of inflammation in clinical medicine. Although it is well established that inflammatory cytokines, mainly interleukin-1 (IL-1), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) induce SAA in the liver, the biological functions of elicited SAA remain an enigma. By the classical multi-step protein purification studies of chemotactic factors present in plasma or serum, we discovered novel chemokines and SAA1 fragments, which are induced during inflammatory reactions. In contrast to earlier literature, pure SAA1 fails to induce chemokines, an ascribed function that most probably originates from contaminating lipopolysaccharide (LPS). However, intact SAA1 and fragments thereof synergize with CXC and CC chemokines to enhance chemotaxis. Natural SAA1 fragments are generated by inflammatory proteinases such as matrix metalloproteinase-9 (MMP-9). They mediate synergy with chemokines by the interaction with cognate G protein-coupled receptors (GPCRs), formyl peptide receptor 2 (FPR2) and (CC and CXC) chemokine receptors. In conclusion, SAA1 enforces the action of many chemokines and assists in local leukocyte recruitment, in particular, when the concentrations of specifically-induced chemokines are still low.
Collapse
Affiliation(s)
| | | | | | | | - Mieke Gouwy
- Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium; (J.V.D.); (S.S.); (P.P.); (G.O.)
| |
Collapse
|
3
|
Celik HA, Gurbuz N, Turantepe E, Secme M, Dodurga Y. Profiling of Toll-like Receptors and Related Signaling Mediators in the Pathogenesis of Morphea. Dermatol Pract Concept 2024; 14:dpc.1404a219. [PMID: 39652944 PMCID: PMC11619943 DOI: 10.5826/dpc.1404a219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2024] [Indexed: 12/12/2024] Open
Abstract
INTRODUCTION Morphea, also known as localized scleroderma, is a rare fibrosing inflammatory disease of unknown pathogenesis. OBJECTIVES Although the genetic basis for morphea is important, reports on the evaluation of Toll-like receptors (TLR) in this disease is quite limited. We aimed to evaluate TLR expression levels and serum IL-6, IL-17A, TGF-β1, FGF, and VEGF levels in patients with morphea and compare these results with healthy controls. METHODS The expression levels of TLRs in the lesional and non-lesional adjacent skin of patients with morphea and in normal skin of healthy controls were evaluated by RT-PCR, whereas serum levels of IL-6, IL-17A, TGF-β1, FGF, and VEGF were evaluated by ELISA. RESULTS Based on our findings, TLR1 gene expression increased 34.3-fold in the lesional skin of patients with morphea. In addition, IL-6, IL-17A, TGF-β, FGF, and VEGF were found to be higher in the blood samples of the patient group than in the healthy group. CONCLUSION TLRs are important parts of the pathogenesis of morphea, and a better understanding of them will lead to more directed, effective treatments. We believe that this study will be important for pioneering TLR-targeted therapeutic approaches in the treatment of morphea in the future.
Collapse
Affiliation(s)
- Hilal Ayvaz Celik
- Department of Dermatology, Faculty of Medicine, Suleyman Demirel University, Isparta, Türkiye
| | - Nilgun Gurbuz
- Department of Medical Biology, Faculty of Medicine, Suleyman Demirel University, Isparta, Türkiye
| | - Ergin Turantepe
- Department of Medical Biology, Faculty of Medicine, Suleyman Demirel University, Isparta, Türkiye
| | - Mücahit Secme
- Department of Medical Biology, Faculty of Medicine, Ordu University, Ordu, Türkiye
| | - Yavuz Dodurga
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Denizli, Türkiye
| |
Collapse
|
4
|
Wang B, Li H, Gill G, Zhang X, Tao G, Liu B, Zhai L, Chen W, Wang H, Gu HM, Qin S, Zhang DW. Hepatic Surf4 Deficiency Impairs Serum Amyloid A1 Secretion and Attenuates Liver Fibrosis in Mice. RESEARCH (WASHINGTON, D.C.) 2024; 7:0435. [PMID: 39105051 PMCID: PMC11298252 DOI: 10.34133/research.0435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024]
Abstract
Liver fibrosis is a severe global health problem. However, no effective antifibrotic drugs have been approved. Surf4 is primarily located in the endoplasmic reticulum (ER) and mediates the transport of secreted proteins from the ER to the Golgi apparatus. Knockout of hepatic Surf4 (Surf4 LKO) in mice impairs very-low-density lipoprotein secretion without causing overt liver damage. Here, we found that collagen levels are significantly reduced in the liver of Surf4 LKO mice compared with control Surf4 flox mice, as demonstrated by proteomics, Western blot, and quantitative reverse transcription polymerase chain reaction. Therefore, this study aims to investigate whether and how hepatic Surf4 affects liver fibrosis. We observed that CCl4-induced liver fibrosis is significantly lower in Surf4 LKO mice than in Surf4 flox mice. Mechanistically, hepatic Surf4 deficiency reduces serum amyloid A1 (SAA1) secretion and hepatic stellate cell (HSC) activation. Surf4 coimmunoprecipitates and colocalizes with SAA1. Lack of hepatic Surf4 significantly reduces SAA1 secretion from hepatocytes, and SAA1 activates cultured human HSCs (LX-2 cells). Conditioned medium (CM) from Surf4-deficient primary hepatocytes activates LX-2 cells to a much lesser extent than CM from Surf4 flox primary hepatocytes, and this reduced effect is restored by the addition of recombinant SAA1 to CM from Surf4-deficient hepatocytes. Knockdown of SAA1 in primary hepatocytes or TLR2 in LX-2 cells significantly reduces LX-2 activation induced by CM from Surf4 flox hepatocytes but not from Surf4 LKO hepatocytes. Furthermore, knockdown of SAA1 significantly ameliorates liver fibrosis in Surf4 flox mice but does not further reduce liver fibrosis in Surf4 LKO mice. We also observe substantial expression of Surf4 and SAA1 in human fibrotic livers. Therefore, hepatic Surf4 facilitates SAA1 secretion, activates HSCs, and aggravates liver fibrosis, suggesting that hepatic Surf4 and SAA1 may serve as treatment targets for liver fibrosis.
Collapse
Affiliation(s)
- Bingxiang Wang
- School of Clinic and Basic Medical Sciences, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan,China
- Institute of Atherosclerosis in Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Huili Li
- School of Clinic and Basic Medical Sciences, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan,China
- Institute of Atherosclerosis in Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Govind Gill
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Xiangyan Zhang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Geru Tao
- School of Clinic and Basic Medical Sciences, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan,China
- Institute of Atherosclerosis in Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Boyan Liu
- School of Clinic and Basic Medical Sciences, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan,China
- Institute of Atherosclerosis in Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Lei Zhai
- School of Clinic and Basic Medical Sciences, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan,China
- Institute of Atherosclerosis in Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Wei Chen
- School of Clinic and Basic Medical Sciences, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan,China
- Institute of Atherosclerosis in Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Hao Wang
- School of Clinic and Basic Medical Sciences, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan,China
- Institute of Atherosclerosis in Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Hong-mei Gu
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shucun Qin
- School of Clinic and Basic Medical Sciences, The Second Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan,China
- Institute of Atherosclerosis in Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Da-wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
5
|
Możdżan M, Węgiel A, Biskup L, Brzezińska O, Makowska J. Anti-Th/To Antibodies in Scleroderma: Good Prognosis or Serious Concern? J Clin Med 2024; 13:3022. [PMID: 38892733 PMCID: PMC11172938 DOI: 10.3390/jcm13113022] [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: 04/15/2024] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Systemic sclerosis (SSc) represents a rare and intricate autoimmune connective tissue disease, the pathophysiology of which has not been fully understood. Its key features include progressive fibrosis of the skin and internal organs, vasculopathy and aberrant immune activation. While various anti-nuclear antibodies can serve as biomarkers for the classification and prognosis of SSc, their direct role in organ dysfunction remains unclear. Anti-Th/To antibodies are present in approximately 5% of SSc patients, and are particularly prevalent among those with the limited subtype of the disease. Although the presence of these autoantibodies is associated with a mild course of the disease, there is a strong connection between them and severe clinical manifestations of SSc, including interstitial lung disease, pulmonary arterial hypertension and gastrointestinal involvement. Also, the additional clinical correlations, particularly with malignancies, need further research. Moreover, the disease's course seems to be influenced by antibodies, specific serum cytokines and TLR signaling pathways. Understanding the relationships between presence of anti-Th/To, its molecular aspects and response to treatment options is crucial for the development of novel, personalized therapeutic techniques and should undergo profound analysis in future studies.
Collapse
Affiliation(s)
- Maria Możdżan
- Department of Rheumatology, Medical University of Lodz, 90-549 Lodz, Poland; (A.W.); (L.B.); (O.B.)
| | | | | | | | - Joanna Makowska
- Department of Rheumatology, Medical University of Lodz, 90-549 Lodz, Poland; (A.W.); (L.B.); (O.B.)
| |
Collapse
|
6
|
Huang X, An X, Gao X, Wang N, Liu J, Zhang Y, Qi G, Zhang C. Serum amyloid A facilitates expansion of CD4 + T cell and CD19 + B cell subsets implicated in the severity of myasthenia gravis patients. J Neurochem 2024; 168:224-237. [PMID: 38214332 DOI: 10.1111/jnc.16047] [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: 11/05/2023] [Revised: 12/12/2023] [Accepted: 12/29/2023] [Indexed: 01/13/2024]
Abstract
Serum amyloid A (SAA) is a clinically useful inflammatory marker involved in the pathogenesis of autoimmune diseases. This study aimed to explore the SAA levels in a cohort of patients with myasthenia gravis (MG) in relation to disease-related clinical parameters and myasthenic crisis (MC) and elucidate the effects of SAA on immune response. A total of 82 MG patients including 50 new-onset MG patients and 32 MC patients were enrolled in this study. Baseline data and laboratory parameters of all enrolled MG patients were routinely recorded through electronic medical systems. SAA levels were measured by enzyme-linked immunosorbent assay (ELISA) kit. CD4+ T and CD19+ B cell subsets were analyzed by flow cytometry. In vitro, human recombinant SAA (Apo-SAA) was applied to stimulate peripheral blood mononuclear cells (PBMCs) from MG patients to observe the effect on T and B cell differentiation. Our results indicated that SAA levels in new-onset MG patients were higher than those in controls and were positively correlated with QMG score, MGFA classification, plasmablast cells, IL-6, and IL-17 levels. Subgroup analysis revealed that SAA levels were increased in generalized MG (GMG) patients than in ocular MG (OMG), as well as elevated in late-onset MG (LOMG) than in early-onset MG (EOMG) and higher in MGFA III/IV compared with MGFA I/II. The ROC curve demonstrated that SAA showed good diagnostic value for MC, especially when combined with NLR. In vitro, Apo-SAA promoted the Th1 cells, Th17 cells, plasmablast cells, and plasma cells differentiation in MG PBMCs. The present findings suggested that SAA was increased in MG patients and promoted expansion of CD4+ T cell and CD19+ B cell subsets, which implicated in the severity of MG patients.
