1
|
Xian G, Huang R, Hu D, Xu M, Chen Y, Ren H, Xu D, Zeng Q. Interleukin-37 attenuates aortic valve lesions by inhibiting N6-methyladenosine-mediated interleukin-1 receptor-associated kinase M degradation. Cardiovasc Res 2025; 121:492-506. [PMID: 39913240 DOI: 10.1093/cvr/cvaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/11/2024] [Accepted: 11/12/2024] [Indexed: 02/26/2025] Open
Abstract
AIMS Calcific aortic valve disease (CAVD) has become an increasingly important global medical problem without effective pharmacological intervention. Accumulating evidence indicates that aortic valve calcification is driven by inflammation. Interleukin-1 receptor-associated kinase M (IRAK-M) is a well-known negative regulator of inflammation, but its role in CAVD remains unclear. METHODS AND RESULTS Here, we stimulated aortic valve interstitial cells (AVICs) with low-dose lipopolysaccharide (LPS) to mimic the inflammatory response in aortic valve calcification and observed the expression pattern of IRAK-M. Furthermore, we generated IRAK-M-/- mice to explore the effect of IRAK-M deficiency on the aortic valve in vivo. Additionally, overexpression and knockdown experiments were performed to verify the role of IRAK-M in AVICs. Methylated RNA immunoprecipitation-quantitative polymerase chain reaction was used to detect the N6-methyladenosine (m6A) level of IRAK-M, and recombinant interleukin (IL)-37-treated AVICs were used to determine the regulatory relationship between IL-37 and IRAK-M. We found that IRAK-M expression was upregulated in the early stages of inflammation as part of a negative feedback mechanism to modulate the immune response. However, persistent inflammation increased overall m6A levels, ultimately leading to reduced IRAK-M expression. In vivo, IRAK-M-/- mice exhibited a propensity for aortic valve thickening and calcification. Overexpression and knockdown experiments showed that IRAK-M inhibited inflammation and osteogenic responses in AVICs. In addition, IL-37 restored IRAK-M expression by inhibiting m6A-mediated IRAK-M degradation to suppress inflammation and aortic valve calcification. CONCLUSION Our findings confirm that inflammation and epigenetic modifications synergistically regulate IRAK-M expression. Moreover, IRAK-M represents a potential target for mitigating aortic valve calcification. Meanwhile, IL-37 exhibited inhibitory effects on CAVD development both in vivo and in vitro, giving us hope that CAVD can be treated with drugs rather than surgery.
Collapse
Affiliation(s)
- Gaopeng Xian
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Rong Huang
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Dongtu Hu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Minhui Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Yangchao Chen
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Hao Ren
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Northern Guangzhou Ave, Guangzhou 510515, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, Guangzhou 510515, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| |
Collapse
|
2
|
Li J, Luo R, Fu Y, Liu S, Dong Q, Sun Y, Tian X, Zhu Y, Wang P, Guo L, Lu Q, Ye C, Fu S, Qiu Y. Baicalin, Amoxicillin, and Probenecid Provide Protection in Mice Against Glaesserella parasuis Challenge. Biomolecules 2025; 15:507. [PMID: 40305201 PMCID: PMC12024593 DOI: 10.3390/biom15040507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 05/02/2025] Open
Abstract
Glaesserella parasuis (G. parasuis) causes Glässer's disease and systemic inflammatory responses in the host. The currently available therapies have limited efficacy and fail to achieve a balance between anti-inflammatory and antibacterial effects. In this study, we investigated the effects of baicalin, amoxicillin, and probenecid on blood biochemical parameters, routine blood indicators, survival rate, bacterial burden, and pathological tissue damage in G. parasuis-challenged mice. Treatment with baicalin, amoxicillin, and probenecid significantly modified the blood biochemical parameters and routine blood test indicators, increased the survival rate, attenuated the bacterial burden, and alleviated pathological tissue damage in G. parasuis-challenged mice. Treatment with baicalin, amoxicillin, and probenecid also increased the number of CD3+, CD3+CD4+, and CD3+CD8+ T cells as measured by flow cytometry, and restored the intensity of the CD3, CD4, and CD8 protein expression in the blood vessels of G. parasuis-challenged mice by immunohistochemistry. These compounds reduced interleukin 1β (IL-1β), IL-18, tumor necrosis factor alpha (TNF-α), and high mobility group box 1 protein (HMGB1) expression in the spleen of G. parasuis-challenged mice. Furthermore, baicalin, amoxicillin, and probenecid inhibited activation of the family pyrin domain containing 3 (NLRP3) inflammasome and apoptosis in the spleen of G. parasuis-challenged mice. This study showed the important roles of baicalin, amoxicillin, and probenecid in the modulation of the inflammatory response of Glässer's disease. The findings might provide new strategies for combination therapy using antibiotics and anti-inflammatory drugs to control G. parasuis infection.
Collapse
Affiliation(s)
- Jingyang Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Ronghui Luo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Yunjian Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Siyu Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Qiaoli Dong
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Yamin Sun
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Xinyue Tian
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Yi Zhu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Peiyi Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Ling Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Qirong Lu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China; (J.L.); (R.L.); (Y.F.); (S.L.); (Q.D.); (Y.S.); (X.T.); (Y.Z.); (P.W.); (L.G.); (Q.L.); (C.Y.)
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, China
| |
Collapse
|
3
|
The E, Zhai Y, Yao Q, Ao L, Fullerton DA, Meng X. Molecular Interaction of Soluble Klotho with FGF23 in the Pathobiology of Aortic Valve Lesions Induced by Chronic Kidney Disease. Int J Biol Sci 2024; 20:3412-3425. [PMID: 38993571 PMCID: PMC11234222 DOI: 10.7150/ijbs.92447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 05/03/2024] [Indexed: 07/13/2024] Open
Abstract
Chronic kidney disease (CKD) is linked to greater prevalence and rapid progression of calcific aortic valve disease (CAVD) characterized by valvular leaflet fibrosis and calcification. Fibroblast growth factor 23 (FGF23) level is elevated, and anti-aging protein Klotho is reduced in CKD patients. However, the roles of FGF23 and Klotho in the mechanism of aortic valve fibrosis and calcification remain unclear. We hypothesized that FGF23 mediates CKD-induced CAVD by enhancing aortic valve interstitial cell (AVIC) fibrosis and calcification, while soluble Klotho inhibits FGF23 effect. Methods and Results: In an old mouse model of CKD, kidney damages were accompanied by aortic valve thickening and calcification. FGF23 levels in plasma and aortic valve were increased, while Klotho levels were decreased. Recombinant FGF23 elevated the inflammatory, fibrogenic, and osteogenic activities in AVICs. Neutralizing antibody or shRNA targeting FGF23 suppressed the pathobiological activities in AVICs from valves affected by CAVD. FGF23 exerts its effects on AVICs via FGF receptor (FGFR)/Yes-associated protein (YAP) signaling, and inhibition of FGFR/YAP reduced FGF23's potency in AVICs. Recombinant Klotho downregulated the pathobiological activities in AVICs exposed to FGF23. Incubation of FGF23 with Klotho formed complexes and decreased FGF23's potency. Further, treatment of CKD mice with recombinant Klotho attenuated aortic valve lesions. Conclusion: This study demonstrates that CKD induces FGF23 accumulation, Klotho insufficiency and aortic valve lesions in old mice. FGF23 upregulates the inflammatory, fibrogenic and osteogenic activities in AVICs via the FGFR/YAP signaling pathway. Soluble Klotho suppresses FGF23 effect through molecular interaction and is capable of mitigating CKD-induced CAVD.
