1
|
Niepmann ST, Willemsen N, Boucher AS, Stei M, Goody P, Zietzer A, Bulic M, Billig H, Odainic A, Weisheit CK, Quast C, Adam M, Schmidt SV, Bakhtiary F, Jansen F, Nickenig G, Latz E, Zimmer S. Toll-like receptor-3 contributes to the development of aortic valve stenosis. Basic Res Cardiol 2023; 118:6. [PMID: 36723728 PMCID: PMC9892139 DOI: 10.1007/s00395-023-00980-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/02/2023]
Abstract
Aortic valve stenosis (AS) development is driven by distinct molecular and cellular mechanisms which include inflammatory pathways. Toll-like-receptor-3 (TLR3) is a lysosomal pattern-recognition receptor that binds double-stranded RNA and promotes pro-inflammatory cellular responses. In recent years, TLR3 has emerged as a major regulator of vascular inflammation. The exact role of TLR3 in the development of AS has not been investigated. Isolated human valvular interstitial cells (VICs) were stimulated with the TLR3-agonist polyIC and the resulting pro-inflammatory and pro-osteogenic response measured. Severe AS was induced in wildtype- and TLR3-/- mice via mechanical injury of the aortic valve with a coronary springwire. TLR3 activation was achieved by polyIC injection every 24 h after wire injury, while TLR3 inhibition was realized using Compound 4a (C4a) every 48 h after surgery. Endothelial mesenchymal transition (EndoMT) of human valvular endothelial cells (VECs) was assessed after polyIC stimulation. Stimulation of human VICs with polyIC promoted a strong inflammatory and pro-osteogenic reaction. Similarly, injection of polyIC marginally increased AS development in mice after wire injury. AS induction was significantly decreased in TLR3-/- mice, confirming the role of endogenous TLR3 ligands in AS pathology. Pharmacological inhibition of TLR3 with C4a not only prevented the upregulation of inflammatory cytokines and osteogenic markers in VICs, and EndoMT in VECs, but also significantly abolished the development of AS in vivo. Endogenous TLR3 activation significantly contributes to AS development in mice. Pharmacological inhibition of TLR3 with C4a prevented AS formation. Therefore, targeting TLR3 may be a viable treatment option.
Collapse
Affiliation(s)
- Sven Thomas Niepmann
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany.
| | - Nicola Willemsen
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Ann Sophie Boucher
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Marta Stei
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Philip Goody
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Andreas Zietzer
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Marko Bulic
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Hannah Billig
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Alexandru Odainic
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany ,Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC Australia
| | | | - Christine Quast
- Cardiovascular Research Laboratory, Division of Cardiology, Pulmonary Diseases and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany ,Department of Anaesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Matti Adam
- Clinic for Cardiology, University Hospital Cologne, Cologne, Germany
| | | | - Farhad Bakhtiary
- Heart Center Bonn, Clinic for Heard Surgery, University Hospital Bonn, Bonn, Germany
| | - Felix Jansen
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Georg Nickenig
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Eike Latz
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Sebastian Zimmer
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
2
|
Yang JH, Zhao Z, Niu W, Choi HP, Azadzoi KM. Formation of Double Stranded RNA Provokes Smooth Muscle Contractions and Structural Modifications in Bladder Ischemia. Res Rep Urol 2022; 14:399-414. [PMID: 36415310 PMCID: PMC9676006 DOI: 10.2147/rru.s388464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/11/2022] [Indexed: 08/30/2023] Open
Abstract
Purpose Growing evidence suggests that ischemia provokes detrusor overactivity and degenerative responses in the bladder. Underlying mechanisms appear to involve modification of smooth muscle contractile rudiments by hypoxia, redox, cellular stress and cell survival signaling. Downstream pathways of cellular stress and stress response molecules eliciting bladder dysfunction in ischemia remain largely elusive. Our goal was to define the role of double stranded RNA (dsRNA), a stress response molecule provoked by redox, in ischemia mediated bladder dysfunction. Methods A rat model of pelvic ischemia along with a cell culture hypoxia model were used to investigate the expression levels, functional consequences, structural aspects, and regulatory mechanisms of dsRNA in the bladder. Gene and protein expression were examined by reverse transcription polymerase chain reaction (RT-PCR), dot blot, and Western blotting, respectively. Tissue structure and function were assessed using histological staining and organ bath. Regulatory mechanisms were analyzed in cultured bladder smooth muscle cells. Results The data presented here provide the first evidence of the formation of dsRNA in the overactive bladder. dsRNA is a cellular stress response molecule that sensitizes smooth muscle and regulates inflammatory and degenerative rejoinders. Our data suggest that the production of dsRNA in the bladder is provoked by ischemia. Formation of dsRNA appears to augment bladder smooth muscle contractions and provoke fibrotic and apoptotic responses. Downstream actions of dsRNA in the bladder may involve upregulation of dsRNA-activated protein kinase R (PKR) and caspase-3, the executioner of apoptosis. Conclusion Activation of dsRNA/PKR pathway may play a role in sensitization of bladder smooth muscle cells to contractile stimuli, whereas dsRNA and caspase-3 crosstalk appear to modulate cellular stress and instigate degenerative responses in bladder ischemia. These observations suggest the role of dsRNA in bladder dysfunction and may open new perspectives to overcome overactive smooth muscle contractions and structural damage in the bladder.