Collapse
Affiliation(s)
- Xiaoyu Huang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xueting An
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xue Gao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Ningning Wang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jia Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yong Zhang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guoyan Qi
- Center of Treatment of Myasthenia Gravis Hebei Province, First Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Chao Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
7
|
Wang Q, Li H, Lu H, Wang S, Li Y, Zhang Z, Han J, Yang Z, Yang Y, Hong Y. SAA1 exacerbates pancreatic β-cell dysfunction through activation of NF-κB signaling in high-fat diet-induced type 2 diabetes mice. Mol Cell Endocrinol 2023; 576:112043. [PMID: 37574124 DOI: 10.1016/j.mce.2023.112043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
Insufficient decompensated insulin secretion and insulin resistance caused by pancreatic β-cell dysfunction are the pathological bases of type 2 diabetes mellitus (T2DM). Glucolipotoxicity in pancreatic β-cells is an important factor leading to their dysfunction, closely related to inflammatory signals, oxidative stress, mitochondrial dysfunction, and endoplasmic reticulum stress (ERs). However, there may be other unproven regulatory mechanisms that govern pancreatic β-cell dysfunction. Therefore, further elucidation of the underlying mechanisms that lead to pancreatic β-cells dysfunction will provide a sufficient theoretical basis for the more effective prevention and treatment of T2DM. As a stress protein with pro-inflammatory properties, Serum Amyloid 1 (SAA1) promotes the progression of metabolic syndrome-related diseases by activating immune cells and damaging endothelial cells. In the development of T2DM, the activation of nuclear factor-kappa B (NF-κB) signaling aggravates pancreatic β-cells dysfunction under the stimulation of free fatty acids (FFAs), inflammatory factors, and chemokines. Moreover, the facilitating effect of SAA1 on the activation of the NF-κB signaling pathway has been demonstrated in other studies. In the present study, we demonstrated that SAA1 inhibits insulin secretion and promotes apoptotic molecular expression in pancreatic cells and islets and that NF-κB signaling inhibitors could reduce this effect of SAA1. SAA1 deficiency improved high-fat diet (HFD)-induced pancreatic β-cell dysfunction and decreased expression of NF-κB signaling molecules. Our findings suggested that HFD-induced SAA1 might exacerbate T2DM by enhancing pancreatic β-cell dysfunction; such a function of SAA1 might depend on NF-κB signaling activation.
Collapse
Affiliation(s)
- Qi Wang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, 550025, PR China
| | - Hong Li
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, 550025, PR China
| | - Henghao Lu
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, 550025, PR China
| | - Shumin Wang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, 550025, PR China
| | - Yuxiu Li
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, 550025, PR China
| | - Zhenfen Zhang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, 550025, PR China
| | - Jing Han
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, 550025, PR China
| | - Zhe Yang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, 550025, PR China
| | - Yanping Yang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, 550025, PR China
| | - Yan Hong
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, 550025, PR China.
| |
Collapse
|
8
|
O'Reilly S. Toll-like receptor triggering in systemic sclerosis: time to target. Rheumatology (Oxford) 2023; 62:SI12-SI19. [PMID: 35863054 DOI: 10.1093/rheumatology/keac421] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/04/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023] Open
Abstract
SSc is an autoimmune disease that has features of vascular abnormalities, inflammation and skin and lung fibrosis. Toll-like receptors (TLRs) are sentinel receptors that serve to recognize pathogens or internal danger signals leading to downstream signalling pathways that ultimately lead to inflammation and modification of adaptive immunity. Inflammation and fibrosis appear intricately connected in this disease and TLR ligation on fibroblasts can directly activate these cells to produce copious amounts of collagen, a hallmark of disease. The presence of damage-associated molecular patterns in association with fibrosis has been highlighted. Given their prominent role in disease, this review discusses the evidence of their expression and role in disease pathogenesis and possible therapeutic intervention to mitigate fibrosis.
Collapse
|
9
|
Aung WW, Hamaguchi Y, Matsushita T. Targeting cytokines and potentiality of
JAK–STAT
inhibition in systemic sclerosis. JOURNAL OF CUTANEOUS IMMUNOLOGY AND ALLERGY 2022. [DOI: 10.1002/cia2.12288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Wah Wah Aung
- Department of Dermatology, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine Kanazawa University Kanazawa Ishikawa Japan
| | - Yasuhito Hamaguchi
- Department of Dermatology, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine Kanazawa University Kanazawa Ishikawa Japan
| | - Takashi Matsushita
- Department of Dermatology, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine Kanazawa University Kanazawa Ishikawa Japan
| |
Collapse
|
10
|
Regulation of Atherosclerosis by Toll-Like Receptor 4 Induced by Serum Amyloid 1: A Systematic In Vitro Study. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4887593. [PMID: 36158875 PMCID: PMC9499805 DOI: 10.1155/2022/4887593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022]
Abstract
The objective of this study was to investigate the effects of serum amyloid 1 (SAA1) on activation of endothelial cells, formation of foam cells, platelet aggregation, and monocyte/platelet adhesion to endothelial cells. The effect of SAA1 on the inflammatory activation of endothelial cells was investigated by detecting the expression of inflammatory factors and adhesion molecules. The role of SAA1 in formation of foam cells was verified by detecting lipid deposition and expression of molecules related to the formation of foam cells. After platelets were stimulated by SAA1, the aggregation rate was evaluated to determine the effect of SAA1 on platelet aggregation. Monocytes/platelets were cocultured with human umbilical vein endothelial cells (HUVECs) pretreated with or without SAA1 to determine whether SAA1 affected monocyte/platelet adhesion to endothelial cells. By inhibiting toll-like receptor 4 (TLR4) function, we further identified the role of TLR4 signaling in SAA1-mediated endothelial inflammatory activation, foam-cell formation, and monocyte/platelet adhesion to HUVECs. SAA1 significantly increased the expression of adhesion molecules and inflammatory factors in HUVECs. Moreover, SAA1 also promoted lipid deposition and the expression of inflammatory factors and low-density lipoprotein receptor-1 (LOX-1) in THP-1-derived macrophages. In addition, SAA1 induced platelet aggregation and enhanced monocyte/platelet adhesion to HUVECs. However, the TLR4 antagonist significantly inhibited SAA1-induced endothelial cell activation, foam-cell formation, and monocyte/platelet adhesion to HUVECs and downregulated the expression of myeloid differentiation factor 88 (MyD88), phosphor-inhibitor of nuclear factor κB kinase subunit α/β (P-IKKα/β), phospho-inhibitor of nuclear factor κB subunit α (P-IKBα), and phosphorylation of nuclear transcription factor-κB p65 (P-p65) in SAA1-induced HUVECs and THP-1 cells. Conclusively, it is speculated that SAA1 promotes atherosclerosis through enhancing endothelial cell activation, platelet aggregation, foam-cell formation, and monocyte/platelet adhesion to endothelial cells. These biological functions of SAA1 may depend on the activation of TLR4-related nuclear factor-kappa B (NF-κB) signaling pathway.
Collapse
|
11
|
Chepy A, Bourel L, Koether V, Launay D, Dubucquoi S, Sobanski V. Can Antinuclear Antibodies Have a Pathogenic Role in Systemic Sclerosis? Front Immunol 2022; 13:930970. [PMID: 35837382 PMCID: PMC9274282 DOI: 10.3389/fimmu.2022.930970] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/26/2022] [Indexed: 12/30/2022] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease characterized by extensive fibrosis of the skin and internal organs, associated with vasculopathy and autoimmune features. Antinuclear antibodies (ANA) are found in almost all SSc patients and constitute strong diagnosis and prognosis biomarkers. However, it remains unclear whether ANA are simple bystanders or if they can have a role in the pathophysiology of the disease. One might think that the nuclear nature of their targets prevents any accessibility to autoantibodies. Nevertheless, recent data suggest that ANA could be pathogenic or at least contribute to the perennation of the disease. We review here first the indirect clues of the contribution of ANA to SSc: they are associated to the disease subtypes, they may precede disease onset, their titer correlates with disease activity and severity, there is an association between molecular subsets, and some patients can respond to B-cell targeting therapy. Then, we describe in a second part the mechanisms of ANA production in SSc from individual genetic background to post-transcriptional modifications of neoantigens. Finally, we elaborate on the potential mechanisms of pathogenicity: ANA could be pathogenic through immune-complex-mediated mechanisms; other processes potentially involve molecular mimicry and ANA penetration into the target cell, with a focus on anti-topoisomerase-I antibodies, which are the most probable candidate to play a role in the pathophysiology of SSc. Finally, we outline some technical and conceptual ways to improve our understanding in this field.
Collapse
Affiliation(s)
- Aurélien Chepy
- Univ. Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), Lille, France
| | - Louisa Bourel
- Univ. Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for Translational Research in Inflammation, Lille, France
| | - Vincent Koether
- Univ. Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), Lille, France
| | - David Launay
- Univ. Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), Lille, France
| | - Sylvain Dubucquoi
- Univ. Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Institut d’Immunologie, Lille, France
| | - Vincent Sobanski
- Univ. Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for Translational Research in Inflammation, Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), Lille, France
- Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
12
|
Duan T, Du Y, Xing C, Wang HY, Wang RF. Toll-Like Receptor Signaling and Its Role in Cell-Mediated Immunity. Front Immunol 2022. [PMID: 35309296 DOI: 10.3389/fimmu.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Innate immunity is the first defense system against invading pathogens. Toll-like receptors (TLRs) are well-defined pattern recognition receptors responsible for pathogen recognition and induction of innate immune responses. Since their discovery, TLRs have revolutionized the field of immunology by filling the gap between the initial recognition of pathogens by innate immune cells and the activation of the adaptive immune response. TLRs critically link innate immunity to adaptive immunity by regulating the activation of antigen-presenting cells and key cytokines. Furthermore, recent studies also have shown that TLR signaling can directly regulate the T cell activation, growth, differentiation, development, and function under diverse physiological conditions. This review provides an overview of TLR signaling pathways and their regulators and discusses how TLR signaling, directly and indirectly, regulates cell-mediated immunity. In addition, we also discuss how TLR signaling is critically important in the host's defense against infectious diseases, autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Tianhao Duan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yang Du
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Changsheng Xing
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Helen Y Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rong-Fu Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
13
|
Duan T, Du Y, Xing C, Wang HY, Wang RF. Toll-Like Receptor Signaling and Its Role in Cell-Mediated Immunity. Front Immunol 2022; 13:812774. [PMID: 35309296 PMCID: PMC8927970 DOI: 10.3389/fimmu.2022.812774] [Citation(s) in RCA: 378] [Impact Index Per Article: 126.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
Innate immunity is the first defense system against invading pathogens. Toll-like receptors (TLRs) are well-defined pattern recognition receptors responsible for pathogen recognition and induction of innate immune responses. Since their discovery, TLRs have revolutionized the field of immunology by filling the gap between the initial recognition of pathogens by innate immune cells and the activation of the adaptive immune response. TLRs critically link innate immunity to adaptive immunity by regulating the activation of antigen-presenting cells and key cytokines. Furthermore, recent studies also have shown that TLR signaling can directly regulate the T cell activation, growth, differentiation, development, and function under diverse physiological conditions. This review provides an overview of TLR signaling pathways and their regulators and discusses how TLR signaling, directly and indirectly, regulates cell-mediated immunity. In addition, we also discuss how TLR signaling is critically important in the host's defense against infectious diseases, autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Tianhao Duan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yang Du
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Changsheng Xing
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Helen Y. Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rong-Fu Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
14
|
Hinchcliff M, Garcia-Milian R, Di Donato S, Dill K, Bundschuh E, Galdo FD. Cellular and Molecular Diversity in Scleroderma. Semin Immunol 2021; 58:101648. [PMID: 35940960 DOI: 10.1016/j.smim.2022.101648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
With the increasing armamentarium of high-throughput tools available at manageable cost, it is attractive and informative to determine the molecular underpinnings of patient heterogeneity in systemic sclerosis (SSc). Given the highly variable clinical outcomes of patients labelled with the same diagnosis, unravelling the cellular and molecular basis of disease heterogeneity will be crucial to predicting disease risk, stratifying management and ultimately informing a patient-centered precision medicine approach. Herein, we summarise the findings of the past several years in the fields of genomics, transcriptomics, and proteomics that contribute to unraveling the cellular and molecular heterogeneity of SSc. Expansion of these findings and their routine integration with quantitative analysis of histopathology and imaging studies into clinical care promise to inform a scientifically driven patient-centred personalized medicine approach to SSc in the near future.