Collapse
Affiliation(s)
| | | | | | | | | | - Xianzhong Meng
- Departments of Surgery and Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| |
Collapse
|
4
|
Yu C, Zhang Y, Yang L, Aikebaier M, Shan S, Zha Q, Yang K. Identification of pyroptosis-associated genes with diagnostic value in calcific aortic valve disease. Front Cardiovasc Med 2024; 11:1340199. [PMID: 38333413 PMCID: PMC10850341 DOI: 10.3389/fcvm.2024.1340199] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Background Calcific aortic valve disease (CAVD) is one of the most prevalent valvular diseases and is the second most common cause for cardiac surgery. However, the mechanism of CAVD remains unclear. This study aimed to investigate the role of pyroptosis-related genes in CAVD by performing comprehensive bioinformatics analysis. Methods Three microarray datasets (GSE51472, GSE12644 and GSE83453) and one RNA sequencing dataset (GSE153555) were obtained from the Gene Expression Omnibus (GEO) database. Pyroptosis-related differentially expressed genes (DEGs) were identified between the calcified and the normal valve samples. LASSO regression and random forest (RF) machine learning analyses were performed to identify pyroptosis-related DEGs with diagnostic value. A diagnostic model was constructed with the diagnostic candidate pyroptosis-related DEGs. Receiver operating characteristic (ROC) curve analysis was performed to estimate the diagnostic performances of the diagnostic model and the individual diagnostic candidate genes in the training and validation cohorts. CIBERSORT analysis was performed to estimate the differences in the infiltration of the immune cell types. Pearson correlation analysis was used to investigate associations between the diagnostic biomarkers and the immune cell types. Immunohistochemistry was used to validate protein concentration. Results We identified 805 DEGs, including 319 down-regulated genes and 486 up-regulated genes. These DEGs were mainly enriched in pathways related to the inflammatory responses. Subsequently, we identified 17 pyroptosis-related DEGs by comparing the 805 DEGs with the 223 pyroptosis-related genes. LASSO regression and RF algorithm analyses identified three CAVD diagnostic candidate genes (TREM1, TNFRSF11B, and PGF), which were significantly upregulated in the CAVD tissue samples. A diagnostic model was constructed with these 3 diagnostic candidate genes. The diagnostic model and the 3 diagnostic candidate genes showed good diagnostic performances with AUC values >0.75 in both the training and the validation cohorts based on the ROC curve analyses. CIBERSORT analyses demonstrated positive correlation between the proportion of M0 macrophages in the valve tissues and the expression levels of TREM1, TNFRSF11B, and PGF. Conclusion Three pyroptosis-related genes (TREM1, TNFRSF11B and PGF) were identified as diagnostic biomarkers for CAVD. These pyroptosis genes and the pro-inflammatory microenvironment in the calcified valve tissues are potential therapeutic targets for alleviating CAVD.
Collapse
Affiliation(s)
- Chenxi Yu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yifeng Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ling Yang
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Mirenuer Aikebaier
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shuyao Shan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qing Zha
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ke Yang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Conte M, Poggio P, Monti M, Petraglia L, Cabaro S, Bruzzese D, Comentale G, Caruso A, Grimaldi M, Zampella E, Gencarelli A, Cervasio MR, Cozzolino F, Monaco V, Myasoedova V, Valerio V, Ferro A, Insabato L, Bellino M, Galasso G, Graziani F, Pucci P, Formisano P, Pilato E, Cuocolo A, Perrone Filardi P, Leosco D, Parisi V. Isolated Valve Amyloid Deposition in Aortic Stenosis: Potential Clinical and Pathophysiological Relevance. Int J Mol Sci 2024; 25:1171. [PMID: 38256243 PMCID: PMC10815971 DOI: 10.3390/ijms25021171] [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: 11/27/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
Amyloid deposition within stenotic aortic valves (AVs) also appears frequent in the absence of cardiac amyloidosis, but its clinical and pathophysiological relevance has not been investigated. We will elucidate the rate of isolated AV amyloid deposition and its potential clinical and pathophysiological significance in aortic stenosis (AS). In 130 patients without systemic and/or cardiac amyloidosis, we collected the explanted AVs during cardiac surgery: 57 patients with calcific AS and 73 patients with AV insufficiency (41 with AV sclerosis and 32 without, who were used as controls). Amyloid deposition was found in 21 AS valves (37%), 4 sclerotic AVs (10%), and none of the controls. Patients with and without isolated AV amyloid deposition had similar clinical and echocardiographic characteristics and survival rates. Isolated AV amyloid deposition was associated with higher degrees of AV fibrosis (p = 0.0082) and calcification (p < 0.0001). Immunohistochemistry analysis suggested serum amyloid A1 (SAA1), in addition to transthyretin (TTR), as the protein possibly involved in AV amyloid deposition. Circulating SAA1 levels were within the normal range in all groups, and no difference was observed in AS patients with and without AV amyloid deposition. In vitro, AV interstitial cells (VICs) were stimulated with interleukin (IL)-1β which induced increased SAA1-mRNA both in the control VICs (+6.4 ± 0.5, p = 0.02) and the AS VICs (+7.6 ± 0.5, p = 0.008). In conclusion, isolated AV amyloid deposition is frequent in the context of AS, but it does not appear to have potential clinical relevance. Conversely, amyloid deposition within AV leaflets, probably promoted by local inflammation, could play a role in AS pathophysiology.
Collapse
Affiliation(s)
- Maddalena Conte
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini, 5, 80131 Naples, Italy
- Casa di Cura San Michele, 81024 Caserta, Italy; (A.C.)
| | - Paolo Poggio
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (P.P.)
| | - Maria Monti
- Dipartimento di Scienze Chimiche, University of Naples Federico II, 5, 80131 Naples, Italy (F.C.); (V.M.)
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Laura Petraglia
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini, 5, 80131 Naples, Italy
| | - Serena Cabaro
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini, 5, 80131 Naples, Italy
| | - Dario Bruzzese
- Department of Public Health, University of Naples Federico II, 5, 80131 Naples, Italy
| | - Giuseppe Comentale
- Department of Advanced Biomedical Science, University of Naples Federico II, 5, 80131 Naples, Italy (A.C.)
| | | | | | - Emilia Zampella
- Department of Advanced Biomedical Science, University of Naples Federico II, 5, 80131 Naples, Italy (A.C.)
| | - Annarita Gencarelli
- Department of Advanced Biomedical Science, University of Naples Federico II, 5, 80131 Naples, Italy (A.C.)
| | - Maria Rosaria Cervasio
- Department of Advanced Biomedical Science, University of Naples Federico II, 5, 80131 Naples, Italy (A.C.)
| | - Flora Cozzolino
- Dipartimento di Scienze Chimiche, University of Naples Federico II, 5, 80131 Naples, Italy (F.C.); (V.M.)
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Vittoria Monaco
- Dipartimento di Scienze Chimiche, University of Naples Federico II, 5, 80131 Naples, Italy (F.C.); (V.M.)