Collapse
Affiliation(s)
- Jing-Hua Yang
- Department of Surgery, Boston University School of Medicine and Proteomics Laboratory, VA Boston Healthcare System, Boston, MA, USA
| | - Zuohui Zhao
- Department of Urology, Boston University School of Medicine, Boston, MA, USA
| | - Wanting Niu
- Research Department, VA Boston Healthcare System, Boston, MA, USA
| | - Han-Pil Choi
- Research Department, VA Boston Healthcare System, Boston, MA, USA
| | - Kazem M Azadzoi
- Departments of Urology and Pathology, VA Boston Healthcare System and Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
3
|
Guo S, Zhang E, Zhang B, Liu Q, Meng Z, Li Z, Wang C, Gong Z, Wu Y. Identification of Key Non-coding RNAs and Transcription Factors in Calcific Aortic Valve Disease. Front Cardiovasc Med 2022; 9:826744. [PMID: 35845040 PMCID: PMC9276990 DOI: 10.3389/fcvm.2022.826744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 06/01/2022] [Indexed: 11/25/2022] Open
Abstract
Background Calcific aortic valve disease (CAVD) is one of the most frequently occurring valvular heart diseases among the aging population. Currently, there is no known pharmacological treatment available to delay or reverse CAVD progression. The regulation of gene expression could contribute to the initiation, progression, and treatment of CAVD. Non-coding RNAs (ncRNAs) and transcription factors play essential regulatory roles in gene expression in CAVD; thus, further research is urgently needed. Materials and Methods The gene-expression profiles of GSE51472 and GSE12644 were obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) were identified in each dataset. A protein-protein-interaction (PPI) network of DEGs was then constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins database, and functional modules were analyzed with ClusterOne plugin in Cytoscape. Furthermore, Gene Ontology-functional annotation and Kyoto Encyclopedia of Genes and Genomes-pathway analysis were conducted for each functional module. Most crucially, ncRNAs and transcription factors acting on each functional module were separately identified using the RNAInter and TRRUST databases. The expression of predicted transcription factors and key genes was validated using GSE51472 and GSE12644. Furthermore, quantitative real-time PCR (qRT-PCR) experiments were performed to validate the differential expression of most promising candidates in human CAVD and control samples. Results Among 552 DEGs, 383 were upregulated and 169 were downregulated. In the PPI network, 15 functional modules involving 182 genes and proteins were identified. After hypergeometric testing, 45 ncRNAs and 33 transcription factors were obtained. Among the predicted transcription factors, CIITA, HIF1A, JUN, POU2F2, and STAT6 were differentially expressed in both the training and validation sets. In addition, we found that key genes, namely, CD2, CD86, CXCL8, FCGR3B, GZMB, ITGB2, LY86, MMP9, PPBP, and TYROBP were also differentially expressed in both the training and validation sets. Among the most promising candidates, differential expressions of ETS1, JUN, NFKB1, RELA, SP1, STAT1, ANCR, and LOC101927497 were identified via qRT-PCR experiments. Conclusion In this study, we identified functional modules with ncRNAs and transcription factors involved in CAVD pathogenesis. The current results suggest candidate molecules for further research on CAVD.
Collapse
|
4
|
Wang L, Zhou S, Liu Y, Li Y, Sun X. Bibliometric analysis of the inflammatory mechanism in aortic disease. Rev Cardiovasc Med 2022; 23:67. [PMID: 35229558 DOI: 10.31083/j.rcm2302067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/07/2021] [Accepted: 10/14/2021] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND In view of the key role of inflammation in the pathogenesis of aortic disease, we visually analyzed the research hotspots of inflammatory mechanism in aortic disease in this work through the method of bibliometrics from the Web of Science (WOS) Core database over the past three decades. METHODS A visual bibliometric network of research articles on inflammatory mechanisms in aortic disease was obtained from VOSviewer and Citespace based on the WOS Core Collection. RESULTS A total of 1278 documents from January 1990 to February 2021 were selected for analysis. The United States and China had the highest percentage of articles, comprising 34.01% and 24.92% of articles worldwide, respectively. Harvard University has published the most articles in this field, followed by the University of Michigan and Huazhong University of Science and Technology. The top 3 research hotspots were atherosclerosis, oxidative stress, and macrophages. The journal with the most articles in this area was Arteriosclerosis Thrombosis and Vascular Biology, followed by Atherosclerosis and PLOS One. The research trend on inflammatory mechanisms in the aortic system has 5 distinct directions: (1) atherosclerosis, NF-κB, expression, smooth muscle cell, and oxidative stress; (2) coronary artery disease, C-reactive protein, risk factors, endothelial dysfunction, and aortic stenosis; (3) abdominal aortic aneurysm, matrix metalloproteinases, macrophage, and pathogenesis; (4) cholesterol, metabolism, low-density lipoprotein, gene expression, and a therosclerotic lesions; and (5) calcific aortic valve disease, interstitial cells, calcification, and stenosis. CONCLUSIONS Inflammatory mechanism research has shown a tendency to rise gradually in the aortic field. Numerous studies have explored the role of inflammatory responses in aortic disease, which may increase the risk of endothelial dysfunction (aortic fibrosis and stiffness) and induce plaque formation. Among them, NFκB activation, nitric-oxide synthase expression, and oxidative stress are particularly essential.