Collapse
Affiliation(s)
- Monique Hinchcliff
- Yale School of Medicine, Department of Internal Medicine, Section of Rheumatology, Allergy & Immunology, USA.
| | | | - Stefano Di Donato
- Raynaud's and Scleroderma Programme, Leeds Institute of Rheumatic and Musculoskeletal Medicine and NIHR Biomedical Research Centre, University of Leeds, UK
| | | | - Elizabeth Bundschuh
- Yale School of Medicine, Department of Internal Medicine, Section of Rheumatology, Allergy & Immunology, USA
| | - Francesco Del Galdo
- Raynaud's and Scleroderma Programme, Leeds Institute of Rheumatic and Musculoskeletal Medicine and NIHR Biomedical Research Centre, University of Leeds, UK.
| |
Collapse
|
15
|
Thoreau B, Chaigne B, Renaud A, Mouthon L. Pathophysiology of systemic sclerosis. Presse Med 2021; 50:104087. [PMID: 34718115 DOI: 10.1016/j.lpm.2021.104087] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 10/20/2021] [Indexed: 10/20/2022] Open
Abstract
Systemic sclerosis (SSc) is a rare connective tissue disease characterized by vascular remodeling, fibroblast activation and extra-cellular matrix production in excess and autoimmunity. Environmental factors including mainly silica and solvents have been assumed to contribute to the development of SSc, together with genetic factors including gene variants implicated in innate immunity such as IRF5 and STAT4, and epigenetic factors including histone post-translational modifications, DNA hypomethylation, and microRNAs or long- non coding RNAs system were reported to participate in immune activation and fibrosis processes in patients with SSc. A number of animal models of SSc have been set up over the years, including genetic and induced SSc models. These models, together with data obtained from human SSc patients, contributed to better understand the mechanisms contributing to vasculopathy and fibrosis. Alongside the pathophysiological process of SSc, several cellular and molecular actors are involved, such as dysregulations in the innate and adaptive immune cells, of the fibroblast, the implication of pro-inflammatory and pro-fibrosing signaling pathways such as the Wnt, TGF-β pathways or other cytokines, with a strong imprint of oxidative stress. The whole lead to the overactivity of the fibroblast with genetic dysregulation, apoptosis defect, hyperproduction of elements of extracellular matrix, and finally the phenomena of vasculopathy and fibrosis. These advances contribute to open new therapeutic areas through the design of biologics and small molecules.
Collapse
Affiliation(s)
- Benjamin Thoreau
- Institut Cochin, Inserm U1016, CNRS UMR 8104,Université de Paris, Paris, France; Service de Médecine Interne, Centre de Référence Maladies Systémiques Autoimmunes Rares d'Ile de France, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Benjamin Chaigne
- Institut Cochin, Inserm U1016, CNRS UMR 8104,Université de Paris, Paris, France; Service de Médecine Interne, Centre de Référence Maladies Systémiques Autoimmunes Rares d'Ile de France, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Arthur Renaud
- Institut Cochin, Inserm U1016, CNRS UMR 8104,Université de Paris, Paris, France
| | - Luc Mouthon
- Institut Cochin, Inserm U1016, CNRS UMR 8104,Université de Paris, Paris, France; Service de Médecine Interne, Centre de Référence Maladies Systémiques Autoimmunes Rares d'Ile de France, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.
| |
Collapse
|
16
|
Akl EM, Halim WAA. Serum level of serum amyloid A1 protein in patients with acne vulgaris. J Cosmet Dermatol 2021; 21:2597-2601. [PMID: 34449969 DOI: 10.1111/jocd.14405] [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: 07/12/2021] [Accepted: 08/12/2021] [Indexed: 11/30/2022]
Abstract
INTRODUCTION The serum amyloid A1 (SAA1) protein is involved in many pathological diseases. The increased secretion of SAA1 can maintain inflammatory conditions. Acne vulgaris (AV) is a chronic inflammatory disease of pilosebaceous glands and may be associated with systemic manifestations. AIM OF THE WORK This study aimed to evaluate the serum level of SAA1 in patients with AV. SUBJECTS AND METHODS 120 participants were included in this study: 60 patients with AV and 60 apparently healthy volunteers as a control group. These participants were subjected to dermatological examination and assessments of lipid profile, fasting blood glucose (FBG), and serum SAA1. RESULTS Both serum SAA1 and FBG are significantly elevated in patients with AV than control (p < 0.0001 and p < 0.001, respectively). Furthermore, there are positive correlations between AV severity and SAA1 and FBG (p < 0.001 and p < 0.0001, respectively). CONCLUSIONS Serum amyloid A1 is increased in AV, and this elevation may play a role in the inflammatory milieu of AV.
Collapse
Affiliation(s)
- Essam Mohamed Akl
- Faculty of Medicine, Department of Dermatology, Venereology & Andrology, Benha University, Benha, Egypt
| | - Walid A Abdel Halim
- Faculty of Medicine, Department of Clinical and Chemical Pathology, Benha University, Benha, Egypt
| |
Collapse
|
17
|
Hemed-Shaked M, Cowman MK, Kim JR, Huang X, Chau E, Ovadia H, Amar KO, Eshkar-Sebban L, Melamed M, Lev LB, Kedar E, Armengol J, Alemany J, Beyth S, Okon E, Kanduc D, Elgavish S, Wallach-Dayan SB, Cohen SJ, Naor D. MTADV 5-MER peptide suppresses chronic inflammations as well as autoimmune pathologies and unveils a new potential target-Serum Amyloid A. J Autoimmun 2021; 124:102713. [PMID: 34390919 DOI: 10.1016/j.jaut.2021.102713] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 11/17/2022]
Abstract
Despite the existence of potent anti-inflammatory biological drugs e.g., anti-TNF and anti IL-6 receptor antibodies, for treating chronic inflammatory and autoimmune diseases, these are costly and not specific. Cheaper oral available drugs remain an unmet need. Expression of the acute phase protein Serum Amyloid A (SAA) is dependent on release of pro-inflammatory cytokines IL-1, IL-6 and TNF-α during inflammation. Conversely, SAA induces pro-inflammatory cytokine secretion, including Th17, leading to a pathogenic vicious cycle and chronic inflammation. 5- MER peptide (5-MP) MTADV (methionine-threonine-alanine-aspartic acid-valine), also called Amilo-5MER, was originally derived from a sequence of a pro-inflammatory CD44 variant isolated from synovial fluid of a Rheumatoid Arthritis (RA) patient. This human peptide displays an efficient anti-inflammatory effects to ameliorate pathology and clinical symptoms in mouse models of RA, Inflammatory Bowel Disease (IBD) and Multiple Sclerosis (MS). Bioinformatics and qRT-PCR revealed that 5-MP, administrated to encephalomyelytic mice, up-regulates genes contributing to chronic inflammation resistance. Mass spectrometry of proteins that were pulled down from an RA synovial cell extract with biotinylated 5-MP, showed that it binds SAA. 5-MP disrupted SAA assembly, which is correlated with its pro-inflammatory activity. The peptide MTADV (but not scrambled TMVAD) significantly inhibited the release of pro-inflammatory cytokines IL-6 and IL-1β from SAA-activated human fibroblasts, THP-1 monocytes and peripheral blood mononuclear cells. 5-MP suppresses the pro-inflammatory IL-6 release from SAA-activated cells, but not from non-activated cells. 5-MP could not display therapeutic activity in rats, which are SAA deficient, but does inhibit inflammations in animal models of IBD and MS, both are SAA-dependent, as shown by others in SAA knockout mice. In conclusion, 5-MP suppresses chronic inflammation in animal models of RA, IBD and MS, which are SAA-dependent, but not in animal models, which are SAA-independent.
Collapse
Affiliation(s)
- Maayan Hemed-Shaked
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Mary K Cowman
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY, USA
| | - Jin Ryoun Kim
- Othmer-Jacobs Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, Brooklyn, USA
| | - Xiayun Huang
- Othmer-Jacobs Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, Brooklyn, USA
| | - Edward Chau
- Othmer-Jacobs Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, Brooklyn, USA
| | - Haim Ovadia
- Department of Neurology, Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Keren-Or Amar
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Lora Eshkar-Sebban
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Michal Melamed
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Libat Bar Lev
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Eli Kedar
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | | | | | - Shaul Beyth
- Orthopedic Surgery Department, Hadassah University Hospital, Jerusalem, Israel
| | - Eli Okon
- Department of Pathology, Hadassah University Hospital, Jerusalem, Israel
| | - Darja Kanduc
- Biotechnologies and Biopharmaceutics, University of Bari, Bari, 70126, Italy
| | - Sharona Elgavish
- Bioinformatics Unit of the Hebrew University of Jerusalem and Hadassah Medical Center, Israel
| | - Shulamit B Wallach-Dayan
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah-Hebrew University Medical Center, Israel
| | - Shmuel Jaffe Cohen
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - David Naor
- The Lautenberg Center of Immunology and Cancer Research, Faculty of Medicine, Hebrew University of Jerusalem, Israel.
| |
Collapse
|
18
|
Olivier DW, Pretorius E, Engelbrecht AM. Serum amyloid A1: Innocent bystander or active participant in cell migration in triple-negative breast cancer? Exp Cell Res 2021; 406:112759. [PMID: 34332984 DOI: 10.1016/j.yexcr.2021.112759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 01/26/2023]
Abstract
The Serum Amyloid A (SAA) family of proteins is associated with various pathological conditions, including cancer. However, their role in cancer is incompletely understood. Here, we investigated the role of SAA1 in cell cycle regulation, apoptosis, survival signaling, metabolism, and metastasis in models of triple-negative breast cancer (TNBC), using RNAi. Our data show that in untransformed epithelial cells (MCF12A), the knockdown of SAA1 induces the expression of cell cycle regulators (MCM2, p53), the activation of DNA repair (PARP synthesis), and survival signaling (NFκB). In contrast, knockdown of SAA1 in the TNBC cell line (MDA-MB-231) induced the expression p16 and shifted cells in the cell cycle from the S to G2/M phase, without the activation of DNA repair. Moreover, in SAA1-deficient MDA-MB-231 and HCC70 cells, metabolism (NADH oxidation) continually increased while cell migration (% wound closure and the rate of wound closure) decreased. However, silencing of SAA1 altered epithelial and mesenchymal markers in MCF12A (E-cadherin, Laminin 1β, Vimentin) and MDA-MB-231 (α-Smooth muscle actin) cells, associated with the metastatic program of epithelial-mesenchymal transition. Nonetheless, our data provide evidence that SAA1 could potentially serve as a therapeutic target in TNBC.