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | | | | | - Adele Ferro
- Institute of Biostructure and Bioimaging, CNR, 80145 Naples, Italy
| | - Luigi Insabato
- Department of Advanced Biomedical Science, University of Naples Federico II, 5, 80131 Naples, Italy (A.C.)
| | - Michele Bellino
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, 84081 Salerno, Italy
| | - Gennaro Galasso
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, 84081 Salerno, Italy
| | - Francesca Graziani
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Pietro Pucci
- Dipartimento di Scienze Chimiche, University of Naples Federico II, 5, 80131 Naples, Italy (F.C.); (V.M.)
| | - Pietro Formisano
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini, 5, 80131 Naples, Italy
| | - Emanuele Pilato
- Department of Advanced Biomedical Science, University of Naples Federico II, 5, 80131 Naples, Italy (A.C.)
| | - Alberto Cuocolo
- Department of Advanced Biomedical Science, University of Naples Federico II, 5, 80131 Naples, Italy (A.C.)
| | - Pasquale Perrone Filardi
- Department of Advanced Biomedical Science, University of Naples Federico II, 5, 80131 Naples, Italy (A.C.)
| | - Dario Leosco
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini, 5, 80131 Naples, Italy
| | - Valentina Parisi
- Department of Translational Medical Sciences, University of Naples Federico II, Via S. Pansini, 5, 80131 Naples, Italy
| |
Collapse
|
6
|
Bańka P, Wybraniec M, Bochenek T, Gruchlik B, Burchacka A, Swinarew A, Mizia-Stec K. Influence of Aortic Valve Stenosis and Wall Shear Stress on Platelets Function. J Clin Med 2023; 12:6301. [PMID: 37834945 PMCID: PMC10573628 DOI: 10.3390/jcm12196301] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Aortic valve stenosis (AS) is a common heart valve disease in the elderly population, and its pathogenesis remains an interesting area of research. The degeneration of the aortic valve leaflets gradually progresses to valve sclerosis. The advanced phase is marked by the presence of extracellular fibrosis and calcification. Turbulent, accelerated blood flow generated by the stenotic valve causes excessive damage to the aortic wall. Elevated shear stress due to AS leads to the degradation of high-molecular weight multimers of von Willebrand factor, which may involve bleeding in the mucosal tissues. Conversely, elevated shear stress has been associated with the release of thrombin and the activation of platelets, even in individuals with acquired von Willebrand syndrome. Moreover, turbulent blood flow in the aorta may activate the endothelium and promote platelet adhesion and activation on the aortic valve surface. Platelets release a wide range of mediators, including lysophosphatidic acid, which have pro-osteogenic effects in AS. All of these interactions result in blood coagulation, fibrinolysis, and the hemostatic process. This review summarizes the current knowledge on high shear stress-induced hemostatic disorders, the influence of AS on platelets and antiplatelet therapy.
Collapse
Affiliation(s)
- Paweł Bańka
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| | - Maciej Wybraniec
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| | - Tomasz Bochenek
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| | - Bartosz Gruchlik
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| | - Aleksandra Burchacka
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| | - Andrzej Swinarew
- Faculty of Science and Technology, University of Silesia in Katowice, 40-007 Katowice, Poland
- Department of Swimming and Water Rescue, Institute of Sport Science, The Jerzy Kukuczka Academy of Physical Education, 40-065 Katowice, Poland
| | - Katarzyna Mizia-Stec
- First Department of Cardiology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| |
Collapse
|
7
|
Bouhamida E, Morciano G, Pedriali G, Ramaccini D, Tremoli E, Giorgi C, Pinton P, Patergnani S. The Complex Relationship between Hypoxia Signaling, Mitochondrial Dysfunction and Inflammation in Calcific Aortic Valve Disease: Insights from the Molecular Mechanisms to Therapeutic Approaches. Int J Mol Sci 2023; 24:11105. [PMID: 37446282 DOI: 10.3390/ijms241311105] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Calcific aortic valve stenosis (CAVS) is among the most common causes of cardiovascular mortality in an aging population worldwide. The pathomechanisms of CAVS are such a complex and multifactorial process that researchers are still making progress to understand its physiopathology as well as the complex players involved in CAVS pathogenesis. Currently, there is no successful and effective treatment to prevent or slow down the disease. Surgical and transcatheter valve replacement represents the only option available for treating CAVS. Insufficient oxygen availability (hypoxia) has a critical role in the pathogenesis of almost all CVDs. This process is orchestrated by the hallmark transcription factor, hypoxia-inducible factor 1 alpha subunit (HIF-1α), which plays a pivotal role in regulating various target hypoxic genes and metabolic adaptations. Recent studies have shown a great deal of interest in understanding the contribution of HIF-1α in the pathogenesis of CAVS. However, it is deeply intertwined with other major contributors, including sustained inflammation and mitochondrial impairments, which are attributed primarily to CAVS. The present review aims to cover the latest understanding of the complex interplay effect of hypoxia signaling pathways, mitochondrial dysfunction, and inflammation in CAVS. We propose further hypotheses and interconnections on the complexity of these impacts in a perspective of better understanding the pathophysiology. These interplays will be examined considering recent studies that shall help us better dissect the molecular mechanism to enable the design and development of potential future therapeutic approaches that can prevent or slow down CAVS processes.
Collapse
Affiliation(s)
- Esmaa Bouhamida
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Giampaolo Morciano
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Gaia Pedriali
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Daniela Ramaccini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Elena Tremoli
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Simone Patergnani
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
8
|
Rani A, Toor D. Plausible Role of NLRP3 Inflammasome and Associated Cytokines in Pathogenesis of Rheumatic Heart Disease. Crit Rev Immunol 2023; 43:1-14. [PMID: 37824373 DOI: 10.1615/critrevimmunol.2023049463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Rheumatic heart disease (RHD) is a post-streptococcal sequela caused by Streptococcus pyogenes. The global burden of disease is high among people with low socio-economic status, with significant cases emerging every year despite global eradication efforts. The current treatment includes antibiotic therapies to target strep throat and rheumatic fever and valve replacement strategies as a corrective measure for chronic RHD patients. Valvular damage and valve calcification are considered to be the end-stage processes of the disease resulting from impairment of the endothelial arrangement due to immune infiltration. This immune infiltration is mediated by a cascade of events involving NLRP3 inflammasome activation. NLRP3 inflammasome is activated by wide range of stimuli including bacterial cell wall components like M proteins and leukocidal toxins like nicotinamide dehydrogenase (NADase) and streptolysin O (SLO) and these play a major role in sustaining the virulence of Streptococcus pyogenes and progression of RHD. In this review, we are discussing NLRP3 inflammasome and its plausible role in the pathogenesis of RHD by exploiting the host-pathogen interaction mainly focusing on the NLRP3 inflammasome-mediated cytokines IL-1β and IL-18. Different therapeutic approaches involving NLRP3 inflammasome inactivation, caspase-1 inhibition, and blockade of IL-1β and IL-18 are discussed in this review and may be promising for treating RHD patients.
Collapse
Affiliation(s)
- Aishwarya Rani
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Sector-125, Noida, 201313, Uttar Pradesh, India
| | - Devinder Toor
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Sector-125, Noida, 201313, Uttar Pradesh, India
| |
Collapse
|
9
|
Li X, Guo D, Zhou W, Hu Y, Zhou H, Chen Y. The Potential Prognostic, Diagnostic and Therapeutic Targets for Recurrent Arrhythmias in Patients with Coronary Restenosis and Reocclusions After Coronary Stenting. Curr Pharm Des 2022; 28:3500-3512. [PMID: 36424794 DOI: 10.2174/1381612829666221124110445] [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: 04/07/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND The interplay of oxidative stress, proinflammatory microparticles, and proinflammatory cytokines in recurrent arrhythmias is unknown in elderly patients with coronary restenosis and reocclusions after coronary stenting. OBJECTIVE This research sought to investigate the potential diagnostic and therapeutic targets for recurrent arrhythmias in patients with coronary restenosis and reocclusions after coronary stenting. METHODS We examined whether oxidative stress, proinflammatory microparticles, and proinflammatory cytokines could have effects that lead to recurrent arrhythmias in elderly patients with coronary restenosis and reocclusions. We measured the levels of malondialdehyde (MDA), CD31 + endothelial microparticle (CD31 EMP), CD62E + endothelial microparticle (CD62E + EMP), high-sensitivity C-reactive protein (hs-CRP), interleukin- 1β (IL-1β), interleukin-6 (IL-6), interleukin-8 (IL-8) and tumor necrosis factor-α (TNF-α), as well as oxidized low-density lipoprotein (OX-LDL), and assessed the effects of relationship between oxidative stress, proinflammatory microparticles, and proinflammatory cytokines on recurrent atrial and ventricular arrhythmias in elderly patients with coronary restenosis and reocclusions after coronary stenting. RESULTS The levels of CD31 + EMP, CD62E + EMP, MDA, hs-CRP, IL-1β, IL-6, IL-8, TNF-α and OX-LDL were found to be increased significantly in coronary restenosis + recurrent atrial arrhythmia group compared to without coronary restenosis and coronary restenosis + without recurrent atrial arrhythmia groups, respectively (P < 0.001). Patients in the coronary reocclusion + recurrent ventricular arrhythmia group also exhibited significantly increased levels of CD31 + EMP, CD62E + EMP, MDA, hs-CRP, IL-1β, IL-6, IL-8, TNF-α and OXLDL compared to without coronary reocclusion and coronary reocclusion + without recurrent ventricular arrhythmia groups, respectively (P < 0.001). CONCLUSION Proinflammatory microparticles, proinflammatory cytokines, and oxidative stress might act as potential targets for recurrent arrhythmias in patients with coronary restenosis and reocclusions after coronary stenting.