Collapse
Affiliation(s)
- Luchen Wang
- Aortic and Vascular Surgery Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| | - Sangyu Zhou
- Aortic and Vascular Surgery Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| | - Yanxiang Liu
- Aortic and Vascular Surgery Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| | - Yunfeng Li
- Aortic and Vascular Surgery Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
- Shandong University, Qilu Hospital, 250012 Jinan, Shandong, China
| | - Xiaogang Sun
- Aortic and Vascular Surgery Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| |
Collapse
|
5
|
Zhang P, The E, Luo Z, Zhai Y, Yao Q, Ao L, Fullerton DA, Xu D, Meng X. Pro-inflammatory mediators released by activated monocytes promote aortic valve fibrocalcific activity. Mol Med 2022; 28:5. [PMID: 35062861 PMCID: PMC8780233 DOI: 10.1186/s10020-022-00433-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 01/07/2022] [Indexed: 11/24/2022] Open
Abstract
Background Calcific aortic valve disease (CAVD) is the most prevalent heart valve disorder in the elderly. Valvular fibrocalcification is a characteristic pathological change. In diseased valves, monocyte accumulation is evident, and aortic valve interstitial cells (AVICs) display greater fibrogenic and osteogenic activities. However, the impact of activated monocytes on valular fibrocalcification remains unclear. We tested the hypothesis that pro-inflammatory mediators from activated monocytes elevate AVIC fibrogenic and osteogenic activities.
Methods and results Picro-sirius red staining and Alizarin red staining revealed collagen and calcium depositions in cultured human AVICs exposed to conditioned media derived from Pam3CSK4-stimulated monocytes (Pam3 CM). Pam3 CM up-regulated alkaline phosphatase (ALP), an osteogenic biomarker, and extracellular matrix proteins collagen I and matrix metalloproteinase-2 (MMP-2). ELISA analysis identified high levels of RANTES and TNF-α in Pam3 CM. Neutralizing RANTES in the Pam3 CM reduced its effect on collagen I and MMP-2 production in AVICs while neutralizing TNF-α attenuated the effect on AVIC ALP production. In addition, Pam3 CM induced NF-κB and JNK activation. While JNK mediated the effect of Pam3 CM on collagen I and MMP-2 production, NF-κB was critical for the effect of Pam3 CM on ALP production in AVICs. Conclusions This study demonstrates that activated monocytes elevate the fibrogenic and osteogenic activities in human AVICs through a paracrine mechanism. TNF-α and RANTES mediate the pro-fibrogenic effect of activated monocytes on AVICs through activation of JNK, and TNF-α also activates NF-κB to elevate AVIC osteogenic activity. The results suggest that infiltrated monocytes elevate AVIC fibrocalcific activity to promote CAVD progression.
Collapse
Affiliation(s)
- Peijian Zhang
- Department of Surgery, University of Colorado Denver, Aurora, CO, 80045, USA.,Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Erlinda The
- Department of Surgery, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Zichao Luo
- Department of Surgery, University of Colorado Denver, Aurora, CO, 80045, USA.,Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yufeng Zhai
- Department of Surgery, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Qingzhou Yao
- Department of Surgery, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Lihua Ao
- Department of Surgery, University of Colorado Denver, Aurora, CO, 80045, USA
| | - David A Fullerton
- Department of Surgery, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Dingli Xu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Xianzhong Meng
- Department of Surgery, University of Colorado Denver, Aurora, CO, 80045, USA.
| |
Collapse
|
6
|
Bayer AL, Alcaide P. MyD88: At the heart of inflammatory signaling and cardiovascular disease. J Mol Cell Cardiol 2021; 161:75-85. [PMID: 34371036 PMCID: PMC8629847 DOI: 10.1016/j.yjmcc.2021.08.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease is a leading cause of death worldwide and is associated with systemic inflammation. In depth study of the cell-specific signaling mechanisms mediating the inflammatory response is vital to improving anti-inflammatory therapies that reduce mortality and morbidity. Cellular damage in the cardiovascular system results in the release of damage associated molecular patterns (DAMPs), also known as "alarmins," which activate myeloid cells through the adaptor protein myeloid differentiation primary response 88 (MyD88). MyD88 is broadly expressed in most cell types of the immune and cardiovascular systems, and its role often differs in a cardiovascular disease context and cell specific manner. Herein we review what is known about MyD88 in the setting of a variety of cardiovascular diseases, discussing cell specific functions and the relative contributions of MyD88-dependent vs. independent alarmin triggered inflammatory signaling. The widespread involvement of these pathways in cardiovascular disease, and their largely unexplored complexity, sets the stage for future in depth mechanistic studies that may place MyD88 in both immune and non-immune cell types as an attractive target for therapeutic intervention in cardiovascular disease.
Collapse
Affiliation(s)
- Abraham L Bayer
- Department of Immunology, Tufts University School of Medicine. 136 Harrison Ave, Boston, MA 02111, United States of America.