Collapse
Affiliation(s)
- Daniel Wilhelm Olivier
- Department of Physiological Sciences, Stellenbosch University, Mike De Vries Building, Corner Merriman and Bosman Road, Stellenbosch, 7602, South Africa.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Mike De Vries Building, Corner Merriman and Bosman Road, Stellenbosch, 7602, South Africa.
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Mike De Vries Building, Corner Merriman and Bosman Road, Stellenbosch, 7602, South Africa.
| |
Collapse
|
19
|
O'Reilly S. Serum amyloid A as a universal biomarker in lung disease. Intern Med J 2021; 51:1195-1196. [PMID: 34278698 DOI: 10.1111/imj.15324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 11/29/2022]
|
20
|
Jung BG, Vankayalapati R, Samten B. Mycobacterium tuberculosis stimulates IL-1β production by macrophages in an ESAT-6 dependent manner with the involvement of serum amyloid A3. Mol Immunol 2021; 135:285-293. [PMID: 33957478 DOI: 10.1016/j.molimm.2021.04.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/26/2021] [Accepted: 04/25/2021] [Indexed: 12/16/2022]
Abstract
Despite its critical roles in immune responses against tuberculosis infection and immune pathology, the molecular details of interleukin (IL)-1β production in tuberculosis infection remain elusive. To explore IL-1β production in tuberculosis infection, we infected mouse bone marrow-derived macrophages (BMDM) with Mycobacterium tuberculosis (Mtb) H37Rv, its early secreted antigenic target protein of 6 kDa (ESAT-6) gene deletion (H37Rv:Δ3875) or complemented strain (H37Rv:Δ3875C) and evaluated IL-1β production. H37Rv induced significantly increased IL-1β production by BMDMs compared to non-infected BMDMs. In contrast, H37Rv:Δ3875 induced significantly less mature IL-1β production despite eliciting comparable levels of pro-IL-1β and IL-8 from BMDMs compared to H37Rv and H37Rv:Δ3875C. Blocking either NLRP3 or K+ efflux diminished H37Rv-induced IL-1β production by BMDMs. Infection of mice intranasally with H37Rv:Δ3875 induced less IL-1β production in the lungs compared with H37Rv. Intranasal delivery of ESAT-6 but not CFP10 induced production of IL-1β in mouse lungs and RNA-Seq analysis identified serum amyloid A (SAA) 3 as one of the highly expressed genes in mouse lungs. Infection of mice with H37Rv but not H37Rv:Δ3875 induced expression of lung SAA3 mRNA and protein, consistent with the effect of intranasal delivery of ESAT-6. Silencing SAA3 reduced Mtb-induced IL-1β production by BMDMs. We conclude that SAA3 plays critical role in ESAT-6 dependent IL-1β production by macrophages in tuberculosis infection.
Collapse
Affiliation(s)
- Bock-Gie Jung
- Department of Pulmonary Immunology, University of Texas Health Science Center at Tyler, TX, 75708, USA
| | - Ramakrishna Vankayalapati
- Department of Pulmonary Immunology, University of Texas Health Science Center at Tyler, TX, 75708, USA
| | - Buka Samten
- Department of Pulmonary Immunology, University of Texas Health Science Center at Tyler, TX, 75708, USA.
| |
Collapse
|
21
|
Sorić Hosman I, Kos I, Lamot L. Serum Amyloid A in Inflammatory Rheumatic Diseases: A Compendious Review of a Renowned Biomarker. Front Immunol 2021; 11:631299. [PMID: 33679725 PMCID: PMC7933664 DOI: 10.3389/fimmu.2020.631299] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Serum amyloid A (SAA) is an acute phase protein with a significant importance for patients with inflammatory rheumatic diseases (IRD). The central role of SAA in pathogenesis of IRD has been confirmed by recent discoveries, including its involvement in the activation of the inflammasome cascade and recruitment of interleukin 17 producing T helper cells. Clinical utility of SAA in IRD was originally evaluated nearly half a century ago. From the first findings, it was clear that SAA could be used for evaluating disease severity and monitoring disease activity in patients with rheumatoid arthritis and secondary amyloidosis. However, cost-effective and more easily applicable markers, such as C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR), overwhelmed its use in clinical practice. In the light of emerging evidences, SAA has been discerned as a more sensitive biomarker in a wide spectrum of IRD, especially in case of subclinical inflammation. Furthermore, a growing number of studies are confirming the advantages of SAA over many other biomarkers in predicting and monitoring response to biological immunotherapy in IRD patients. Arising scientific discoveries regarding the role of SAA, as well as delineating SAA and its isoforms as the most sensitive biomarkers in various IRD by recently developing proteomic techniques are encouraging the revival of its clinical use. Finally, the most recent findings have shown that SAA is a biomarker of severe Coronavirus disease 2019 (COVID-19). The aim of this review is to discuss the SAA-involving immune system network with emphasis on mechanisms relevant for IRD, as well as usefulness of SAA as a biomarker in various IRD. Therefore, over a hundred original papers were collected through an extensive PubMed and Scopus databases search. These recently arising insights will hopefully lead to a better management of IRD patients and might even inspire the development of new therapeutic strategies with SAA as a target.
Collapse
Affiliation(s)
- Iva Sorić Hosman
- Department of Pediatrics, Zadar General Hospital, Zadar, Croatia
| | - Ivanka Kos
- Division of Nephrology, Dialysis and Transplantation, Department of Pediatrics, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Lovro Lamot
- Division of Nephrology, Dialysis and Transplantation, Department of Pediatrics, University Hospital Centre Zagreb, Zagreb, Croatia.,Department of Pediatrics, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
22
|
Innate immune response in systemic autoimmune diseases: a potential target of therapy. Inflammopharmacology 2020; 28:1421-1438. [DOI: 10.1007/s10787-020-00762-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023]
|
23
|
Worrell JC, O'Reilly S. Bi-directional communication: Conversations between fibroblasts and immune cells in systemic sclerosis. J Autoimmun 2020; 113:102526. [PMID: 32713676 DOI: 10.1016/j.jaut.2020.102526] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 02/09/2023]
Abstract
Systemic Sclerosis (SSc) is an autoimmune idiopathic connective tissue disease, characterized by aberrant fibro-proliferative and inflammatory responses, causing fibrosis of multiple organs. In recent years the interactions between innate and adaptive immune cells with resident fibroblasts have been uncovered. Cross-talk between immune and stromal cells mediates activation of stromal cells to myofibroblasts; key cells in the pathophysiology of fibrosis. These cells and their cytokines appear to mediate their effects in both a paracrine and autocrine fashion. This review examines the role of innate and adaptive immune cells in SSc, focusing on recent advances that have illuminated our understanding of ongoing bi-directional communication between immune and stromal cells. Finally, we appraise current and future therapies and how these may be useful in a disease that currently has no specific disease modifying treatment.
Collapse
Affiliation(s)
- Julie C Worrell
- Insititute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Steven O'Reilly
- Durham University, Biosciences, Faculty of Science, Durham, UK. steven.o'
| |
Collapse
|
24
|
Biological Characterization of Commercial Recombinantly Expressed Immunomodulating Proteins Contaminated with Bacterial Products in the Year 2020: The SAA3 Case. Mediators Inflamm 2020; 2020:6087109. [PMID: 32694927 PMCID: PMC7362292 DOI: 10.1155/2020/6087109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/19/2020] [Accepted: 06/03/2020] [Indexed: 01/20/2023] Open
Abstract
The serum amyloid A (SAA) gene family is highly conserved and encodes acute phase proteins that are upregulated in response to inflammatory triggers. Over the years, a considerable amount of literature has been published attributing a wide range of biological effects to SAAs such as leukocyte recruitment, cytokine and chemokine expression and induction of matrix metalloproteinases. Furthermore, SAAs have also been linked to protumorigenic, proatherogenic and anti-inflammatory effects. Here, we investigated the biological effects conveyed by murine SAA3 (mu rSAA3) recombinantly expressed in Escherichia coli. We observed the upregulation of a number of chemokines including CCL2, CCL3, CXCL1, CXCL2, CXCL6 or CXCL8 following stimulation of monocytic, fibroblastoid and peritoneal cells with mu rSAA3. Furthermore, this SAA variant displayed potent in vivo recruitment of neutrophils through the activation of TLR4. However, a major problem associated with proteins derived from recombinant expression in bacteria is potential contamination with various bacterial products, such as lipopolysaccharide, lipoproteins and formylated peptides. This is of particular relevance in the case of SAA as there currently exists a discrepancy in biological activity between SAA derived from recombinant expression and that of an endogenous source, i.e. inflammatory plasma. Therefore, we subjected commercial recombinant mu rSAA3 to purification to homogeneity via reversed-phase high-performance liquid chromatography (RP-HPLC) and re-assessed its biological potential. RP-HPLC-purified mu rSAA3 did not induce chemokines and lacked in vivo neutrophil chemotactic activity, but retained the capacity to synergize with CXCL8 in the activation of neutrophils. In conclusion, experimental results obtained when using proteins recombinantly expressed in bacteria should always be interpreted with care.
Collapse
|
25
|
Investigation of Potential Genetic Biomarkers and Molecular Mechanism of Ulcerative Colitis Utilizing Bioinformatics Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4921387. [PMID: 32190668 PMCID: PMC7073481 DOI: 10.1155/2020/4921387] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/28/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022]
Abstract
Objectives To reveal the molecular mechanisms of ulcerative colitis (UC) and provide potential biomarkers for UC gene therapy. Methods We downloaded the GSE87473 microarray dataset from the Gene Expression Omnibus (GEO) and identified the differentially expressed genes (DEGs) between UC samples and normal samples. Then, a module partition analysis was performed based on a weighted gene coexpression network analysis (WGCNA), followed by pathway and functional enrichment analyses. Furthermore, we investigated the hub genes. At last, data validation was performed to ensure the reliability of the hub genes. Results Between the UC group and normal group, 988 DEGs were investigated. The DEGs were clustered into 5 modules using WGCNA. These DEGs were mainly enriched in functions such as the immune response, the inflammatory response, and chemotaxis, and they were mainly enriched in KEGG pathways such as the cytokine-cytokine receptor interaction, chemokine signaling pathway, and complement and coagulation cascades. The hub genes, including dual oxidase maturation factor 2 (DUOXA2), serum amyloid A (SAA) 1 and SAA2, TNFAIP3-interacting protein 3 (TNIP3), C-X-C motif chemokine (CXCL1), solute carrier family 6 member 14 (SLC6A14), and complement decay-accelerating factor (CD antigen CD55), were revealed as potential tissue biomarkers for UC diagnosis or treatment. Conclusions This study provides supportive evidence that DUOXA2, A-SAA, TNIP3, CXCL1, SLC6A14, and CD55 might be used as potential biomarkers for tissue biopsy of UC, especially SLC6A14 and DUOXA2, which may be new targets for UC gene therapy. Moreover, the DUOX2/DUOXA2 and CXCL1/CXCR2 pathways might play an important role in the progression of UC through the chemokine signaling pathway and inflammatory response.