Collapse
Affiliation(s)
- Xia Li
- Department of Geriatrics, Second People's Hospital of Huai'an, Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huai'an 223002, China
| | - Dianxuan Guo
- Department of Geriatrics, Second People's Hospital of Huai'an, Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huai'an 223002, China
| | - Wenhang Zhou
- Department of Geriatrics, Second People's Hospital of Huai'an, Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huai'an 223002, China
| | - Youdong Hu
- Department of Geriatrics, Second People's Hospital of Huai'an, Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huai'an 223002, China
| | - Hualan Zhou
- Department of Geriatrics, Second People's Hospital of Huai'an, Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huai'an 223002, China
| | - Ying Chen
- Department of Geriatrics, Second People's Hospital of Huai'an, Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huai'an 223002, China
| |
Collapse
|
10
|
Saucedo-Orozco H, Vargas-Barron J, Márquez-Velazco R, Farjat-Pasos JI, Martinez-Zavala KS, Jiménez-Rojas V, Criales-Vera SA, Arias-Godínez JA, Fuentevilla-Alvarez G, Guarner-Lans V, Perez-Torres I, Melendez-Ramirez G, Sanchez Perez TE, Soto ME. Bioprosthesis in aortic valve replacement: long-term inflammatory response and functionality. Open Heart 2022; 9:openhrt-2022-002065. [PMID: 35926961 PMCID: PMC9358956 DOI: 10.1136/openhrt-2022-002065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Abstract
Background The evaluation of long-term inflammatory response and function in postoperative patients with aortic valve replacement (AVR) deserves special analysis because it is important to try to prevent reoperation and improve durability and functionality of the prostheses. It is our objective Methods In this study, we included a cohort of patients with aortic valve damage treated by AVR with mechanical prosthesis, bio prosthesis and we included a control group. Results We found that IL-4 and osteopontin levels were higher in patients with mechanical vs biological prostheses (p=0.01 and p=0.04, respectively), osteoprotegerin (OPG) levels were decreased (p=0.01), women had lower levels of ET-1 and IL-6, (p=0.02) (p=0.04), respectively. Patients older than 60 years had decreased levels of IL-1ß p<0.001) and a higher concentration of IL-4 p<0.05). IL-1ß, OPG and TNFα were higher in patients with less than 5 years of evolution vs more than 10 years (p=0.004, p=0.02 and p=0.03, respectively). Factors such as age, gender, prosthetic and elevated IL-1B and ET-1 levels are associated with valve dysfunction prosthetic. These results indicate that the inflammatory involvement present prior to valve replacement may be perpetuated by various factors in the long term. Conclusions The findings provide us with the opportunity to effectively treat patients with AVR in the postoperative period, which could prolong the functionality of the bio prostheses. Trial registration number NCT04557345.
Collapse
Affiliation(s)
- Huitzilihuitl Saucedo-Orozco
- Cardioneumology, Instituto Nacional de Cardiologia Ignacio Chavez, Ciudad de Mexico, Mexico.,Speciality Hospital, National Medical Center "La Raza", Cardioneumology, Instituto Mexicano del Seguro Social, Ciudad de Mexico, Mexico
| | - Jesus Vargas-Barron
- Pharmacology, Instituto Nacional de Cardiologia Ignacio Chavez, CDMX, Mexico
| | - Ricardo Márquez-Velazco
- Department of Immunology, Instituto Nacional de Cardiologia Ignacio Chavez, Mexico City, Mexico
| | - Julio Iván Farjat-Pasos
- Interventional Cardiology, Instituto Nacional de Cardiologia Ignacio Chavez, Tlalpan, Mexico
| | | | | | | | | | | | | | - Israel Perez-Torres
- Cardiovascular Biomedicine, Instituto Nacional de Cardiologia Ignacio Chavez, CDMX, Mexico
| | | | | | - Maria Elena Soto
- Immunology, Instituto Nacional de Cardiologia Ignacio Chavez, CDMX, Mexico .,Cardiovascular Line, Hospital ABC, Mexico City, Mexico
| |
Collapse
|
11
|
Ferrari S, Pesce M. The Complex Interplay of Inflammation, Metabolism, Epigenetics, and Sex in Calcific Disease of the Aortic Valve. Front Cardiovasc Med 2022; 8:791646. [PMID: 35071359 PMCID: PMC8770423 DOI: 10.3389/fcvm.2021.791646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/30/2021] [Indexed: 12/20/2022] Open
Abstract
Calcification of the aortic valve is one of the most rapidly increasing pathologies in the aging population worldwide. Traditionally associated to cardiovascular risk conditions, this pathology is still relatively unaddressed on a molecular/cellular standpoint and there are no available treatments to retard its progression unless valve substitution. In this review, we will describe some of the most involved inflammatory players, the metabolic changes that may be responsible of epigenetic modifications and the gender-related differences in the onset of the disease. A better understanding of these aspects and their integration into a unique pathophysiology context is relevant to improve current therapies and patients management.
Collapse
Affiliation(s)
- Silvia Ferrari
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| |
Collapse
|
12
|
Innate immune cells in the pathophysiology of calcific aortic valve disease: lessons to be learned from atherosclerotic cardiovascular disease? Basic Res Cardiol 2022; 117:28. [PMID: 35581364 PMCID: PMC9114076 DOI: 10.1007/s00395-022-00935-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 01/31/2023]
Abstract
Calcific aortic valve disease (CAVD) is the most common valvular disease in the developed world with currently no effective pharmacological treatment available. CAVD results from a complex, multifactorial process, in which valvular inflammation and fibro-calcific remodelling lead to valve thickening and cardiac outflow obstruction. The exact underlying pathophysiology of CAVD is still not fully understood, yet the development of CAVD shows many similarities with the pathophysiology of atherosclerotic cardiovascular disease (ASCVD), such as coronary artery disease. Innate immune cells play a crucial role in ASCVD and might also play a pivotal role in the development of CAVD. This review summarizes the current knowledge on the role of innate immune cells, both in the circulation and in the aortic valve, in the development of CAVD and the similarities and differences with ASCVD. Trained immunity and clonal haematopoiesis of indeterminate potential are proposed as novel immunological mechanisms that possibly contribute to the pathophysiology of CAVD and new possible treatment targets are discussed.