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine. 136 Harrison Ave, Boston, MA 02111, United States of America.
| |
Collapse
|
7
|
Parra-Izquierdo I, Sánchez-Bayuela T, Castaños-Mollor I, López J, Gómez C, San Román JA, Sánchez Crespo M, García-Rodríguez C. Clinically used JAK inhibitor blunts dsRNA-induced inflammation and calcification in aortic valve interstitial cells. FEBS J 2021; 288:6528-6542. [PMID: 34009721 DOI: 10.1111/febs.16026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/31/2021] [Accepted: 05/17/2021] [Indexed: 01/25/2023]
Abstract
Calcific aortic valve disease (CAVD) is the most prevalent valvulopathy worldwide. Growing evidence supports a role for viral and cell-derived double-stranded (ds)-RNA in cardiovascular pathophysiology. Poly(I:C), a dsRNA surrogate, has been shown to induce inflammation, type I interferon (IFN) responses, and osteogenesis through Toll-like receptor 3 in aortic valve interstitial cells (VIC). Here, we aimed to determine whether IFN signaling via Janus kinase (JAK)/Signal transducers and activators of transcription (STAT) mediates dsRNA-induced responses in primary human VIC. Western blot, ELISA, qPCR, calcification, flow cytometry, and enzymatic assays were performed to evaluate the mechanisms of dsRNA-induced inflammation and calcification. Poly(I:C) triggered a type I IFN response characterized by IFN-regulatory factors gene upregulation, IFN-β secretion, and STAT1 activation. Additionally, Poly(I:C) promoted VIC inflammation via NF-κB and subsequent adhesion molecule expression, and cytokine secretion. Pretreatment with ruxolitinib, a clinically used JAK inhibitor, abrogated these responses. Moreover, Poly(I:C) promoted a pro-osteogenic phenotype and increased VIC calcification to a higher extent in cells from males. Inhibition of JAK with ruxolitinib or a type I IFN receptor blocking antibody blunted Poly(I:C)-induced calcification. Mechanistically, Poly(I:C) promoted VIC apoptosis in calcification medium, which was inhibited by ruxolitinib. Moreover, Poly(I:C) co-operated with IFN-γ to increase VIC calcification by synergistically activating extracellular signal-regulated kinases and hypoxia-inducible factor-1α pathways. In conclusion, JAK/STAT signaling mediates dsRNA-triggered inflammation, apoptosis, and calcification and may contribute to a positive autocrine loop in human VIC in the presence of IFN-γ. Blockade of dsRNA responses with JAK inhibitors may be a promising therapeutic avenue for CAVD.
Collapse
Affiliation(s)
- Iván Parra-Izquierdo
- Unidad de Excelencia Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Spain
| | - Tania Sánchez-Bayuela
- Unidad de Excelencia Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Spain
| | - Irene Castaños-Mollor
- Unidad de Excelencia Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Spain
| | - Javier López
- ICICOR, Hospital Clínico Universitario, Valladolid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Cristina Gómez
- Unidad de Excelencia Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Spain
| | - J Alberto San Román
- ICICOR, Hospital Clínico Universitario, Valladolid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Mariano Sánchez Crespo
- Unidad de Excelencia Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Spain
| | - Carmen García-Rodríguez
- Unidad de Excelencia Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
8
|
Wang J, Sun L, Nie Y, Duan S, Zhang T, Wang W, Ye RD, Hou S, Qian F. Protein Kinase C δ (PKCδ) Attenuates Bleomycin Induced Pulmonary Fibrosis via Inhibiting NF-κB Signaling Pathway. Front Physiol 2020; 11:367. [PMID: 32390869 PMCID: PMC7188947 DOI: 10.3389/fphys.2020.00367] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 03/30/2020] [Indexed: 12/21/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive and lethal interstitial lung disease characterized by consistent pulmonary inflammation. Although protein kinase C delta (PKCδ) is involved in broad scope cellular response, the role of PKCδ in IPF is complicated and has not been fully defined yet. Here, we reported that PKCδ deficiency (PKCδ-/-) aggravated bleomycin (BLM)-induced pulmonary fibrosis and inflammation. Upon challenge with BLM, the pulmonary capillary permeability, immune cell infiltration, inflammatory cytokine production, and collagen deposition were enhanced in PKCδ-/- mice compared to that in PKCδ+/+ mice. In response to poly(I:C) stimulation, PKCδ deficient macrophages displayed an increased production of IL-1β, IL-6, TNF-α, and IL-33, which were associated with an enhanced NF-κB activation. Furthermore, we found that PKCδ could directly bind to and phosphorylate A20, an inhibitory protein of NF-κB signal. These results suggested that PKCδ may inhibit the NF-κB signaling pathway via enhancing the stability and activity of A20, which in turn attenuates pulmonary fibrosis, suggesting that PKCδ is a promising target for treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Jun Wang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Sun
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Shixin Duan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Weiwei Wang
- College of Pharmacy and Chemistry, Dali University, Dali, China
| | - Richard D Ye
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Shangwei Hou
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Qian
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China.,Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, China