Collapse
|
26
|
Frasca L, Lande R. Toll-like receptors in mediating pathogenesis in systemic sclerosis. Clin Exp Immunol 2020; 201:14-24. [PMID: 32048277 DOI: 10.1111/cei.13426] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2020] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors (TLRs) are evolutionarily conserved receptors essential for the host defence against pathogens. Both immune and non-immune cells can express TLRs, although at different levels. Systemic sclerosis (SSc) is a chronic disease in which autoimmunity, dysregulated profibrotic mediator release and activation of fibroblasts lead to dysregulated collagen deposition and fibrosis. There is now increasing knowledge that the innate immune system and, in particular, TLRs take a part in SSc pathogenesis. The list of endogenous ligands that can stimulate TLRs in SSc is growing: these ligands represent specific danger-associated molecular patterns (DAMPs), involved either in the initiation or the perpetuation of inflammation, and in the release of factors that sustain the fibrotic process or directly stimulate the cells that produce collagen and the endothelial cells. This review reports evidences concerning TLR signalling involvement in SSc. We report the new DAMPs, as well as the TLR-linked pathways involved in disease, with emphasis on type I interferon signature in SSc, the role of plasmacytoid dendritic cells (pDCs) and platelets. The dissection of the contribution of all these pathways to disease, and their correlation with the disease status, as well as their values as prognostic tools, can help to plan timely intervention and design new drugs for more appropriate therapeutic strategies.
Collapse
Affiliation(s)
- L Frasca
- National Centre for Drug Research and Evaluation, Pharmacological Research and Experimental Therapy Unit, Istituto Superiore di Sanità, Rome, Italy
| | - R Lande
- National Centre for Drug Research and Evaluation, Pharmacological Research and Experimental Therapy Unit, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
27
|
Hahn A, Kny M, Pablo-Tortola C, Todiras M, Willenbrock M, Schmidt S, Schmoeckel K, Jorde I, Nowak M, Jarosch E, Sommer T, Bröker BM, Felix SB, Scheidereit C, Weber-Carstens S, Butter C, Luft FC, Fielitz J. Serum amyloid A1 mediates myotube atrophy via Toll-like receptors. J Cachexia Sarcopenia Muscle 2020; 11:103-119. [PMID: 31441598 PMCID: PMC7015249 DOI: 10.1002/jcsm.12491] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 06/28/2019] [Accepted: 07/22/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Critically ill patients frequently develop muscle atrophy and weakness in the intensive-care-unit setting [intensive care unit-acquired weakness (ICUAW)]. Sepsis, systemic inflammation, and acute-phase response are major risk factors. We reported earlier that the acute-phase protein serum amyloid A1 (SAA1) is increased and accumulates in muscle of ICUAW patients, but its relevance was unknown. Our objectives were to identify SAA1 receptors and their downstream signalling pathways in myocytes and skeletal muscle and to investigate the role of SAA1 in inflammation-induced muscle atrophy. METHODS We performed cell-based in vitro and animal in vivo experiments. The atrophic effect of SAA1 on differentiated C2C12 myotubes was investigated by analysing gene expression, protein content, and the atrophy phenotype. We used the cecal ligation and puncture model to induce polymicrobial sepsis in wild type mice, which were treated with the IкB kinase inhibitor Bristol-Myers Squibb (BMS)-345541 or vehicle. Morphological and molecular analyses were used to investigate the phenotype of inflammation-induced muscle atrophy and the effects of BMS-345541 treatment. RESULTS The SAA1 receptors Tlr2, Tlr4, Cd36, P2rx7, Vimp, and Scarb1 were all expressed in myocytes and skeletal muscle. Treatment of differentiated C2C12 myotubes with recombinant SAA1 caused myotube atrophy and increased interleukin 6 (Il6) gene expression. These effects were mediated by Toll-like receptors (TLR) 2 and 4. SAA1 increased the phosphorylation and activity of the transcription factor nuclear factor 'kappa-light-chain-enhancer' of activated B-cells (NF-κB) p65 via TLR2 and TLR4 leading to an increased binding of NF-κB to NF-κB response elements in the promoter region of its target genes resulting in an increased expression of NF-κB target genes. In polymicrobial sepsis, skeletal muscle mass, tissue morphology, gene expression, and protein content were associated with the atrophy response. Inhibition of NF-κB signalling by BMS-345541 increased survival (28.6% vs. 91.7%, P < 0.01). BMS-345541 diminished inflammation-induced atrophy as shown by a reduced weight loss of the gastrocnemius/plantaris (vehicle: -21.2% and BMS-345541: -10.4%; P < 0.05), tibialis anterior (vehicle: -22.7% and BMS-345541: -17.1%; P < 0.05) and soleus (vehicle: -21.1% and BMS-345541: -11.3%; P < 0.05) in septic mice. Analysis of the fiber type specific myocyte cross-sectional area showed that BMS-345541 reduced inflammation-induced atrophy of slow/type I and fast/type II myofibers compared with vehicle-treated septic mice. BMS-345541 reversed the inflammation-induced atrophy program as indicated by a reduced expression of the atrogenes Trim63/MuRF1, Fbxo32/Atrogin1, and Fbxo30/MuSA1. CONCLUSIONS SAA1 activates the TLR2/TLR4//NF-κB p65 signalling pathway to cause myocyte atrophy. Systemic inhibition of the NF-κB pathway reduced muscle atrophy and increased survival of septic mice. The SAA1/TLR2/TLR4//NF-κB p65 atrophy pathway could have utility in combatting ICUAW.
Collapse
Affiliation(s)
- Alexander Hahn
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Melanie Kny
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Cristina Pablo-Tortola
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Mihail Todiras
- Cardiovascular hormones, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Nicolae Testemiţanu State University of Medicine and Pharmacy, Chișinău, Moldova
| | - Michael Willenbrock
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sibylle Schmidt
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Katrin Schmoeckel
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine, Greifswald, Germany
| | - Ilka Jorde
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine, Greifswald, Germany
| | - Marcel Nowak
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Intracellular Proteolysis, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ernst Jarosch
- Intracellular Proteolysis, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Thomas Sommer
- Intracellular Proteolysis, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Institute of Biology, Humboldt-University Berlin, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Barbara M Bröker
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine, Greifswald, Germany
| | - Stephan B Felix
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Claus Scheidereit
- Signal Transduction in Tumor Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Steffen Weber-Carstens
- Department of Anesthesiology and Intensive Care Medicine, Campus Virchow-Klinikum and Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Christian Butter
- Department of Cardiology, Heart Center Brandenburg and Medical University Brandenburg (MHB), Bernau, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jens Fielitz
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
28
|
Wang Y, Cao F, Wang Y, Yu G, Jia BL. Silencing of SAA1 inhibits palmitate- or high-fat diet induced insulin resistance through suppression of the NF-κB pathway. Mol Med 2019; 25:17. [PMID: 31060494 PMCID: PMC6503374 DOI: 10.1186/s10020-019-0075-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 03/13/2019] [Indexed: 12/14/2022] Open
Abstract
Background Obesity is one of the leading causes of insulin resistance. Accumulating reports have highlighted that serum amyloid A-1 (SAA1) is a potential candidate that is capable of attenuating insulin resistance. Hence, we conducted the current study with aims of investigating our proposed hypothesis that silencing SAA1 could inhibit the progression of obesity-induced insulin resistance through the NF-κB pathway. Methods Gene expression microarray analysis was initially performed to screen differentially expressed genes (DEGs) associated with obesity. Palmitate (PA)-induced insulin resistance Huh7 cell models and high-fat diet (HFD)-induced mouse models were established to elucidate the effect of SAA1/Saa1 on insulin resistance. The NF-κB pathway-related expression was subsequently determined through the application of reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. Results Saa1 was identified as an obesity-related gene based on the microarray data of GSE39549. Saa1 was determined to be highly expressed in HFD-induced insulin resistance mouse models. PA-induced Huh7 cells, treated with silenced SAA1 or NF-κB pathway inhibition using BAY 11–7082, displayed a marked decrease in both Saa1 and SOCS3 as well as an elevation in 2DG, IRS1 and the extent of IRS1 phosphorylation. HFD mice treated with silenced Saa1 or inhibited NF-κB pathway exhibited improved fasting blood glucose (FBG) levels as well as fasting plasma insulin (FPI) levels, glucose tolerance and systemic insulin sensitivity. Saa1/SAA1 was determined to show a stimulatory effect on the transport of the NF-κBp65 protein from the cytoplasm to the nucleus both in vivo and in vitro, suggesting that Saa1/SAA1 could activate the NF-κB pathway. Conclusion Taken together, our key findings highlight a novel mechanism by which silencing of SAA1 hinders PA or HFD-induced insulin resistance through inhibition of the NF-κB pathway. Electronic supplementary material The online version of this article (10.1186/s10020-019-0075-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yong Wang
- Department of Gastrointestinal Surgery, the Second Hospital of Anhui Medical University, No. 678, Furong Road, Economic and Technological Development Zone, Hefei, 230601, Anhui Province, People's Republic of China.
| | - Feng Cao
- Department of Gastrointestinal Surgery, the Second Hospital of Anhui Medical University, No. 678, Furong Road, Economic and Technological Development Zone, Hefei, 230601, Anhui Province, People's Republic of China
| | - Yang Wang
- Department of Gastrointestinal Surgery, the Second Hospital of Anhui Medical University, No. 678, Furong Road, Economic and Technological Development Zone, Hefei, 230601, Anhui Province, People's Republic of China
| | - Gang Yu
- Department of Gastrointestinal Surgery, the Second Hospital of Anhui Medical University, No. 678, Furong Road, Economic and Technological Development Zone, Hefei, 230601, Anhui Province, People's Republic of China
| | - Ben-Li Jia
- Department of Gastrointestinal Surgery, the Second Hospital of Anhui Medical University, No. 678, Furong Road, Economic and Technological Development Zone, Hefei, 230601, Anhui Province, People's Republic of China
| |
Collapse
|
29
|
Ignacio RMC, Gibbs CR, Kim S, Lee ES, Adunyah SE, Son DS. Serum amyloid A predisposes inflammatory tumor microenvironment in triple negative breast cancer. Oncotarget 2019; 10:511-526. [PMID: 30728901 PMCID: PMC6355188 DOI: 10.18632/oncotarget.26566] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 12/29/2018] [Indexed: 12/20/2022] Open
Abstract
Acute-phase proteins (APPs) are associated with a variety of disorders such as infection, inflammatory diseases, and cancers. The signature profile of APPs in breast cancer (BC) is poorly understood. Here, we identified serum amyloid A (SAA) for proinflammatory predisposition in BC through the signature profiles of APPs, interleukin (IL) and tumor necrosis factor (TNF) superfamily using publicly available datasets of tumor samples and cell lines. Triple-negative breast cancer (TNBC) subtype highly expressed SAA1/2 compared to HER2, luminal A (LA) and luminal B (LB) subtypes. IL1A, IL1B, IL8/CXCL8, IL32 and IL27RA in IL superfamily and CD70, TNFSF9 and TNFRSF21 in TNF superfamily were highly expressed in TNBC compared to other subtypes. SAA is restrictedly regulated by nuclear factor (NF)-κB and IL-1β, an NF-κB activator highly expressed in TNBC, increased the promoter activity of SAA1 in human TNBC MDA-MB231 cells. Interestingly, two κB-sites contained in SAA1 promoter were involved, and the proximal region (-96/-87) was more critical than the distal site (-288/-279) in regulating IL-1β-induced SAA1. Among the SAA receptors, TLR1 and TLR2 were highly expressed in TNBC. Cu-CPT22, TLR1/2 antagonist, abrogated IL-1β-induced SAA1 promoter activity. In addition, SAA1 induced IL8/CXCL8 promoter activity, which was partially reduced by Cu-CPT22. Notably, SAA1/2, TLR2 and IL8/CXCL8 were associated with a poor overall survival in mesenchymal-like TNBC. Taken together, IL-1-induced SAA via NF-κB-mediated signaling could potentiate an inflammatory burden, leading to cancer progression and high mortality in TNBC patients.