Collapse
|
13
|
Gatera VA, Lesmana R, Musfiroh I, Judistiani RTD, Setiabudiawan B, Abdulah R. Vitamin D Inhibits Lipopolysaccharide (LPS)-Induced Inflammation in A549 Cells by Downregulating Inflammatory Cytokines. Med Sci Monit Basic Res 2021; 27:e931481. [PMID: 34103463 PMCID: PMC8202123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/11/2021] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Studies have shown that lung inflammation affects lung function, with life-threatening results. Vitamin D may play an important role in inhibiting inflammatory cytokines. Vitamin D deficiency is related to several lung problems, including respiratory distress syndrome, alveolar inflammation, epithelial damage, and hypoxia. Few studies have evaluated the benefits of vitamin D in preventing inflammation in alveolar cells. MATERIAL AND METHODS We developed a cell inflammation model induced by lipopolysaccharide (LPS) treatment. The effects of vitamin D on LPS-induced inflammation in A549 cells were examined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and the anti-inflammatory mechanism of vitamin D was evaluated using western blot analysis. RESULTS Our results indicated that vitamin D promoted A549 cell survival following LPS-induced inflammation by downregulating nuclear factor nuclear factor kappa light chain enhancer of activated B cells, tumor necrosis factor-alpha, interleukin (IL)-1ß, IL-6, and IL-12. CONCLUSIONS Our results indicated that vitamin D has the potential to manage lung inflammation, although further studies are needed.
Collapse
Affiliation(s)
- Vesara A. Gatera
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Department of Pharmacy, Faculty of Health, Singaperbangsa University, Karawang, Indonesia
| | - Ronny Lesmana
- Division of Physiology, Department of Biomedical Science, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Division of Biological Activity, Central Laboratory, Universitas Padjadjaran, Bandung, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
| | - Ida Musfiroh
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | | | - Budi Setiabudiawan
- Department of Child Health, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Rizky Abdulah
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
14
|
Noonin C, Thongboonkerd V. Exosome-inflammasome crosstalk and their roles in inflammatory responses. Am J Cancer Res 2021; 11:4436-4451. [PMID: 33754070 PMCID: PMC7977448 DOI: 10.7150/thno.54004] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammasome is a complex of multiple proteins found in cytoplasm of the cells activated by infectious and/or non-infectious stimuli. This complex involves caspase-1 activation, leading to unconventional secretion of interleukin-1β (IL-1β) and IL-18 and inflammatory cascade. Exosome is the nanoscale membrane-bound extracellular vesicle that plays significant roles in intercellular communications by carrying bioactive molecules, e.g., proteins, RNAs, microRNAs (miRNAs), DNAs, from one cell to the others. In this review, we provide the update information on the crosstalk between exosome and inflammasome and their roles in inflammatory responses. The effects of inflammasome activation on exosomal secretion are summarized. On the other hand, the (dual) effects of exosomes on inhibiting and promoting inflammasome activation are discussed. Finally, perspectives on therapeutic roles of exosomes in human diseases and future direction of the research on exosome-inflammasome crosstalk are provided.
Collapse
|
15
|
Jarrett MJ, Houk AK, McCuistion PE, Weyant MJ, Reece TB, Meng X, Fullerton DA. Wnt Signaling Mediates Pro-Fibrogenic Activity in Human Aortic Valve Interstitial Cells. Ann Thorac Surg 2020; 112:519-525. [PMID: 33189669 DOI: 10.1016/j.athoracsur.2020.08.068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 07/27/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Proinflammatory activation of toll-like receptor-4 (TLR4) drives phenotypic changes in aortic valve interstitial cells (AVICs) and produces a fibrogenic phenotype that mediates valvular fibrosis and contributes to aortic stenosis. Prior work identified upregulated Wnt signaling in AVICs taken from valves affected by aortic stenosis. Our purpose was to determine the contribution of Wnt signaling to TLR4-dependent fibrogenic activity in isolated human AVICs. METHODS Human AVICs were isolated from hearts explanted for cardiac transplantation (N = 4). To test whether Wnt signaling contributed to TLR4-dependent fibrogenic activity, AVICs were treated with Wnt inhibitor (Dkk1) prior to TLR4 activation (LPS) and fibrogenic markers assessed. To determine the mediator of TLR4-to-Wnt signaling, expression of the key Wnt ligand, Wnt3a, was assessed after TLR4 activation and neutralizing antibodies confirmed the identity of the mediator. Fibrogenic activity was assessed after AVICs were treated with recombinant Wnt3a. Statistics were by analysis of variance (P < .05). RESULTS TLR4 activation upregulated in vitro collagen deposition, type IV collagen and MMP2 expression, and Dkk1 inhibited these responses (P < .05). Expression of Wnt3a was upregulated after TLR4 activation (P < .05). Anti-Wnt3a neutralizing antibodies abrogated TLR4-dependent type IV collagen and MMP2 expression (P < .05). Wnt3a upregulated type IV collagen and MMP2 expression independent of TLR4 activation (P < .05). CONCLUSIONS This study found that TLR4-dependent fibrogenic activity was mediated through Wnt signaling. The mediator of profibrogenic TLR4-to-Wnt signaling was a key Wnt ligand, Wnt3a. The abrogation of TLR4-induced fibrogenic activity in human AVICs by Wnt blockade illustrates a potential therapeutic role for Wnt inhibition in treatment and/or prevention of aortic stenosis.
Collapse
Affiliation(s)
- Michael J Jarrett
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado.
| | - Anna K Houk
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Peyton E McCuistion
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Michael J Weyant
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - T Brett Reece
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Xianzhong Meng
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - David A Fullerton
- Department of Surgery, Division of Cardiothoracic Surgery, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
16
|
Nakamura Y, Aihara R, Iwata H, Kuwayama T, Shirasuna K. IL1B triggers inflammatory cytokine production in bovine oviduct epithelial cells and induces neutrophil accumulation via CCL2. Am J Reprod Immunol 2020; 85:e13365. [PMID: 33099841 DOI: 10.1111/aji.13365] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/13/2020] [Accepted: 10/21/2020] [Indexed: 12/16/2022] Open
Abstract
PROBLEM The oviduct is essential for reproduction. We previously showed that oviduct epithelial cells (OECs) isolated from aged cows expressed higher levels of inflammatory cytokines, including interleukin (IL) 1A and IL1B. In addition, aging is associated with tissue dysfunction and cellular senescence via a senescence-associated secretory phenotype (SASP) and immune cell accumulation. We investigated whether IL1A or IL1B causes SASP production, cellular senescence, and inflammatory responses in bovine OECs. METHOD OF STUDY The OECs were isolated from bovine oviducts from young (mean 50.3 months) and aged cows (mean 157.0 months) and cultured. RESULTS Treatment with IL1A or IL1B induced SASP production (IL8, IL6, TNFA, and CCL2) and mRNA expression of cell adhesion molecules in bovine OECs, but both IL1s did not induce cellular senescence in OECs and migration of polymorphonuclear neutrophils (PMNs). Cultured medium of OECs treated with IL1s, especially IL1B, dramatically induced PMN migration. Treatment with the CCL2 inhibitor, but not IL8 or its receptor CXCR2 inhibitors, significantly reduced immune cell migration in IL1B-treated OEC-cultured medium. Treatment with IL1B increased PMN adhesion to OECs, resulting in further SASP production in OECs due to a PMN-OEC interaction. CONCLUSION We suggest that senescence-associated IL1s cause SASP production in bovine OECs and CCL2 induced by IL1B is essential for the migration of immune cells to OECs. Specifically, IL1B regulates PMN migration and adhesion to bovine OECs, and PMNs accelerate inflammatory cytokine production from bovine OECs via a direct interaction. These phenomena may contribute to chronic oviductal inflammation, resulting in subfertility.