| |
Collapse
|
9
|
Yao Q, The E, Ao L, Zhai Y, Osterholt MK, Fullerton DA, Meng X. TLR4 Stimulation Promotes Human AVIC Fibrogenic Activity through Upregulation of Neurotrophin 3 Production. Int J Mol Sci 2020; 21:ijms21041276. [PMID: 32074942 PMCID: PMC7072994 DOI: 10.3390/ijms21041276] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is a chronic inflammatory disease that manifests as progressive valvular fibrosis and calcification. An inflammatory milieu in valvular tissue promotes fibrosis and calcification. Aortic valve interstitial cell (AVIC) proliferation and the over-production of the extracellular matrix (ECM) proteins contribute to valvular thickening. However, the mechanism underlying elevated AVIC fibrogenic activity remains unclear. Recently, we observed that AVICs from diseased aortic valves express higher levels of neurotrophin 3 (NT3) and that NT3 exerts pro-osteogenic and pro-fibrogenic effects on human AVICs. HYPOTHESIS Pro-inflammatory stimuli upregulate NT3 production in AVICs to promote fibrogenic activity in human aortic valves. METHODS AND RESULTS AVICs were isolated from normal human aortic valves and were treated with lipopolysaccharide (LPS, 0.20 µg/mL). LPS induced TLR4-dependent NT3 production. This effect of LPS was abolished by inhibition of the Akt and extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) pathways. The stimulation of TLR4 in human AVICs with LPS resulted in a greater proliferation rate and an upregulated production of matrix metallopeptidases-9 (MMP-9) and collagen III, as well as augmented collagen deposition. Recombinant NT3 promoted AVIC proliferation in a tropomyosin receptor kinase (Trk)-dependent fashion. The neutralization of NT3 or the inhibition of Trk suppressed LPS-induced AVIC fibrogenic activity. CONCLUSIONS The stimulation of TLR4 in human AVICs upregulates NT3 expression and promotes cell proliferation and collagen deposition. The NT3-Trk cascade plays a critical role in the TLR4-mediated elevation of fibrogenic activity in human AVICs. Upregulated NT3 production by endogenous TLR4 activators may contribute to aortic valve fibrosis associated with CAVD progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xianzhong Meng
- Correspondence: ; Tel.: +303-724-6303; Fax: +303-724-6330
| |
Collapse
|
10
|
Tang X, Pan L, Zhao S, Dai F, Chao M, Jiang H, Li X, Lin Z, Huang Z, Meng G, Wang C, Chen C, Liu J, Wang X, Ferro A, Wang H, Chen H, Gao Y, Lu Q, Xie L, Han Y, Ji Y. SNO-MLP (S-Nitrosylation of Muscle LIM Protein) Facilitates Myocardial Hypertrophy Through TLR3 (Toll-Like Receptor 3)-Mediated RIP3 (Receptor-Interacting Protein Kinase 3) and NLRP3 (NOD-Like Receptor Pyrin Domain Containing 3) Inflammasome Activation. Circulation 2020; 141:984-1000. [PMID: 31902237 DOI: 10.1161/circulationaha.119.042336] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND S-nitrosylation (SNO), a prototypic redox-based posttranslational modification, is involved in the pathogenesis of cardiovascular disease. The aim of this study was to determine the role of SNO of MLP (muscle LIM protein) in myocardial hypertrophy, as well as the mechanism by which SNO-MLP modulates hypertrophic growth in response to pressure overload. METHODS Myocardial samples from patients and animal models exhibiting myocardial hypertrophy were examined for SNO-MLP level using biotin-switch methods. SNO sites were further identified through liquid chromatography-tandem mass spectrometry. Denitrosylation of MLP by the mutation of nitrosylation sites or overexpression of S-nitrosoglutathione reductase was used to analyze the contribution of SNO-MLP in myocardial hypertrophy. Downstream effectors of SNO-MLP were screened through mass spectrometry and confirmed by coimmunoprecipitation. Recruitment of TLR3 (Toll-like receptor 3) by SNO-MLP in myocardial hypertrophy was examined in TLR3 small interfering RNA-transfected neonatal rat cardiomyocytes and in a TLR3 knockout mouse model. RESULTS SNO-MLP level was significantly higher in hypertrophic myocardium from patients and in spontaneously hypertensive rats and mice subjected to transverse aortic constriction. The level of SNO-MLP also increased in angiotensin II- or phenylephrine-treated neonatal rat cardiomyocytes. S-nitrosylated site of MLP at cysteine 79 was identified by liquid chromatography-tandem mass spectrometry and confirmed in neonatal rat cardiomyocytes. Mutation of cysteine 79 significantly reduced hypertrophic growth in angiotensin II- or phenylephrine-treated neonatal rat cardiomyocytes and transverse aortic constriction mice. Reducing SNO-MLP level by overexpression of S-nitrosoglutathione reductase greatly attenuated myocardial hypertrophy. Mechanistically, SNO-MLP stimulated TLR3 binding to MLP in response to hypertrophic stimuli, and disrupted this interaction by downregulating TLR3-attenuated myocardial hypertrophy. SNO-MLP also increased the complex formation between TLR3 and RIP3 (receptor-interacting protein kinase 3). This interaction in turn induced NLRP3 (nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3) inflammasome activation, thereby promoting the development of myocardial hypertrophy. CONCLUSIONS Our findings revealed a key role of SNO-MLP in myocardial hypertrophy and demonstrated TLR3-mediated RIP3 and NLRP3 inflammasome activation as the downstream signaling pathway, which may represent a therapeutic target for myocardial hypertrophy and heart failure.