Collapse
Affiliation(s)
- Rosa Mistica C Ignacio
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN, USA
| | - Carla R Gibbs
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN, USA
| | - Soohyun Kim
- Department of Veterinary Sciences, College of Veterinary Medicine, Kon-Kuk University, Seoul, Republic of Korea
| | - Eun-Sook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, USA
| | - Samuel E Adunyah
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN, USA
| | - Deok-Soo Son
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN, USA
| |
Collapse
|
30
|
Kim S, Chun SH, Park HJ, Lee SI. Systemic Sclerosis and Microbiota: Overview of Current Research Trends and Future Perspective. JOURNAL OF RHEUMATIC DISEASES 2019. [DOI: 10.4078/jrd.2019.26.4.235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Suhee Kim
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, Korea
| | - Sung Hak Chun
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, Korea
| | - Hee Jin Park
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, Korea
| | - Sang-Il Lee
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, Korea
| |
Collapse
|
31
|
Brown M, O'Reilly S. The immunopathogenesis of fibrosis in systemic sclerosis. Clin Exp Immunol 2018; 195:310-321. [PMID: 30430560 DOI: 10.1111/cei.13238] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2018] [Indexed: 02/07/2023] Open
Abstract
Systemic sclerosis (SSc) is an idiopathic systemic autoimmune disease. It is characterized by a triad of hallmarks: immune dysfunction, fibrosis and vasculopathy. Immune dysfunction in SSc is characterized by the activation and recruitment of immune cells and the production of autoantibodies and cytokines. How immune abnormalities link the fibrosis and vasculopathy in SSc is poorly understood. A plethora of immune cell types are implicated in the immunopathogenesis of SSc, including T cells, B cells, dendritic cells, mast cells and macrophages. How these different cell types interact to contribute to SSc is complicated, and can involve cell-to-cell interactions and communication via cytokines, including transforming growth factor (TGF)-β, interleukin (IL)-6 and IL-4. We will attempt to review significant and recent research demonstrating the importance of immune cell regulation in the immunopathogenesis of SSc with a particular focus on fibrosis.
Collapse
Affiliation(s)
- M Brown
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - S O'Reilly
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| |
Collapse
|
32
|
Luckett-Chastain LR, Gipson JR, Gillaspy AF, Gallucci RM. Transcriptional profiling of irritant contact dermatitis (ICD) in a mouse model identifies specific patterns of gene expression and immune-regulation. Toxicology 2018; 410:1-9. [DOI: 10.1016/j.tox.2018.08.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/07/2018] [Accepted: 08/27/2018] [Indexed: 12/16/2022]
|
33
|
Burgess EJ, Hoyt LR, Randall MJ, Mank MM, Bivona JJ, Eisenhauer PL, Botten JW, Ballif BA, Lam YW, Wargo MJ, Boyson JE, Ather JL, Poynter ME. Bacterial Lipoproteins Constitute the TLR2-Stimulating Activity of Serum Amyloid A. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2377-2384. [PMID: 30158125 PMCID: PMC6179936 DOI: 10.4049/jimmunol.1800503] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/04/2018] [Indexed: 12/21/2022]
Abstract
Studies comparing endogenous and recombinant serum amyloid A (SAA) have generated conflicting data on the proinflammatory function of these proteins. In exploring this discrepancy, we found that in contrast to commercially sourced recombinant human SAA1 (hSAA1) proteins produced in Escherichia coli, hSAA1 produced from eukaryotic cells did not promote proinflammatory cytokine production from human or mouse cells, induce Th17 differentiation, or stimulate TLR2. Proteomic analysis of E. coli-derived hSAA1 revealed the presence of numerous bacterial proteins, with several being reported or probable lipoproteins. Treatment of hSAA1 with lipoprotein lipase or addition of a lipopeptide to eukaryotic cell-derived hSAA1 inhibited or induced the production of TNF-α from macrophages, respectively. Our results suggest that a function of SAA is in the binding of TLR2-stimulating bacterial proteins, including lipoproteins, and demand that future studies of SAA employ a recombinant protein derived from eukaryotic cells.
Collapse
Affiliation(s)
- Edward J Burgess
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Laura R Hoyt
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Matthew J Randall
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Madeleine M Mank
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Joseph J Bivona
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Philip L Eisenhauer
- Immunobiology Division, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Jason W Botten
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Immunobiology Division, Department of Medicine, University of Vermont, Burlington, VT 05405
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405
| | - Bryan A Ballif
- Department of Biology, University of Vermont, Burlington, VT 05405; and
| | - Ying-Wai Lam
- Department of Biology, University of Vermont, Burlington, VT 05405; and
| | - Matthew J Wargo
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405
| | - Jonathan E Boyson
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Department of Surgery, University of Vermont, Burlington, VT 05405
| | - Jennifer L Ather
- Vermont Lung Center, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| | - Matthew E Poynter
- Vermont Lung Center, University of Vermont, Burlington, VT 05405;
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, VT 05405
- Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405
| |
Collapse
|
34
|
Henderson J, Bhattacharyya S, Varga J, O'Reilly S. Targeting TLRs and the inflammasome in systemic sclerosis. Pharmacol Ther 2018; 192:163-169. [PMID: 30081049 DOI: 10.1016/j.pharmthera.2018.08.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Systemic sclerosis (SSc) is an idiopathic autoimmune disease characterised by inflammation, vascular problems, cytokine dysregulation and ultimately fibrosis, which accounts for poor prognosis and eventual mortality. At present no curative treatments exist, hence there is an urgent need to better understand the aetiology and develop improved therapies accordingly. Although still widely debated, significant evidence points to upregulation of the innate immune response via the activity of Toll-like receptors (TLRs) and the NLRP3 inflammasome as the start points in a cascade of signaling events which drives excessive extracellular matrix protein production, causing fibrosis. Herein the recent breakthroughs which have implicated TLR signaling and the NLRP3 inflammasome in SSc and the novel therapeutic possibilities this introduces to the field will be discussed.
Collapse
Affiliation(s)
- John Henderson
- Faculty of Health and Life Sciences, Northumbria University, Ellison Building, Newcastle upon Tyne NE2 8ST, United Kingdom
| | | | - John Varga
- Department of Medicine, Northwestern University, Chicago, USA
| | - Steven O'Reilly
- Faculty of Health and Life Sciences, Northumbria University, Ellison Building, Newcastle upon Tyne NE2 8ST, United Kingdom.
| |
Collapse
|
35
|
Meiners S, Evankovich J, Mallampalli RK. The ubiquitin proteasome system as a potential therapeutic target for systemic sclerosis. Transl Res 2018; 198:17-28. [PMID: 29702079 DOI: 10.1016/j.trsl.2018.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/20/2018] [Accepted: 03/24/2018] [Indexed: 01/16/2023]
Abstract
The present review aims to summarize available knowledge on the role of the ubiquitin-proteasome system (UPS) in the pathogenesis of scleroderma and scleroderma-related disease mechanisms. This will provide the reader with a more mechanistic understanding of disease pathogenesis and help to identify putative novel targets within the UPS for potential therapeutic intervention. Because of the heterogenous manifestations of scleroderma, we will primarily focus on conserved mechanisms that are involved in the development of lung scleroderma phenotypes.
Collapse
Affiliation(s)
- Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig Maximilians University, Helmholtz Zentrum München, Germany; Comprehensive Pneumology Center, Munich (CPC-M), Germany; Member of the German Center for Lung Research (DZL), Munich, Germany.
| | - John Evankovich
- Pulmonary, Allergy, and Critical Care Medicine, Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rama K Mallampalli
- Pulmonary, Allergy, and Critical Care Medicine, Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Cell Biology and Physiology, University of Pittsburgh, Pittsburgh, PA, USA; Medical Specialty Service Line, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
36
|
Laurent P, Sisirak V, Lazaro E, Richez C, Duffau P, Blanco P, Truchetet ME, Contin-Bordes C. Innate Immunity in Systemic Sclerosis Fibrosis: Recent Advances. Front Immunol 2018; 9:1702. [PMID: 30083163 PMCID: PMC6064727 DOI: 10.3389/fimmu.2018.01702] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022] Open
Abstract
Systemic sclerosis (SSc) is a heterogeneous autoimmune disease characterized by three interconnected hallmarks (i) vasculopathy, (ii) aberrant immune activation, and (iii) fibroblast dysfunction leading to extracellular matrix deposition and fibrosis. Blocking or reversing the fibrotic process associated with this devastating disease is still an unmet clinical need. Although various components of innate immunity, including macrophages and type I interferon, have long been implicated in SSc, the precise mechanisms that regulate the global innate immune contribution to SSc pathogenesis remain poorly understood. Recent studies have identified new innate immune players, such as pathogen-recognition receptors, platelet-derived danger-associated molecular patterns, innate lymphoid cells, and plasmacytoid dendritic cells in the pathophysiology of SSc, including vasculopathy and fibrosis. In this review, we describe the evidence demonstrating the importance of innate immune processes during SSc development with particular emphasis on their role in the initiation of pathology. We also discuss potential therapeutic options to modulate innate immune cells or signaling in SSc that are emerging from these recent advances.