Collapse
Affiliation(s)
- Yuki Nakamura
- Laboratory of Animal Reproduction, Department of Agriculture, Tokyo University of Agriculture, Atsugi, Japan
| | - Riho Aihara
- Laboratory of Animal Reproduction, Department of Agriculture, Tokyo University of Agriculture, Atsugi, Japan
| | - Hisataka Iwata
- Laboratory of Animal Reproduction, Department of Agriculture, Tokyo University of Agriculture, Atsugi, Japan
| | - Takehito Kuwayama
- Laboratory of Animal Reproduction, Department of Agriculture, Tokyo University of Agriculture, Atsugi, Japan
| | - Koumei Shirasuna
- Laboratory of Animal Reproduction, Department of Agriculture, Tokyo University of Agriculture, Atsugi, Japan
| |
Collapse
|
17
|
Alushi B, Curini L, Christopher MR, Grubitzch H, Landmesser U, Amedei A, Lauten A. Calcific Aortic Valve Disease-Natural History and Future Therapeutic Strategies. Front Pharmacol 2020; 11:685. [PMID: 32477143 PMCID: PMC7237871 DOI: 10.3389/fphar.2020.00685] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 04/27/2020] [Indexed: 12/20/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the most frequent heart valve disorder. It is characterized by an active remodeling process accompanied with valve mineralization, that results in a progressive aortic valve narrowing, significant restriction of the valvular area, and impairment of blood flow.The pathophysiology of CAVD is a multifaceted process, involving genetic factors, chronic inflammation, lipid deposition, and valve mineralization. Mineralization is strictly related to the inflammatory process in which both, innate, and adaptive immunity are involved. The underlying pathophysiological pathways that go from inflammation to calcification and, finally lead to severe stenosis, remain, however, incompletely understood. Histopathological studies are limited to patients with severe CAVD and no samples are available for longitudinal studies of disease progression. Therefore, alternative routes should be explored to investigate the pathogenesis and progression of CAVD.Recently, increasing evidence suggests that epigenetic markers such as non-coding RNAs are implicated in the landscape of phenotypical changes occurring in CAVD. Furthermore, the microbiome, an essential player in several diseases, including the cardiovascular ones, has recently been linked to the inflammation process occurring in CAVD. In the present review, we analyze and discuss the CAVD pathophysiology and future therapeutic strategies, focusing on the real and putative role of inflammation, calcification, and microbiome.
Collapse
Affiliation(s)
- Brunilda Alushi
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Department of General and Interventional Cardiology, Helios Klinikum Erfurt, Erfurt, Germany
| | - Lavinia Curini
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Mary Roxana Christopher
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Herko Grubitzch
- Berlin Institute of Health, Berlin, Germany
- Department of Cardiology, German Heart Centre Berlin (DHZB), Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
- Sod of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Florence, Italy
| | - Alexander Lauten
- Department of Cardiology, Charite´ Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Department of General and Interventional Cardiology, Helios Klinikum Erfurt, Erfurt, Germany
| |
Collapse
|
18
|
Singh S, Torzewski M. Fibroblasts and Their Pathological Functions in the Fibrosis of Aortic Valve Sclerosis and Atherosclerosis. Biomolecules 2019; 9:biom9090472. [PMID: 31510085 PMCID: PMC6769553 DOI: 10.3390/biom9090472] [Citation(s) in RCA: 48] [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: 07/11/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases, such as atherosclerosis and aortic valve sclerosis (AVS) are driven by inflammation induced by a variety of stimuli, including low-density lipoproteins (LDL), reactive oxygen species (ROS), infections, mechanical stress, and chemical insults. Fibrosis is the process of compensating for tissue injury caused by chronic inflammation. Fibrosis is initially beneficial and maintains extracellular homeostasis. However, in the case of AVS and atherosclerosis, persistently active resident fibroblasts, myofibroblasts, and smooth muscle cells (SMCs) perpetually remodel the extracellular matrix under the control of autocrine and paracrine signaling from the immune cells. Myofibroblasts also produce pro-fibrotic factors, such as transforming growth factor-β1 (TGF-β1), angiotensin II (Ang II), and interleukin-1 (IL-1), which allow them to assist in the activation and migration of resident immune cells. Post wound repair, these cells undergo apoptosis or become senescent; however, in the presence of unresolved inflammation and persistence signaling for myofibroblast activation, the tissue homeostasis is disturbed, leading to excessive extracellular matrix (ECM) secretion, disorganized ECM, and thickening of the affected tissue. Accumulating evidence suggests that diverse mechanisms drive fibrosis in cardiovascular pathologies, and it is crucial to understand the impact and contribution of the various mechanisms for the control of fibrosis before the onset of a severe pathological consequence.
Collapse
Affiliation(s)
- Savita Singh
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology and University of Tuebingen, 70376 Stuttgart, Germany.
| | - Michael Torzewski
- Department of Laboratory Medicine and Hospital Hygiene, Robert-Bosch-Hospital, 70376 Stuttgart, Germany.
| |
Collapse
|
19
|
Development of calcific aortic valve disease: Do we know enough for new clinical trials? J Mol Cell Cardiol 2019; 132:189-209. [PMID: 31136747 DOI: 10.1016/j.yjmcc.2019.05.016] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/11/2019] [Accepted: 05/19/2019] [Indexed: 12/19/2022]
Abstract
Calcific aortic valve disease (CAVD), previously thought to represent a passive degeneration of the valvular extracellular matrix (VECM), is now regarded as an intricate multistage disorder with sequential yet intertangled and interacting underlying processes. Endothelial dysfunction and injury, initiated by disturbed blood flow and metabolic disorders, lead to the deposition of low-density lipoprotein cholesterol in the VECM further provoking macrophage infiltration, oxidative stress, and release of pro-inflammatory cytokines. Such changes in the valvular homeostasis induce differentiation of normally quiescent valvular interstitial cells (VICs) into synthetically active myofibroblasts producing excessive quantities of the VECM and proteins responsible for its remodeling. As a result of constantly ongoing degradation and re-deposition, VECM becomes disorganised and rigid, additionally potentiating myofibroblastic differentiation of VICs and worsening adaptation of the valve to the blood flow. Moreover, disrupted and excessively vascularised VECM is susceptible to the dystrophic calcification caused by calcium and phosphate precipitating on damaged collagen fibers and concurrently accompanied by osteogenic differentiation of VICs. Being combined, passive calcification and biomineralisation synergistically induce ossification of the aortic valve ultimately resulting in its mechanical incompetence requiring surgical replacement. Unfortunately, multiple attempts have failed to find an efficient conservative treatment of CAVD; however, therapeutic regimens and clinical settings have also been far from the optimal. In this review, we focused on interactions and transitions between aforementioned mechanisms demarcating ascending stages of CAVD, suggesting a predisposing condition (bicuspid aortic valve) and drug combination (lipid-lowering drugs combined with angiotensin II antagonists and cytokine inhibitors) for the further testing in both preclinical and clinical trials.
Collapse
|
20
|
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: 57] [Impact Index Per Article: 9.5] [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
|
21
|
Lee SH, Choi JH. Involvement of inflammatory responses in the early development of calcific aortic valve disease: lessons from statin therapy. Anim Cells Syst (Seoul) 2018; 22:390-399. [PMID: 30533261 PMCID: PMC6282465 DOI: 10.1080/19768354.2018.1528175] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 08/09/2018] [Accepted: 08/10/2018] [Indexed: 12/15/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the most common degenerative heart valve disease. Among the many risk factors for this disease are age, hypercholesterolemia, hypertension, smoking, type-2 diabetes, rheumatic fever, and chronic kidney disease. Since many of these overlap with risk factors for atherosclerosis, the molecular and cellular mechanisms of CAVD development have been presumed to be similar to those for atherogenesis. Thus, attempts have been made to evaluate the therapeutic efficacy of statins, representative anti-atherosclerosis drugs with lipid-lowering and anti-inflammatory effects, against CAVD. Unfortunately, statins have shown little or no effect on CAVD development. But some reports suggest that statins may prevent or reduce the development of early stage CAVD in which having calcification is absent or minimal. These results suggest that therapeutic approaches should differ according to the stage of disease, and that a precise understanding of the mechanism of aortic valve calcification is required to identify novel therapeutic targets for advanced CAVD. Given the involvement of inflammatory processes in the development and progression of CAVD, current therapeutic approaches for chronic inflammatory cardiovascular disease like atherosclerosis may help to prevent or minimize the early development of CAVD. In this review, we focus on several inflammatory cellular and molecular components involved in CAVD that might be considered drug targets for preventing CAVD.