Collapse
Affiliation(s)
- Xin Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Lihong Pan
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Shuang Zhao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Feiyue Dai
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Menglin Chao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Hong Jiang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Xuesong Li
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Zhe Lin
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Zhengrong Huang
- Department of Cardiology, the First Affiliated Hospital of Xiamen University, China (Z.H.)
| | - Guoliang Meng
- Nanjing Medical University, Nanjing, China (G.M.).,Department of Pharmacology, School of Pharmacy, Nantong University, China (G.M.)
| | - Chun Wang
- Department of Geriatrics, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, China (C.W.)
| | - Chan Chen
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China (C.C., J.L.)
| | - Jin Liu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China (C.C., J.L.)
| | - Xin Wang
- Faculty of Biology, Medicine and Health, the University of Manchester, United Kingdom (X.W.)
| | - Albert Ferro
- Cardiovascular Clinical Pharmacology, British Heart Foundation Centre of Research Excellence, Cardiovascular Division, King's College London, United Kingdom (A.F.)
| | - Hong Wang
- Department of Pharmacology, Lewis Kats School of Medicine, Temple University, Philadelphia, PA (H.W.)
| | - Hongshan Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Yuanqing Gao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Qiulun Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Liping Xie
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.)
| | - Yi Han
- Department of Geriatrics, First Affiliated Hospital of Nanjing Medical University, China (Y.H.)
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Medical University, Nanjing, China (X.T., L.P., S.Z., F.D., M.C., H.J., X.L., Z.L., H.C., Y.G., Q.L., L.X., Y.J.).,State Key Laboratory of Reproductive Medicine (Y.J.)
| |
Collapse
|
11
|
Sastre B, García-García ML, Calvo C, Casas I, Rodrigo-Muñoz JM, Cañas JA, Mora I, del Pozo V. Immune recovery following bronchiolitis is linked to a drop in cytokine and LTC4 levels. Pediatr Res 2020; 87:581-587. [PMID: 31600771 PMCID: PMC7086521 DOI: 10.1038/s41390-019-0606-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/25/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Bronchiolitis is the main cause of hospitalization of children younger than 1 year; however, the immune mechanism of bronchiolitis is not completely understood. The aim of this study was to analyze the recovery of immune response after a bronchiolitis episode. METHODS Forty-nine infants hospitalized with bronchiolitis diagnosis were enrolled. Nasopharyngeal aspirates (NPAs) were processed. Twenty-seven pro-inflammatory biomarkers linked to innate immunity, inflammation, and epithelial damage, as well as nitrites and lipid mediators, were evaluated in the NPA supernatant by ELISA (enzyme-linked immunosorbent assay) and Luminex. Also, 11 genes were analyzed in NPA cells by quantitative PCR. RESULTS A widespread statistically significant decline of multiple pro-inflammatory parameters and cytokines were detected in the recovery period after respiratory infection: interferon-α2 (IFNα2), IFNγ, interleukin-10 (IL-10), IL-1β, IL-8, IFN-γ-inducible protein-10, vascular endothelial growth factor, monocyte chemoattractant protein-1, macrophage inflammatory protein-1α (MIP-1α), and MIP-1β. Supporting these results, a decreased nuclear factor-κB gene expression was observed (P = 0.0116). A significant diminution of cysteinyl leukotriene C4 (LTC4) soluble levels (P = 0.0319) and cyclooxygenase-2 (COX-2) gene expression were observed in the recovery sample. In children classified by post-bronchiolitis wheezing, LTC4 remains elevated in the NPA supernatant. CONCLUSIONS After bronchiolitis, cytokines and biomarkers linked to innate immune response in NPA decrease significantly in the recovery period accompanied by a drop in LTC4 levels; however, this reduction was lower in infants with post-bronchiolitis wheezing.
Collapse
Affiliation(s)
- Beatriz Sastre
- grid.419651.eDepartment of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain ,0000 0000 9314 1427grid.413448.eCIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - María Luz García-García
- 0000 0001 0635 4617grid.411361.0Pediatrics Department, Severo Ochoa Hospital, Leganés, Spain ,Translational Research Network in Pediatric Infectious Diseases (RITIP), Madrid, Spain ,0000 0001 2323 8386grid.464699.0Alfonso X El Sabio University, Madrid, Spain
| | - Cristina Calvo
- Translational Research Network in Pediatric Infectious Diseases (RITIP), Madrid, Spain ,0000 0001 2323 8386grid.464699.0Alfonso X El Sabio University, Madrid, Spain ,0000 0000 8970 9163grid.81821.32Pediatric Infectious Diseases Department, Hospital Universitario La Paz, Madrid, Spain ,Fundación IdiPaz, Madrid, Spain ,TEDDY Network (European Network of Excellence for Pediatric Clinical Research), Madrid, Spain
| | - Inmaculada Casas
- Translational Research Network in Pediatric Infectious Diseases (RITIP), Madrid, Spain ,Respiratory Virus and Influenza Unit, National Microbiology Center (ISCIII), Madrid, Spain
| | - José Manuel Rodrigo-Muñoz
- grid.419651.eDepartment of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain ,0000 0000 9314 1427grid.413448.eCIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - José Antonio Cañas
- grid.419651.eDepartment of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain ,0000 0000 9314 1427grid.413448.eCIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Inés Mora
- grid.419651.eDepartment of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Victoria del Pozo
- grid.419651.eDepartment of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain ,0000 0000 9314 1427grid.413448.eCIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|
12
|
Raddatz MA, Madhur MS, Merryman WD. Adaptive immune cells in calcific aortic valve disease. Am J Physiol Heart Circ Physiol 2019; 317:H141-H155. [PMID: 31050556 DOI: 10.1152/ajpheart.00100.2019] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Calcific aortic valve disease (CAVD) is highly prevalent and has no pharmaceutical treatment. Surgical replacement of the aortic valve has proved effective in advanced disease but is costly, time limited, and in many cases not optimal for elderly patients. This has driven an increasing interest in noninvasive therapies for patients with CAVD. Adaptive immune cell signaling in the aortic valve has shown potential as a target for such a therapy. Up to 15% of cells in the healthy aortic valve are hematopoietic in origin, and these cells, which include macrophages, T lymphocytes, and B lymphocytes, are increased further in calcified specimens. Additionally, cytokine signaling has been shown to play a causative role in aortic valve calcification both in vitro and in vivo. This review summarizes the physiological presence of hematopoietic cells in the valve, innate and adaptive immune cell infiltration in disease states, and the cytokine signaling pathways that play a significant role in CAVD pathophysiology and may prove to be pharmaceutical targets for this disease in the near future.