Collapse
Affiliation(s)
- Paoline Laurent
- CNRS-UMR 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France
| | - Vanja Sisirak
- CNRS-UMR 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France
| | - Estibaliz Lazaro
- CNRS-UMR 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France.,Internal Medicine Department, Bordeaux University Hospital, Bordeaux, France
| | - Christophe Richez
- CNRS-UMR 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France.,Rheumatology Department, Bordeaux University Hospital, Bordeaux, France
| | - Pierre Duffau
- CNRS-UMR 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France.,Internal Medicine Department, Bordeaux University Hospital, Bordeaux, France
| | - Patrick Blanco
- CNRS-UMR 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France.,Immunology and Immunogenetic Department, Bordeaux University Hospital, Bordeaux, France
| | - Marie-Elise Truchetet
- CNRS-UMR 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France.,Rheumatology Department, Bordeaux University Hospital, Bordeaux, France
| | - Cécile Contin-Bordes
- CNRS-UMR 5164, ImmunoConcEpT, Bordeaux University, Bordeaux, France.,Immunology and Immunogenetic Department, Bordeaux University Hospital, Bordeaux, France
| |
Collapse
|
37
|
Novel insights into the role of inflammasomes in autoimmune and metabolic rheumatic diseases. Rheumatol Int 2018; 38:1345-1354. [DOI: 10.1007/s00296-018-4074-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/29/2018] [Indexed: 12/12/2022]
|
38
|
Morizane S, Kajita A, Mizuno K, Takiguchi T, Iwatsuki K. Toll-like receptor signalling induces the expression of serum amyloid A in epidermal keratinocytes and dermal fibroblasts. Clin Exp Dermatol 2018; 44:40-46. [PMID: 29770468 DOI: 10.1111/ced.13604] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Toll-like receptors (TLRs) play critical roles in innate immune response by sensing pathogen- or damage-associated molecular patterns. Epidermal keratinocytes and dermal fibroblasts also produce proinflammatory cytokines and chemokines under stimulation with TLR ligands. Serum amyloid A (SAA) is an essential factor in the pathogenesis of secondary amyloidosis, and also has immunomodulatory functions. SAA are produced mainly by hepatocytes but also by a variety of cells, including immune cells, endothelial cells, synoviocytes, and epidermal keratinocytes. However, SAA expression in human dermal fibroblasts has not been shown to date. AIM To investigate the effect of TLR ligands on SAA expression in epidermal keratinocytes and dermal fibroblasts. METHODS We investigated whether TLR ligands induce the expression of SAA in normal human epidermal keratinocytes (NHEKs) and normal human dermal fibroblasts (NHDFs) by real-time quantitative PCR and ELISA. The effect of SAA on its own expression in NHDFs was also studied. RESULTS SAA expression was induced via nuclear factor-κB by TLR1/2, 3, 5 and 2/6 ligands in NHEKs. In NHDFs, TLR1/2 and TLR2/6 ligands increased SAA expression. SAA further induced its own expression via TLR1/2 and NF-κB in NHDFs, as previously reported for NHEKs. CONCLUSIONS Our results provide new evidence that the skin's innate immune response contributes to the production of SAA, which might lead to an increased risk of systemic complications such as secondary amyloidosis of recessive dystrophic epidermolysis bullosa.
Collapse
Affiliation(s)
- S Morizane
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - A Kajita
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - K Mizuno
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - T Takiguchi
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.,Department of Plastic Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - K Iwatsuki
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
39
|
Ng CT, Yip GWC, Chen ES, Poh WYR, Bay BH, Yung LYL. Gold nanoparticles induce serum amyloid A 1-Toll-like receptor 2 mediated NF-kB signaling in lung cells in vitro. Chem Biol Interact 2018; 289:81-89. [PMID: 29705079 DOI: 10.1016/j.cbi.2018.04.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 02/05/2018] [Accepted: 04/23/2018] [Indexed: 11/19/2022]
Abstract
Gold nanoparticles (AuNPs) have emerging applications in biomedicine and the industry. Exposure to AuNPs has previously been shown to alter the transcriptional activity of nuclear factor kappa B (NF-kB), which is known to mediate physiological and pathological processes. This study seeks to provide mechanistic insights into AuNP-induced NF-kB activation in Small Airway Epithelial Cells (SAECs) in vitro. Increased NF-kB transcriptional activity (quantified by the luciferase reporter assay) was observed in AuNP-treated SAECs. Transcriptomic analysis revealed differential expression of 42 genes, which regulate functional processes that include cellular response to stimulus, chemicals and stress as well as immune response. Notably, the gene expression of serum amyloid A1 (SAA1), an acute phase protein and Toll-like receptor 2 (TLR2) were found to be up-regulated. As TLR2 is known to be a functional receptor of SAA1, a co-immunoprecipitation assay was performed. SAA1 was observed to be co-immunoprecipitated with the TLR2 protein and this protein-protein interaction was further supported by in silico computer based protein modeling. The present study suggests that AuNPs may potentially induce SAA1-TLR2-mediated NF-kB transcription factor activation in lung epithelial cells, highlighting that nano-bio interactions could result in biological effects that may affect cells.
Collapse
Affiliation(s)
- Cheng Teng Ng
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117594, Singapore.
| | - George Wai Cheong Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117594, Singapore
| | - Ee Sin Chen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Wan Yan Rebecca Poh
- Carl Zeiss Pte Ltd, Microscopy Business Group, Electron Microscopy, Singapore
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117594, Singapore.
| | - Lin Yue Lanry Yung
- Department of Chemical & Biomolecular Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117585, Singapore.
| |
Collapse
|
40
|
Lis-Święty A, Widuchowska M, Brzezińska-Wcisło L, Kucharz E. High acute phase protein levels correlate with pulmonary and skin involvement in patients with diffuse systemic sclerosis. J Int Med Res 2018; 46:1634-1639. [PMID: 29512396 PMCID: PMC6091829 DOI: 10.1177/0300060518760955] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Objective This study was performed to evaluate the serum amyloid A (SAA) and C-reactive protein (CRP) levels in patients with diffuse systemic sclerosis (dSSc) in relation to a control group, disease duration, and skin and pulmonary involvement. Methods This case-control study included 18 patients with early dSSc, 15 patients with late dSSc, and 15 healthy controls. The SAA and CRP levels, modified Rodnan skin score (mRSS), and diffusing capacity of the lungs for carbon monoxide (DLCO) were determined in all patients. Results The SAA and CRP levels were significantly higher in patients with early and late dSSc than in healthy controls. The frequency of detection of elevated SAA and CRP levels was approximately 66% and 85%, respectively. A significant correlation was found between the SAA and CRP levels in patients with dSSc. The SAA and CRP levels were inversely correlated with DLCO. The CRP level was positively correlated with the mRSS. Conclusions High SAA and CRP levels could serve as biomarkers for pulmonary involvement. The serum CRP level accurately reflects the extension of skin thickening in patients with dSSc.
Collapse
Affiliation(s)
- Anna Lis-Święty
- 1 473237 Medical University of Silesia in Katowice, School of Medicine in Katowice, Chair and Department of Dermatology, Katowice, Poland
| | - Małgorzata Widuchowska
- 2 473237 Medical University of Silesia in Katowice, School of Medicine in Katowice, Chair and Department of Internal Medicine and Rheumatology, Katowice, Poland
| | - Ligia Brzezińska-Wcisło
- 1 473237 Medical University of Silesia in Katowice, School of Medicine in Katowice, Chair and Department of Dermatology, Katowice, Poland
| | - Eugeniusz Kucharz
- 2 473237 Medical University of Silesia in Katowice, School of Medicine in Katowice, Chair and Department of Internal Medicine and Rheumatology, Katowice, Poland
| |
Collapse
|
41
|
Sun C, Chen SY. RGC32 Promotes Bleomycin-Induced Systemic Sclerosis in a Murine Disease Model by Modulating Classically Activated Macrophage Function. THE JOURNAL OF IMMUNOLOGY 2018; 200:2777-2785. [PMID: 29507108 DOI: 10.4049/jimmunol.1701542] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/10/2018] [Indexed: 12/20/2022]
Abstract
Systemic sclerosis (SSc) is a multisystem autoimmune disorder that is characterized by inflammation and fibrosis in the skin and internal organs. Previous studies indicate that inflammatory cells and cytokines play essential roles in the pathogenesis of SSc; however, the mechanisms that underlie the inflammation-driven development of SSc are not fully understood. In this study, we show that response gene to complement 32 (RGC32) is abundantly expressed in mouse macrophages in the early stage of bleomycin-induced SSc. Importantly, RGC32 is required to induce the inflammatory response during the onset of SSc, because RGC32 deficiency in mice significantly ameliorates skin and lung sclerosis and inhibits the expression of inflammatory mediators inducible NO synthase (iNOS) and IL-1β in macrophages. RGC32 appears to be a novel regulator for the differentiation of classically activated macrophages (M1 macrophages). IFN-γ and LPS stimulation induces RGC32 expression in primary peritoneal macrophages and bone marrow-derived macrophages. RGC32 deficiency impairs the polarization of M1 macrophages and attenuates iNOS and IL-1β production. Mechanistically, RGC32 interacts with NF-κB proteins and promotes iNOS and IL-1β expression by binding to their promoters. Collectively, our data reveal that RGC32 promotes the onset of SSc by regulating the inflammatory response of M1 macrophages, and it may serve as a promising therapeutic target for treating SSc.
Collapse
Affiliation(s)
- Chenming Sun
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602
| | - Shi-You Chen
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602
| |
Collapse
|
42
|
Affandi AJ, Carvalheiro T, Radstake TRDJ, Marut W. Dendritic cells in systemic sclerosis: Advances from human and mice studies. Immunol Lett 2017; 195:18-29. [PMID: 29126878 DOI: 10.1016/j.imlet.2017.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/05/2017] [Accepted: 11/06/2017] [Indexed: 12/20/2022]
Abstract
Systemic sclerosis (SSc) is a complex heterogeneous fibrotic autoimmune disease with an unknown exact etiology, and characterized by three hallmarks: fibrosis, vasculopathy, and immune dysfunction. Dendritic cells (DCs) are specialized cells in pathogen sensing with high potency of antigen presentation and capable of releasing mediators to shape the immune response. Altered DCs distributions and their impaired functions may account for their role in breaking the immune tolerance and driving inflammation in SSc, and the direct contribution of DCs in promoting endothelial dysfunction and fibrotic process has only begun to be understood. Plasmacytoid dendritic cells in particular have been implicated due to their high production of type I interferon as well as other cytokines and chemokines, including the pro-inflammatory and anti-angiogenic CXCL4. Furthermore, a deeper understanding of human and mouse DC biology has clarified their identification and function in different tissues, and novel DC subsets have only recently been discovered. In this review, we highlight key findings and recent advances exploring DC role in the pathogenesis of SSc and other related autoimmune diseases, and consideration of their potential use as targeted therapy in SSc.
Collapse
Affiliation(s)
- Alsya J Affandi
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tiago Carvalheiro
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Timothy R D J Radstake
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wioleta Marut
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Systemic sclerosis (SSc) is a heterogeneous autoimmune disease which has defined three hallmarks: Small vessel vasculopathy, production of autoantibodies and fibroblast dysfunction. The exact aetiology of the disease remains unknown, due to the complex nature of the cellular signalling pathways involved. However, there is strong and consistent evidence that the innate system, in particular toll-like receptor signalling, is contributing to the progression and perhaps onset of systemic sclerosis. In light of this evidence, this review examines the role of innate immunity in systemic sclerosis and where appropriate suggests avenues for therapeutic modulation in SSc. RECENT FINDINGS Multiple lines of evidence suggest that Toll-like receptors (TLRs) are dysregulated and emerging evidence suggests that many endogenous ligands are also elevated in the disease leading to 'sterile inflammation' and ultimately the induction of fibrosis. Currently, no effective therapy exists and exploiting the innate immune system perturbation may be one possible avenue. Innate immune dysregulation is key in SSc pathogenesis and may represent a novel target.