Collapse
Affiliation(s)
- Seung Hyun Lee
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Jae-Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
22
|
Rutkovskiy A, Malashicheva A, Sullivan G, Bogdanova M, Kostareva A, Stensløkken KO, Fiane A, Vaage J. Valve Interstitial Cells: The Key to Understanding the Pathophysiology of Heart Valve Calcification. J Am Heart Assoc 2017; 6:e006339. [PMID: 28912209 PMCID: PMC5634284 DOI: 10.1161/jaha.117.006339] [Citation(s) in RCA: 228] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Arkady Rutkovskiy
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Norway
- Centre for Heart Failure Research, University of Oslo, Norway
- Department of Emergency Medicine and Intensive Care, Oslo University Hospital, Oslo, Norway
- Division of Medicine, Akershus University Hospital, Lørenskog, Norway
- ITMO University, St. Petersburg, Russia
| | - Anna Malashicheva
- Almazov National Medical Research Centre, St. Petersburg, Russia
- ITMO University, St. Petersburg, Russia
| | - Gareth Sullivan
- Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Norway
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Norway
- Institute of Immunology, Oslo University Hospital, Oslo, Norway
- Norwegian Center for Stem Cell Research, Oslo, Norway
| | - Maria Bogdanova
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Anna Kostareva
- Almazov National Medical Research Centre, St. Petersburg, Russia
- ITMO University, St. Petersburg, Russia
| | - Kåre-Olav Stensløkken
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Norway
- Centre for Heart Failure Research, University of Oslo, Norway
| | - Arnt Fiane
- Institute of Clinical Medicine, University of Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Jarle Vaage
- Institute of Clinical Medicine, University of Oslo, Norway
- Department of Emergency Medicine and Intensive Care, Oslo University Hospital, Oslo, Norway
- ITMO University, St. Petersburg, Russia
| |
Collapse
|
23
|
Takahashi Y, Matsutani N, Dejima H, Nakayama T, Uehara H, Kawamura M. Nuclear factor-kappa B influences early phase of compensatory lung growth after pneumonectomy in mice. J Biomed Sci 2017; 24:41. [PMID: 28679393 PMCID: PMC5499001 DOI: 10.1186/s12929-017-0350-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 06/27/2017] [Indexed: 02/08/2023] Open
Abstract
Background Compensatory lung growth (CLG) is a well-established lung regeneration model. However, the sequential mechanisms, including unknown molecular triggers or regulators, remain unclear. Nuclear factor- kappa B (NF-κB) is known to be essential for inflammation and tissue regeneration; therefore, we investigated the role of NF-κB in CLG. Methods C57BL/6 J mice underwent either a left pneumonectomy or a thoracotomy (n = 77). Gene microarray analysis was performed to detect genes that were upregulated at 12 h after pneumonectomy. NF-κB protein expression was examined by immunohistochemistry and Western blot. To investigate the influence of NF-κB on CLG, either an NF-κB inhibitor SN50 or saline was administered following pneumonectomy and the degree of CLG was evaluated in each group by measuring the lung dry weight index (LDWI) and the mean linear intercept. Results Gene microarray analysis identified 11 genes that were significantly but transiently increased at 12 h after pneumonectomy. Among the 11 genes, NF-κB was selected based on its reported functions. Western blot analysis showed that NF-κB protein expression after pneumonectomy was significantly higher at 12 h compared to 48 h. Additionally, NF-κB protein expression at 12 h after pneumonectomy was significantly higher than at both 12 and 48 h after thoracotomy (p < 0.029 for all). NF-κB protein expression, evaluated through immunohistochemistry, was expressed mainly in type 2 alveolar epithelial cells and was significant increased 12 h after pneumonectomy compared to 48 h after pneumonectomy and both 12 and 48 h after thoracotomy (p < 0.001 for all). SN50 administration following pneumonectomy induced a significant decrease in NF-κB expression (p = 0.004) and LDWI compared to the vehicle administration (p = 0.009). Conclusions This is the first report demonstrating that NF-κB signaling may play a key role in CLG. Given its pathway is crucial in tissue regeneration of various organs, NF-κB may shed light on identification of molecular triggers or clinically usable key regulators of CLG.
Collapse
Affiliation(s)
- Yusuke Takahashi
- Department of General Thoracic Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan.
| | - Noriyuki Matsutani
- Department of General Thoracic Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Hitoshi Dejima
- Department of General Thoracic Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Takashi Nakayama
- Department of General Thoracic Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Hirofumi Uehara
- Department of General Thoracic Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| | - Masafumi Kawamura
- Department of General Thoracic Surgery, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo, 173-8606, Japan
| |
Collapse
|
24
|
Xiao F, Zheng R, Yang D, Cao K, Zhang S, Wu B, Shao Y, Zhou B. Sex-dependent aortic valve pathology in patients with rheumatic heart disease. PLoS One 2017; 12:e0180230. [PMID: 28662157 PMCID: PMC5491156 DOI: 10.1371/journal.pone.0180230] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 06/12/2017] [Indexed: 12/15/2022] Open
Abstract
Background Rheumatic heart disease is an autoimmune disease caused by group A streptococci infection and frequently affects the aortic valve. Sex differences are common in the disease progression, treatment, and outcome. However, little is known about the sex differences in the pathology of aortic valves in rheumatic heart disease. Design We studied the end-stage calcific aortic valves from male versus female patients to reveal the sex-dependent pathology differences and molecular changes associated with requiring valve replacement. Methods Aortic valves from 39 patients with rheumatic heart disease (19 males and 20 females) were collected at the time of aortic valve replacement for comparative pathology, immunohistochemistry, and gene expression analyses. Clinical characteristics were also analyzed and compared between the two groups. Results Aortic valves from female patients exhibited increased expression of collagens, infiltration of monocytes/macrophages and neovascularization. Aortic valves from female patients also had increased expression of inflammatory genes involved in the NFKB pathway (phosphorylated NFKB p65 subunit, IL8, and NOS3) and Th1 cytokine genes (IFNA and IL12B). The severe valve pathology in female patients was correlated with a higher serum level of anti-streptolysin O antibodies. Conclusion Inflammation is more prominent in aortic valves of female patients with rheumatic heart disease. This sex difference may contribute to the severe valve pathology and worse outcome of female patients.
Collapse
Affiliation(s)
- Feng Xiao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Departments of Genetics, Pediatrics and Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Rui Zheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Di Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kejiang Cao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shijiang Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bingruo Wu
- Departments of Genetics, Pediatrics and Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Yongfeng Shao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bin Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Departments of Genetics, Pediatrics and Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
25
|
Human interstitial cellular model in therapeutics of heart valve calcification. Amino Acids 2017; 49:1981-1997. [DOI: 10.1007/s00726-017-2432-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/27/2017] [Indexed: 12/27/2022]
|
26
|
Endothelium adhesion molecules ICAM-1, ICAM-2, VCAM-1 and VLA-4 expression in leprosy. Microb Pathog 2017; 104:116-124. [PMID: 28088473 DOI: 10.1016/j.micpath.2017.01.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 01/10/2017] [Accepted: 01/11/2017] [Indexed: 11/21/2022]
Abstract
Leprosy triggers a complex relationship between the pathogen and host immune response. Endothelium plays an important role in this immune response by directly influencing cell migration to infected tissues. The objective of this work is to investigate the possible role of endothelium in M. leprae infection, correlating the characteristics of endothelial markers with the expression pattern of cytokines. Thirty-six skin biopsy samples were cut into 5-μm thick sections and stained with hematoxylin-eosin and Ziehl-Neelsen for morphological analysis and then submitted to immunohistochemical analysis using monoclonal antibodies against ICAM-1, ICAM-2, VCAM-1, and VLA-4. Immunostaining for ICAM-1 showed a significantly larger number of stained endothelial cells in the tuberculoid leprosy (9.92 ± 1.11 cells/mm2) when compared to lepromatous samples (5.87 ± 1.01 cells/mm2) and ICAM-2 revealed no significant difference in the number of endothelial cells expressing this marker between the tuberculoid (13.21 ± 1.27 cells/mm2) and lepromatous leprosy (14.3 ± 1.02 cells/mm2). VCAM-1-immunostained showed 18.28 ± 1.46/mm2 cells in tuberculoid leprosy and 10.67 ± 1.25 cells/mm2 in the lepromatous leprosy. VLA-4 exhibited 22.46 ± 1.38 cells/mm2 in the tuberculoid leprosy 16.04 ± 1.56 cells/mm2 in the lepromatous leprosy. Samples with characteristics of the tuberculoid leprosy exhibited a larger number of cells stained with ICAM-1, VCAM-1 and VLA-4, demonstrating the importance of these molecules in the migration and selection of cells that reach the inflamed tissue.