Collapse
Affiliation(s)
- Michael A Raddatz
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee.,Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Meena S Madhur
- Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee.,Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee.,Division of Clinical Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee
| |
Collapse
|
13
|
Yan X, Xie B, Wu G, Hu J, Wang D, Cai X, Li J. Interleukin-37: The Effect of Anti-Inflammatory Response in Human Coronary Artery Endothelial Cells. Mediators Inflamm 2019; 2019:2650590. [PMID: 30728750 PMCID: PMC6341264 DOI: 10.1155/2019/2650590] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/07/2018] [Accepted: 11/06/2018] [Indexed: 02/05/2023] Open
Abstract
Interleukin-37 (IL-37) is unique in the IL-1 family since it broadly suppresses innate immunity and elevates in humans with inflammatory and autoimmune diseases. IL-37 shows definite groups and transcripts for human IL37 gene, but it is still not completely understood the effect and mechanisms of inflammatory response in endothelial cells. It is well accepted that endothelial dysfunction caused by inflammation is a key initiating event in atherosclerotic plaque formation, which leads to the occurrence and development of the cardiovascular adverse events in clinical since the inflammatory responses of endothelial cells could induce and enhance the deposition of extensive lipid and the formation of atherosclerotic plaque in the intima. Thus, it is essential to investigate the role and potential mechanisms in endothelial inflammatory response to prevent the formation and development of many cardiovascular diseases including atherosclerosis. So far, the recent studies have revealed that IL-37 is able to inhibit inflammatory response by suppressing the TLR2-NF-κB-ICAM-1 pathway intracellularly in human coronary artery endothelial cells (HCAECs). Further, the role of IL-37 may be related to the IL-18 pathway extracellularly and involved in the adhesion and transmigration of neutrophils in HCAECs.
Collapse
Affiliation(s)
- Xianfeng Yan
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Bin Xie
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Guihai Wu
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jing Hu
- Department of Cardiology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, China
| | - Di Wang
- Department of Dermatovenereology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Xiangna Cai
- Department of Plastic Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jilin Li
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| |
Collapse
|
14
|
Ding X, Liang Y, Peng W, Li R, Lin H, Zhang Y, Lu D. Intracellular TLR22 acts as an inflammation equalizer via suppression of NF-κB and selective activation of MAPK pathway in fish. FISH & SHELLFISH IMMUNOLOGY 2018; 72:646-657. [PMID: 29175443 DOI: 10.1016/j.fsi.2017.11.042] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 06/07/2023]
Abstract
TLR22, a typical member of the fish-specific TLRs, is a crucial sensor in virally triggered innate immune signalling retained from natural selection. To elucidate the role of the TLR22-specific signalling cascade mechanism, we provide evidence that the double-stranded (ds) RNA-sensor TLR22 positively regulates the ERK pathway and negatively regulates the JNK, p38 MAP kinase and NF-κB pathway. Here, we show that TLR22 restrains NF-κB activation and IFN (interferon) β and AP-1 (activator protein-1) promoter binding (impairing "primary response" genes (TNF and IL-1)), induces "secondary response" genes (IL-12 and IL-6) and mediates the irregular expression of inflammatory genes. Therefore, TLR22 promotes ERK phosphorylation but impairs the JNK and p38 MAP kinases and IκB phosphorylation. Additionally, TLR22 controls the excessive generation of reactive oxygen species (ROS) to avoid damaging the organism. The specific kinetics of TLR22 depends on its distinct cellular localization. We demonstrate that TLR22 is an intracellular receptor localized in the endosome, and the TLR22-TIR domain is the functional structure inducing the signalling cascade post-viral replication in the body. As mentioned above, our data reveal a novel mechanism whereby TLR22-induced positive adjustment and negative regulation evolved independently to avoid harmful and inappropriate inflammatory responses.
Collapse
Affiliation(s)
- Xu Ding
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China; Department of Obstetrics and Gynecology, First Hospital Affiliated to the Fourth Military Medical University, Xi'an, 710032, China
| | - Yaosi Liang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Wan Peng
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Ruozhu Li
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yong Zhang
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Danqi Lu
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, College of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.