Collapse
|
44
|
Bhattacharyya S, Varga J. Endogenous ligands of TLR4 promote unresolving tissue fibrosis: Implications for systemic sclerosis and its targeted therapy. Immunol Lett 2017; 195:9-17. [PMID: 28964818 DOI: 10.1016/j.imlet.2017.09.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/27/2017] [Accepted: 09/27/2017] [Indexed: 02/07/2023]
Abstract
Fibrosis, the hallmark of scleroderma or systemic sclerosis (SSc), is a complex, dynamic and generally irreversible pathophysiological process that leads to tissue disruption, and lacks effective therapy. While early-stage fibrosis resembles normal wound healing, in SSc fibrosis fails to resolve. Innate immune signaling via toll-like receptors (TLRs) has recently emerged as a key driver of persistent fibrotic response in SSc. Recurrent injury in genetically predisposed individual causes generation of "damage-associated molecular patterns" (DAMPs) such as fibronectin-EDA and tenascin-C. Sensing of these danger signals by TLR4 on resident cells elicits potent stimulatory effects on fibrotic gene expression and myofibroblast differentiation, and appears to sensitize fibroblasts to the profibrotic stimulatory effect of TGF-β. Thus, DAMPs induce TLR4-mediated innate immune signaling on resident mesenchymal cells which drives the emergence and persistence of fibrotic cells in tissues, and underlies the switch from a self-limited repair response to non-resolving pathological fibrosis characteristic of SSc. In this review, we present current views of the DAMP-TLR4 axis in driving sustained fibroblasts activation and its pathogenic roles in fibrosis progression in SSc, and potential anti-fibrotic approaches for selective therapeutic targeting of TLR4 signaling.
Collapse
Affiliation(s)
- Swati Bhattacharyya
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, United States.
| | - John Varga
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
45
|
Su Q, Weindl G. Glucocorticoids and Toll-like receptor 2 cooperatively induce acute-phase serum amyloid A. Pharmacol Res 2017; 128:145-152. [PMID: 28941781 DOI: 10.1016/j.phrs.2017.09.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/08/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022]
Abstract
Serum amyloid A (SAA) is a highly conserved acute-phase protein and extrahepatic produced SAA1/2 contributes to cutaneous inflammation. Prolonged systemic or topical treatment with glucocorticoids can provoke skin diseases such as steroid-induced acne. Glucocorticoids increase Toll-like receptor 2 (TLR2) expression, however, an inflammatory mediator linked to this side effect remains elusive. We report that TLR2 agonists in combination with dexamethasone substantially increase SAA expression and production in human keratinocytes and epithelial cells. Dexamethasone-mediated SAA1 induction depends on the glucocorticoid receptor (GR). In response to Propionibacterium acnes, TLR2-activated signal transducer and activator of transcription 3 (STAT3) and nuclear factor κB (NF-κB) signaling pathways are critically involved in dexamethasone-induced SAA1 production. The formation of transcription factor complexes between GR or p300 and phospho-STAT3 was confirmed by co-immunoprecipitation in dexamethasone- and P. acnes-stimulated keratinocytes. Furthermore, dexamethasone and P. acnes-increased TLR2 and mitogen-activated protein kinase phosphatase-1 (MKP-1) contribute to induction of SAA1 and 2. Likewise, tumor necrosis factor (TNF) induces SAA1 in combination with dexamethasone. GR, transcription factors STAT3 and NF-κB, but not MKP-1, mediate TNF- and dexamethasone-induced SAA1. Conclusively, we provide evidence that glucocorticoids promote SAA1 production under infectious and sterile inflammatory conditions which may provide significant insights to the pathogenesis of steroid-induced acne.
Collapse
Affiliation(s)
- Qi Su
- Institute of Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Berlin, Germany
| | - Günther Weindl
- Institute of Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
46
|
O'Reilly S. Toll Like Receptors in systemic sclerosis: An emerging target. Immunol Lett 2017; 195:2-8. [PMID: 28888416 DOI: 10.1016/j.imlet.2017.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/25/2017] [Accepted: 09/01/2017] [Indexed: 12/21/2022]
Abstract
Pattern Recognition Receptors are critical receptors that elicit an immune response upon their activation that culminates in activation of NF-KB and cytokine secretion. Key among these receptors are the Toll-Like Receptors (TLRs). These evolutionary conserved receptors form a key part in the defence against various pathogens and comprise a key part of the innate immune system. Systemic sclerosis is an autoimmune disease in which a breach of tolerance has occurred and leads to fulminant autoimmunity, dysregulated cytokines, pro-fibrotic mediators and activation of fibroblasts leading to fibrosis via collagen deposition. It has become apparent in recent years that the innate immune system and specifically TLRs are important in disease pathogenesis; responding to internal ligands to initiate an innate immune response ultimately leading to release of a variety of factors that initiate and perpetuate fibrosis. This review will examine the recent evidence of TLR signalling in systemic sclerosis and the internal danger associated molecules that may mediate the fibrotic cascade. Evaluation of their contribution to disease in systemic sclerosis and possible therapeutic targeting will be discussed.
Collapse
Affiliation(s)
- Steven O'Reilly
- Faculty of Health and Life Sciences, Northumbria University, Ellison Building, Newcastle Upon Tyne, United Kingdom.
| |
Collapse
|
47
|
Interleukin-6 as a Multifunctional Regulator: Inflammation, Immune Response, and Fibrosis. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2017. [DOI: 10.5301/jsrd.5000265] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Interleukin 6 (IL-6) is a 184-amino acid protein cytokine that is produced by many types of cells and is expressed during states of cellular stress, such as inflammation, infection, wound sites, and cancer. IL-6 levels may increase several thousand-fold in these states and may help to coordinate the response to dysregulation of tissue homeostasis. IL-6 acts through a membrane-bound IL-6 receptor (mIL-6R), which, together with a second receptor, glycoprotein 130 (gp130), leads to the initiation of intracellular signaling (classic signaling). Given that IL-6R is expressed on only a few types of cells, though all cells express gp130, direct stimulation by IL-6 is limited to cells that express mIL-6R. However, IL-6R is also produced as a soluble, secreted protein that, together with IL-6, can stimulate all gp130-expressing cells by a process termed IL-6 trans-signaling. IL-6 trans-signaling can be blocked without affecting IL-6 classic signaling through mIL-6R. IL-6 has major effects on the adaptive and innate immune system and on mesenchymal and stromal responses during inflammation. It promotes the development of pathogenic T-helper 17 T cells and the maturation of B lymphocytes. Many innate immune cells, neutrophils, and monocytes/macrophages produce and respond to IL-6, resulting in autocrine feedback loops that amplify inflammation. IL-6 has been implicated in the pathogenesis of fibrotic diseases in which IL-6 trans-signaling has been shown to stimulate the proliferation of fibroblasts and the release of procollagen and fibronectin.
Collapse
|
48
|
Devarapu SK, Lorenz G, Kulkarni OP, Anders HJ, Mulay SR. Cellular and Molecular Mechanisms of Autoimmunity and Lupus Nephritis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 332:43-154. [PMID: 28526137 DOI: 10.1016/bs.ircmb.2016.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autoimmunity involves immune responses directed against self, which are a result of defective self/foreign distinction of the immune system, leading to proliferation of self-reactive lymphocytes, and is characterized by systemic, as well as tissue-specific, inflammation. Numerous mechanisms operate to ensure the immune tolerance to self-antigens. However, monogenetic defects or genetic variants that weaken immune tolerance render susceptibility to the loss of immune tolerance, which is further triggered by environmental factors. In this review, we discuss the phenomenon of immune tolerance, genetic and environmental factors that influence the immune tolerance, factors that induce autoimmunity such as epigenetic and transcription factors, neutrophil extracellular trap formation, extracellular vesicles, ion channels, and lipid mediators, as well as costimulatory or coinhibitory molecules that contribute to an autoimmune response. Further, we discuss the cellular and molecular mechanisms of autoimmune tissue injury and inflammation during systemic lupus erythematosus and lupus nephritis.
Collapse
Affiliation(s)
- S K Devarapu
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - G Lorenz
- Klinikum rechts der Isar, Abteilung für Nephrologie, Technische Universität München, Munich, Germany
| | | | - H-J Anders
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - S R Mulay
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany.
| |
Collapse
|
49
|
Duffy L, O'Reilly SC. Toll-like receptors in the pathogenesis of autoimmune diseases: recent and emerging translational developments. Immunotargets Ther 2016; 5:69-80. [PMID: 27579291 PMCID: PMC5001654 DOI: 10.2147/itt.s89795] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Autoinflammatory diseases are defined as the loss of self-tolerance in which an inflammatory response to self-antigens occurs, which are a significant global burden. Toll-like receptors are key pattern recognition receptors, which integrate signals leading to the activation of transcription factors and ultimately proinflammatory cytokines. Recently, it has become apparent that these are at the nexus of autoinflammatory diseases making them viable and attractive drug targets. The aim of this review was to evaluate the role of innate immunity in autoinflammatory conditions alongside the role of negative regulation while suggesting possible therapeutic targets.
Collapse
Affiliation(s)
- Laura Duffy
- Immunology and Cell Biology Group, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Steven C O'Reilly
- Immunology and Cell Biology Group, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| |
Collapse
|
50
|
Epigenetic Modulation as a Therapeutic Prospect for Treatment of Autoimmune Rheumatic Diseases. Mediators Inflamm 2016; 2016:9607946. [PMID: 27594771 PMCID: PMC4995328 DOI: 10.1155/2016/9607946] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/12/2016] [Indexed: 12/21/2022] Open
Abstract
Systemic inflammatory rheumatic diseases are considered as autoimmune diseases, meaning that the balance between recognition of pathogens and avoidance of self-attack is impaired and the immune system attacks and destroys its own healthy tissue. Treatment with conventional Disease Modifying Antirheumatic Drugs (DMARDs) and/or Nonsteroidal Anti-Inflammatory Drugs (NSAIDs) is often associated with various adverse reactions due to unspecific and toxic properties of those drugs. Although biologic drugs have largely improved the outcome in many patients, such drugs still pose significant problems and fail to provide a solution to all patients. Therefore, development of more effective treatments and improvements in early diagnosis of rheumatic diseases are badly needed in order to increase patient's functioning and quality of life. The reversible nature of epigenetic mechanisms offers a new class of drugs that modulate the immune system and inflammation. In fact, epigenetic drugs are already in use in some types of cancer or cardiovascular diseases. Therefore, epigenetic-based therapeutics that control autoimmunity and chronic inflammatory process have broad implications for the pathogenesis, diagnosis, and management of rheumatic diseases. This review summarises the latest information about potential therapeutic application of epigenetic modification in targeting immune abnormalities and inflammation of rheumatic diseases.
Collapse
|