Collapse
|
27
|
Gabriele LG, Morandini AC, Dionísio TJ, Santos CF. Angiotensin II Type 1 Receptor Knockdown Impairs Interleukin-1β-Induced Cytokines in Human Periodontal Fibroblasts. J Periodontol 2017; 88:e1-e11. [DOI: 10.1902/jop.2016.160354] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
28
|
Sun F, Shi J, Chen S, Deng C, Hu X, Li H, Li G, Liu Y, Dong N. Lazaroid U-74389G inhibits the osteoblastic differentiation of IL-1β-indcued aortic valve interstitial cells through glucocorticoid receptor and inhibition of NF-κB pathway. J Steroid Biochem Mol Biol 2015; 152:114-23. [PMID: 25957738 DOI: 10.1016/j.jsbmb.2015.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/07/2015] [Accepted: 05/01/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Aortic valve calcification is characterized as the active process of aortic valve interstitial cells (AVICs), and considered as an inflammatory disease. As an antioxidant, the anti-inflammatory activity of Lazaroid has been exhibited in various models. We hypothesized that Lazaroid U-74389G would inhibit the osteoblastic differentiation of AVICs induced by IL-1β. METHODS Normal tricuspid aortic valve leaflets were collected from patients with acute aortic dissection (Type A) undergoing the Bentall procedure. AVICs were isolated and stimulated with IL-1β in presence or absence of U-74389G in culture. Cell lysates were analyzed for osteogenic markers and nuclear factor-κB using real-time PCR and Immunoblotting. Culture media was analyzed for IL-6 and IL-8 with enzyme-linked immunosorbent assay. Alizarin Red Staining was adopted to demonstrate the calcium deposition. RESULTS The expression of alkaline phosphatase and bone morphogenetic protein, accompanied by the production of IL-6 and IL-8, was up-regulated in response to IL-1β and was inhibited by the addition of U-74389G. The NF-κB pathway was activated by IL-1β and involved in the suppression of U-74389G on osteoblastic differentiation in AVICs. The negative effects of U-74389G on ostengenic gene expression and mineralization of AVICs were blocked by glucocorticoid receptor antagonist mifepristone and the NF-κB inhibitor Bay 11-7082. CONCLUSIONS U-74389G inhibits the pro-osteogenic response to IL-1β stimulation in AVICs. The osteoblastic differentiation and mineralization of AVICs were inhabited by U-74389G though the modulation of NF-κB activation, and this pathway could be potential therapeutic targets for medical treatment of calcified aortic valve disease.
Collapse
Affiliation(s)
- Fuqiang Sun
- Department of Cardiovascular surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Cardiovascular surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Jiawei Shi
- Department of Cardiovascular surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Si Chen
- Department of Cardiovascular surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Cheng Deng
- Department of Cardiovascular surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xingjian Hu
- Department of Cardiovascular surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Huadong Li
- Department of Cardiovascular surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Geng Li
- Department of Cardiovascular surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Yi Liu
- Department of Cardiovascular surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Nianguo Dong
- Department of Cardiovascular surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
29
|
Abstract
During every heartbeat, cardiac valves open and close coordinately to control the unidirectional flow of blood. In this dynamically challenging environment, resident valve cells actively maintain homeostasis, but the signalling between cells and their microenvironment is complex. When homeostasis is disrupted and the valve opening obstructed, haemodynamic profiles can be altered and lead to impaired cardiac function. Currently, late stages of cardiac valve diseases are treated surgically, because no drug therapies exist to reverse or halt disease progression. Consequently, investigators have sought to understand the molecular and cellular mechanisms of valvular diseases using in vitro cell culture systems and biomaterial scaffolds that can mimic the extracellular microenvironment. In this Review, we describe how signals in the extracellular matrix regulate valve cell function. We propose that the cellular context is a critical factor when studying the molecular basis of valvular diseases in vitro, and one should consider how the surrounding matrix might influence cell signalling and functional outcomes in the valve. Investigators need to build a systems-level understanding of the complex signalling network involved in valve regulation, to facilitate drug target identification and promote in situ or ex vivo heart valve regeneration.
Collapse
|
30
|
Venardos N, Nadlonek NA, Zhan Q, Weyant MJ, Reece TB, Meng X, Fullerton DA. Aortic valve calcification is mediated by a differential response of aortic valve interstitial cells to inflammation. J Surg Res 2014; 190:1-8. [PMID: 24746950 DOI: 10.1016/j.jss.2014.03.051] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/04/2014] [Accepted: 03/12/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND Although calcific aortic stenosis is common, calcification of the other three heart valves is not. The aortic valve interstitial cell (VIC) has been implicated in the pathogenesis of aortic stenosis. Proinflammatory stimulation of aortic VICs induces an osteogenic and inflammatory phenotypic change. We hypothesized that the VICs of the other heart valves do not undergo these changes. Using isolated human VICs from normal aortic, mitral, pulmonary, and tricuspid valves, our purpose was to compare the osteogenic response to proinflammatory stimulation via toll-like receptor 4 (TLR-4). MATERIALS AND METHODS Aortic, pulmonic, mitral, and tricuspid (n=4 for each valve type) VICs were isolated from hearts valves explanted from patients undergoing cardiac transplantation. Cells were cultured and grown to confluence in passage 2-6 before treatment with Lipopolysaccharide (LPS) (100-200 ng/mL) for 24 or 48 h. Cells were characterized by immunofluorescent staining. TLR-4 expression was analyzed (immunoblotting, flow cytometry). Bone morphogenetic protein 2 and intercellular adhesion molecule 1 production were determined (immunoblotting). Monocyte chemoattractant protein 1 levels were determined by enzyme-linked immunosorbent assay. Statistics were by Mann-Whitney U test. RESULTS TLR-4 stimulation induced bone morphogenetic protein 2 production only in aortic VICs (P<0.05). Intra-cellular adhesion molecule 1 production and monocyte chemoattractant protein 1 secretion increased in a similar fashion among TLR-4-stimulated VICs from all four valves. CONCLUSIONS Proinflammatory stimulation induces an osteogenic phenotype in aortic VICs but not mitral, pulmonic, or tricuspid VICs. We conclude that this differential osteogenic response of aortic VICs contributes to the pathogenesis of calcific aortic stenosis.
Collapse
Affiliation(s)
- Neil Venardos
- The Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine.
| | - Nicole A Nadlonek
- The Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine
| | - Qiong Zhan
- The Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine
| | - Michael J Weyant
- The Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine
| | - Thomas Brett Reece
- The Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine
| | - Xianzhong Meng
- The Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine
| | - David A Fullerton
- The Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine
| |
Collapse
|