| |
Collapse
|
15
|
Pham PH, Tong WWL, Misk E, Jones G, Lumsden JS, Bols NC. Atlantic salmon endothelial cells from the heart were more susceptible than fibroblasts from the bulbus arteriosus to four RNA viruses but protected from two viruses by dsRNA pretreatment. FISH & SHELLFISH IMMUNOLOGY 2017; 70:214-227. [PMID: 28882807 DOI: 10.1016/j.fsi.2017.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/23/2017] [Accepted: 09/01/2017] [Indexed: 06/07/2023]
Abstract
Heart diseases caused by viruses are major causes of Atlantic salmon aquaculture loss. Two Atlantic salmon cardiovascular cell lines, an endothelial cell line (ASHe) from the heart and a fibroblast cell line (BAASf) from the bulbus arteriosus, were evaluated for their response to four fish viruses, CSV, IPNV, VHSV IVa and VHSV IVb, and the innate immune agonist, double-stranded RNA mimic poly IC. All four viruses caused cytopathic effects in ASHe and BAASf. However, ASHe was more susceptible to all four viruses than BAASf. When comparing between the viruses, ASHe cells were found to be moderately susceptible to CSV and VHSV IVb, but highly susceptible to IPNV and VHSV IVa induced cell death. All four viruses were capable of propagating in the ASHe cell line, leading to increases in virus titre over time. In BAASf, CSV and IPNV produced more than one log increase in titre from initial infection, but VHSV IVb and IVa did not. When looking at the antiviral response of both cell lines, Mx proteins were induced in ASHe and BAASf by poly IC. All four viruses induced Mx proteins in BAASf, while only CSV and VHSV IVb induced Mx proteins in ASHe. IPNV and VHSV IVa suppressed Mx proteins expression in ASHe. Pretreatment of ASHe with poly IC to allow for Mx proteins accumulation protected the culture from subsequent infections with IPNV and VHSV IVa, resulting in delayed cell death, reduced virus titres and reduced viral proteins expression. These data suggest that endothelial cells potentially can serve as points of infections for viruses in the heart and that two of the four viruses, IPNV and VHSV IVa, have mechanisms to avoid or downregulate antiviral responses in ASHe cells. Furthermore, the high susceptibility of the ASHe cell line to IPNV and VHSV IVa can make it a useful tool for studying antiviral compounds against these viruses and for general detection of fish viruses.
Collapse
Affiliation(s)
- Phuc H Pham
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Winnie W L Tong
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Ehab Misk
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Ginny Jones
- Elanco Canada Limited, Aqua Business R&D, Victoria, PEI, Canada
| | - John S Lumsden
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada; St. George's University, True Blue, Grenada
| | - Niels C Bols
- Department of Biology, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
16
|
Zhan Q, Zeng Q, Song R, Zhai Y, Xu D, Fullerton DA, Dinarello CA, Meng X. IL-37 suppresses MyD88-mediated inflammatory responses in human aortic valve interstitial cells. Mol Med 2017; 23:83-91. [PMID: 28362018 DOI: 10.2119/molmed.2017.00022] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/21/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is common among the elderly, and aortic valve interstitial cells (AVICs) exhibit unique inflammatory and osteogenic responses to pro-inflammatory stimulation which play an important role in valvular fibrosis and calcification. Thus, suppression of AVIC pro-inflammatory response may have therapeutic utility for prevention of CAVD progression. Interleukin (IL)-37, an anti-inflammatory cytokine, reduces tissue inflammation. OBJECTIVE This study was to test the hypothesis that IL-37 suppresses human AVIC inflammatory responses to Toll-like receptor (TLR) agonists. METHODS AND RESULTS Human AVICs were exposed to Pam3CSK4, poly(I:C) and lipopolysaccharide, respectively, in the presence and absence of recombinant human IL-37. Stimulation of TLR4 increased the production of intercellular adhesion molecule-1, IL-6, IL-8 and monocyte chemoattractant protein-1. Knockdown of myeloid differentiation factor 88 (MyD88) or TIR-domain-containing adaptor inducing interferon-β (TRIF) differentially affected inflammatory mediator production following TLR4 stimulation. IL-37 reduced the production of these inflammatory mediators induced by TLR4. Moreover, knockdown of IL-37 enhanced the induction of these mediators by TLR4. IL-37 also suppressed inflammatory mediator production induced by the MyD88-dependent TLR2, but had no effect on the inflammatory responses to the TRIF-dependent TLR3. Furthermore, IL-37 inhibited NF-κB activation induced by TLR2 or TLR4 through a mechanism dependent of IL-18 receptor α-chain. CONCLUSION Activation of TLR2, TLR3 or TLR4 up-regulates the production of inflammatory mediators in human AVICs. IL-37 suppresses MyD88-mediated responses to reduce inflammatory mediator production following stimulation of TLR2 and TLR4. This anti-inflammatory cytokine may be useful for suppression of aortic valve inflammation elicited by MyD88-dependent TLR signaling.
Collapse
Affiliation(s)
- Qiong Zhan
- Department of Surgery, University of Colorado Denver, Aurora, CO 80045.,Department of Cardiology, Nanfang hospital, Southern Medical University, Guangzhou 510515, China
| | - Qingchun Zeng
- Department of Surgery, University of Colorado Denver, Aurora, CO 80045.,Department of Cardiology, Nanfang hospital, Southern Medical University, Guangzhou 510515, China
| | - Rui Song
- Department of Surgery, University of Colorado Denver, Aurora, CO 80045
| | - Yufeng Zhai
- Department of Surgery, University of Colorado Denver, Aurora, CO 80045
| | - Dingli Xu
- Department of Cardiology, Nanfang hospital, Southern Medical University, Guangzhou 510515, China
| | - David A Fullerton
- Department of Surgery, University of Colorado Denver, Aurora, CO 80045
| | | | - Xianzhong Meng
- Department of Surgery, University of Colorado Denver, Aurora, CO 80045
| |
Collapse
|